1
|
Huang Y, Wang Y, Huang X, Yu X. Unveiling the overlooked fungi: the vital of gut fungi in inflammatory bowel disease and colorectal cancer. Gut Pathog 2024; 16:59. [PMID: 39407244 PMCID: PMC11481806 DOI: 10.1186/s13099-024-00651-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
The fungi of the human microbiota play important roles in the nutritional metabolism and immunological balance of the host. Recently, research has increasingly emphasised the role of fungi in modulating inflammation in intestinal diseases and maintaining health in this environment. It is therefore necessary to understand more clearly the interactions and mechanisms of the microbiota/pathogen/host relationship and the resulting inflammatory processes, as well as to offer new insights into the prevention, diagnosis and treatment of inflammatory bowel disease (IBD), colorectal cancer (CRC) and other intestinal pathologies. In this review, we comprehensively elucidate the fungal-associated pathogenic mechanisms of intestinal inflammation in IBD and related CRC, with an emphasis on three main aspects: the direct effects of fungi and their metabolites on the host, the indirect effects mediated by interactions with other intestinal microorganisms and the immune regulation of the host. Understanding these mechanisms will enable the development of innovative approaches based on the use of fungi from the resident human microbiota such as dietary interventions, fungal probiotics and faecal microbiota transplantation in the prevention, diagnosis and treatment of intestinal diseases.
Collapse
Affiliation(s)
- Yilin Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yang Wang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaotian Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Xiaomin Yu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
2
|
Dai X, Li Y, Wang H, Dai Z, Chen Y, Liu Y, Huang S. Development and validation of nomograms based on pre-/post-operative CEA and CA19-9 for survival predicting in stage I-III colorectal cancer patients after radical resection. Front Oncol 2024; 14:1402847. [PMID: 39464705 PMCID: PMC11502300 DOI: 10.3389/fonc.2024.1402847] [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: 03/18/2024] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
Background Carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) are the predominant serum tumour markers (STMs) for predicting the prognosis of colorectal cancer (CRC). The objective of this research is to develop clinical prediction models based on preoperative and postoperative CEA and CA19-9 levels. Methods 1,452 consecutive participants with stage I-III colorectal cancer were included. Kaplan-Meier method, log-rank test, and multivariate COX regression were used to evaluate the significance of preoperative and postoperative STMs. Patients were grouped into a discovery cohort (70%) and a validation cohort (30%). Variables for the nomograms were selected according to the Akaike information criterion (AIC). Subsequently, two clinical predictive models were constructed, evaluated, validated, and then compared with the AJCC 8th TNM stage. Results The overall survival (OS) rate and disease-free survival(DFS) rate declined progressively as the number of positive tumour markers(NPTMs) before and after surgery increased. For both OS and DFS, age, sex, pN stage, and NPTMs before and after surgery were independent prognostic factors, and then clinical prediction models were developed. The Concordance index (C-index), Receiver operating characteristic (ROC) curve, calibration curve, Decision curve analysis (DCA), and risk score stratification all indicated that the models possessed robust predictive efficacy and clinical applicability. The Net reclassification index (NRI) and Integrated discrimination improvement (IDI) indicated that the performance of models was significantly superior to the TNM stage. Conclusion Nomograms based on pre-and postoperative CEA and CA19-9 can accurately predict survival and recurrence for stage I-III CRC patients after radical surgery, and were significantly better than the AJCC 8th TNM stage.
Collapse
Affiliation(s)
- Xuan Dai
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifan Li
- Department of Gastrointestinal Surgery, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Haoran Wang
- The First Clinical School, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhujiang Dai
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Chen
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Liu
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiyong Huang
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Kourie HR, Zouein J, Zalaquett Z, Chebly A, Nasr L, Karak FE, Sadek M, Safar O, Fouani M, Bitar N, Kachmar K, Nasr F, Farhat F, Makarem J, Kattan J, Taieb J. Liquid biopsy as a tool for KRAS/NRAS/BRAF baseline testing in metastatic colorectal cancer. Clin Res Hepatol Gastroenterol 2024; 48:102417. [PMID: 39009221 DOI: 10.1016/j.clinre.2024.102417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND The absence of KRAS and NRAS gene mutations (RAS wild type) in metastatic colorectal cancer (mCRC), is associated with a good response to targeted therapy with anti-EGFR receptor antibodies. The current gold standard for RAS mutational status identification is genetic testing on tissue biopsy samples. OBJECTIVE This study aimed to assess the relevance of liquid biopsy as a less invasive alternative to tissue biopsy for detecting KRAS/NRAS and BRAF mutations in patients with metastatic colorectal cancer (mCRC). The study also aimed to determine the concordance between liquid biopsy and tissue biopsy. METHODS This is a phase IV, observational, uncontrolled, non-comparative, non-randomized, open label study. RAS/BRAF status will be tested at baseline using tissue and liquid biopsy using the Idylla/Biocartis PCR-based device. The primary endpoint is the comparison of the RAS status based on liquid biopsy with the RAS status based on tissue biopsy. RESULTS 100 patients with mCRC were included in the study. 75 % of patients showed concordant results between liquid biopsy and tissue biopsy, while 25 % had discordant results. Liquid biopsy demonstrated a sensitivity of 62 % and a specificity of 93 %. The accuracy of liquid biopsy was 75 %, with a moderate agreement between the two tests. The most frequent mutations in concordant cases were in KRAS (41 %), followed by NRAS (4 %) and BRAF (3 %). Mutations were not detected in 42 % of tissue biopsy samples and 60 % of liquid biopsy samples. The presence of hepatic metastases did not significantly affect the concordance between the biopsy methods. CONCLUSION Liquid biopsy using the Idylla™ system showed a relatively low sensitivity but high specificity for detecting KRAS/NRAS and BRAF mutations in mCRC patients. Despite some discordant cases, liquid biopsy remains a promising alternative to tissue biopsy due to its non-invasiveness, ability to provide multiple samples, and better representation of tumor heterogeneity.
Collapse
Affiliation(s)
- Hampig Raphael Kourie
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University, Boulevard Alfred Naccache, Beirut, Lebanon.
| | - Joseph Zouein
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University, Boulevard Alfred Naccache, Beirut, Lebanon; Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Ziad Zalaquett
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University, Boulevard Alfred Naccache, Beirut, Lebanon
| | - Alain Chebly
- Jacques Loiselet Center for Medical Genetics and Genomics (CGGM), Faculty of Medicine, Saint Joseph University, Beirut, Lebanon; Higher Institute of Public Health, Saint Joseph University, Beirut, Lebanon
| | - Lewis Nasr
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University, Boulevard Alfred Naccache, Beirut, Lebanon
| | - Fadi El Karak
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University, Boulevard Alfred Naccache, Beirut, Lebanon
| | - Maroun Sadek
- Hematology-Oncology Department, Lebanese Hospital Geitaoui, Beirut, Lebanon
| | | | | | | | | | - Fady Nasr
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University, Boulevard Alfred Naccache, Beirut, Lebanon
| | | | | | - Joseph Kattan
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University, Boulevard Alfred Naccache, Beirut, Lebanon
| | - Julien Taieb
- Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
4
|
Zhou T, Wu J, Khan A, Hu T, Wang Y, Salama ES, Su S, Han H, Jin W, Li X. A probiotic Limosilactobacillus fermentum GR-3 mitigates colitis-associated tumorigenesis in mice via modulating gut microbiome. NPJ Sci Food 2024; 8:61. [PMID: 39242568 PMCID: PMC11379937 DOI: 10.1038/s41538-024-00307-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
Bacterial therapy for colorectal cancer (CRC) represents a burgeoning frontier. The probiotic Limosilactobacillus fermentum GR-3, derived from traditional food "Jiangshui", exhibited superior antioxidant capacity by producing indole derivatives ICA and IPA. In an AOM/DSS-induced CRC mouse model, GR-3 treatment alleviated weight loss, colon shortening, rectal bleeding and intestinal barrier disruption by reducing oxidative stress and inflammation. GR-3 colonization in distant colon induced apoptosis and reduced tumor incidence by 51.2%, outperforming the control strain and vitamin C. The beneficial effect of GR-3 on CRC was associated with gut microbiome modulation, increasing SCFA producer Lachnospiraceae NK4A136 group and suppressing pro-inflammatory strain Bacteroides. Metagenomic and metabolic analyses revealed that GR-3 intervention upregulated antioxidant genes (xseA, ALDH) and butyrate synthesis gene (bcd), while increasing beneficial metabolites (SCFAs, ICA, IPA, VB12 and VD3) and reducing harmful secondary bile acids. Overall, GR-3 emerges as a promising candidate in CRC therapy, offering effective gut microbiome remediation.
Collapse
Affiliation(s)
- Tuoyu Zhou
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
- State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agricultural Science and Technology, Lanzhou University, Lanzhou, China
| | - Jingyuan Wu
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, 730000, China
| | - Aman Khan
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Tianxiang Hu
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Yiqing Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, 730000, China
| | - El-Sayed Salama
- Department of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, Gansu Province, PR China
| | - Shaochen Su
- Healthy Examination & Management Center, First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Huawen Han
- State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agricultural Science and Technology, Lanzhou University, Lanzhou, China.
| | - Weilin Jin
- Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, China.
| | - Xiangkai Li
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
5
|
Wang Y, Zhang J, Yang Y, Liu Z, Sun S, Li R, Zhu H, Li T, Zheng J, Li J, Ma L. Circular RNAs in human diseases. MedComm (Beijing) 2024; 5:e699. [PMID: 39239069 PMCID: PMC11374765 DOI: 10.1002/mco2.699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/07/2024] Open
Abstract
Circular RNAs (circRNAs) are a unique class of RNA molecules formed through back-splicing rather than linear splicing. As an emerging field in molecular biology, circRNAs have garnered significant attention due to their distinct structure and potential functional implications. A comprehensive understanding of circRNAs' functions and potential clinical applications remains elusive despite accumulating evidence of their involvement in disease pathogenesis. Recent research highlights their significant roles in various human diseases, but comprehensive reviews on their functions and applications remain scarce. This review provides an in-depth examination of circRNAs, focusing first on their involvement in non-neoplastic diseases such as respiratory, endocrine, metabolic, musculoskeletal, cardiovascular, and renal disorders. We then explore their roles in tumors, with particular emphasis on exosomal circular RNAs, which are crucial for cancer initiation, progression, and resistance to treatment. By detailing their biogenesis, functions, and impact on disease mechanisms, this review underscores the potential of circRNAs as diagnostic biomarkers and therapeutic targets. The review not only enhances our understanding of circRNAs' roles in specific diseases and tumor types but also highlights their potential as novel diagnostic and therapeutic tools, thereby paving the way for future clinical investigations and potential therapeutic interventions.
Collapse
Affiliation(s)
- Yuanyong Wang
- Department of Thoracic Surgery Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) The First Department of Thoracic Surgery Peking University Cancer Hospital and Institute Peking University School of Oncology Beijing China
| | - Jin Zhang
- Department of Traditional Chinese Medicine Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province Xi'an China
| | - Yuchen Yang
- Department of Traditional Chinese Medicine Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province Xi'an China
| | - Zhuofeng Liu
- Department of Traditional Chinese Medicine The Third Affiliated Hospital of Xi'an Medical University Xi'an China
| | - Sijia Sun
- Department of Traditional Chinese Medicine Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province Xi'an China
| | - Rui Li
- Department of Epidemiology School of Public Health Air Force Medical University Xi'an China
| | - Hui Zhu
- Department of Anatomy Medical College of Yan'an University Yan'an China
- Institute of Medical Research Northwestern Polytechnical University Xi'an China
| | - Tian Li
- School of Basic Medicine Fourth Military Medical University Xi'an China
| | - Jin Zheng
- Department of Traditional Chinese Medicine Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province Xi'an China
| | - Jie Li
- Department of Endocrine Xijing 986 Hospital Air Force Medical University Xi'an China
| | - Litian Ma
- Department of Thoracic Surgery Tangdu Hospital Air Force Medical University Xi'an China
- Department of Traditional Chinese Medicine Tangdu Hospital Air Force Medical University Xi'an China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province Xi'an China
- Department of Gastroenterology Tangdu Hospital Air Force Medical University Xi'an China
- School of Medicine Northwest University Xi'an China
| |
Collapse
|
6
|
Jiang J, Li X, Wang J, Chen S, Chen L. SLC25A19 drives colorectal cancer progression by regulating p53. Cancer Med 2024; 13:e70253. [PMID: 39344563 PMCID: PMC11440145 DOI: 10.1002/cam4.70253] [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: 01/24/2024] [Revised: 09/02/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Investigating the molecular mechanism of colorectal cancer (CRC), a common lethal malignancies worldwide, is of great clinical significance. Solute carrier family 25 member 19 (SLC25A19) is a member of the solute carrier family that contribute to cellular functions, including tumor biology. Recently, many studies have attention on uncovering the relationship of SLC25A19 with malignant cancers, but its precise involvement in the regulation of CRC has not been thoroughly understood. This study sought to uncover the role and mechanism of SLC25A19 in CRC development. METHODS The GEPIA database and immunohistochemical staining were utilized to detect the expression of SLC25A19 in CRC tissues. The functional influences of SLC25A19 on CRC cell phenotypes were evaluated through a series of assays including celigo cell count, colony formation, CCK-8, flow cytometry, wound healing, and transwell assays following knocking down SLC25A19. Subsequently, the xenograft tumor model was constructed to evaluate the effect of SLC25A19 on tumor growth in vivo. The underlying mechanisms of SLC25A19 silencing were investigated using the human phospho-kinase array. RESULTS This study demonstrated the upregulation of SLC25A19 in CRC and its significant correlation with unfavorable prognosis in CRC patients. Suppression of SLC25A19 resulted in significant inhibition of cell proliferation, colony formation, and cell migration, alongside a boost in cell apoptosis. In vivo experiments revealed that silenced SLC25A19 displayed reduced growth rates and formed smaller xenografts. Mechanistically, the p53 pathway was found to be upregulated by SLC25A19 knockdown and mediated the function of SLC25A19. CONCLUSIONS Consequently, SLC25A19 was identified as a novel molecule with key regulatory ability in CRC development.
Collapse
Affiliation(s)
- Jinbo Jiang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Xuemei Li
- Advanced Medical Research Institute, School of Medicine, Shandong University, Jinan, Shandong, China
| | - Jiayong Wang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Shaofei Chen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingjuan Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Wu Z, Dai J, Li J, Zhang Z, Shen X. Exploiting the role of O6-methylguanine-DNA-methyltransferase (MGMT) in gastrointestinal cancers. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03365-4. [PMID: 39167167 DOI: 10.1007/s00210-024-03365-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Gastrointestinal (GI) cancer is a prevalent disease and is recognized as the primary cause of cancer-related mortality globally. Therefore, there is an urgent need for novel diagnostic and treatment approaches for GC. The methylation of the O(6)-methylguanine DNA methyltransferase (MGMT) gene promoter is a significant factor in the development of colorectal cancer (CRC), namely in roughly 30-40% of cases where the cancer has spread. MGMT plays a role in the repair of DNA damage caused by methylating drugs like temozolomide (TMZ) and chloroethylating compounds like carmustine. As a result, it contributes to the resistance of chemotherapy when these agents are utilized. Although MGMT's role in the development of CRC is well established, its prognostic significance remains a subject of debate. Only a limited number of research have been conducted to examine the prognostic significance of MGMT methylation, yielding varying outcomes. This review explores the structural functions and repair processes of MGMT, focusing on the putative structural and functional significance of the N-terminal domain of MGMT. It also investigates the advancement of cancer treatment techniques that specifically target MGMT.
Collapse
Affiliation(s)
- Ziming Wu
- School of Public Health, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Jie Dai
- Anqing 116 Hospital, Anqing, 246001, Anhui, China
| | - Jie Li
- Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Zhengyu Zhang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zheijiang, China
| | - Xbing Shen
- School of Public Health, Southeast University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
8
|
Zeng S, Jin N, Yu B, Ren Q, Yan Z, Fu S. Chimeric antigen receptor-T cells targeting epithelial cell adhesion molecule antigens are effective in the treatment of colorectal cancer. BMC Gastroenterol 2024; 24:249. [PMID: 39107717 PMCID: PMC11302356 DOI: 10.1186/s12876-024-03286-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/07/2024] [Indexed: 08/10/2024] Open
Abstract
OBJECTIVE To construct chimeric antigen receptor (CAR)-T cells targeting epithelial cell adhesion molecule (EpCAM) antigen (anti-EpCAM-CAR-T). METHODS A third-generation CAR-T cell construct used a single-chain variable fragment derived from monoclonal antibody against human EpCAM. Peripheral blood mononuclear cells were extracted from volunteers. The proportion of cluster of differentiation 8 positive (CD8+) and CD4 + T cells was measured using flow cytometry. Western blot was used to detect the expression of EpCAM-CAR. The killing efficiency was detected using the MTT assay and transwell assay, and the secretion of killer cytokines tumour necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) was detected using the ELISA. The inhibitory effect of EpCAM-CAR-T on colorectal cancer in vivo was detected using xenografts. RESULTS It was found that T cells expanded greatly, and the proportion of CD3+, CD8 + and CD4 + T cells was more than 60%. Furthermore, EpCAM-CAR-T cells had a higher tumour inhibition rate in the EpCAM expression positive group than in the negative group (P < 0.05). The secretion of killer cytokines TNF-α and IFN-γ in the EpCAM expression positive cell group was higher than that in the negative group (P < 0.05). In the experimental group treated with EpCAM-CAR-T cells, the survival rate of nude mice was higher (P < 0.05), and the tumour was smaller than that in the blank and control groups (P < 0.05). The secretion of serum killer cytokines TNF-α and IFN-γ in tumour-bearing nude mice in the experimental group treated with EpCAM-CAR-T cells was higher than that in the blank and control groups (P < 0.05). CONCLUSION This study successfully constructed EpCAM-CAR cells and found that they can target and recognise EpCAM-positive tumour cells, secrete killer cytokines TNF-α and IFN-γ and better inhibit the growth and metastasis of colorectal cancer in vitro and in vivo than unmodified T cells.
Collapse
Affiliation(s)
- Siheng Zeng
- Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China
- Birth Defects and Regenerative Medicine Laboratory, Department of Biochemistry & Molecular Biology, Biomedicine and Health Graduate Education Innovation Center, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan, Shanxi, 030001, China
| | - Ning Jin
- Birth Defects and Regenerative Medicine Laboratory, Department of Biochemistry & Molecular Biology, Biomedicine and Health Graduate Education Innovation Center, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan, Shanxi, 030001, China
| | - Baofeng Yu
- Birth Defects and Regenerative Medicine Laboratory, Department of Biochemistry & Molecular Biology, Biomedicine and Health Graduate Education Innovation Center, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan, Shanxi, 030001, China
| | - Qing Ren
- Department of Gynecology, Hainan West Central Hospital, Danzhou, Hainan, 571700, China
| | - Zhiqiang Yan
- Department of Gynecology, Hainan West Central Hospital, Danzhou, Hainan, 571700, China
| | - Songtao Fu
- Birth Defects and Regenerative Medicine Laboratory, Department of Biochemistry & Molecular Biology, Biomedicine and Health Graduate Education Innovation Center, Shanxi Medical University, No. 56, Xinjian South Road, Taiyuan, Shanxi, 030001, China.
- Biomedicine and Health Graduate Education Innovation Center, Taiyuan, Shanxi, 030001, China.
| |
Collapse
|
9
|
Lou S, Liu YX, Xia C, Zhang Q, Deng L, Tang JJ. Novel meroterpene-like compounds inhibit ferroptosis through Fe 2+ chelation. Int J Biochem Cell Biol 2024; 173:106610. [PMID: 38879152 DOI: 10.1016/j.biocel.2024.106610] [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: 03/29/2024] [Revised: 05/30/2024] [Accepted: 06/09/2024] [Indexed: 07/22/2024]
Abstract
Colorectal cancer (CRC) is the third most common type of cancer in the world. It is characterized by complex crosstalk between various signaling pathways, as a result of which it is highly challenging to identify optimal therapeutic targets and design treatment strategies. In this study, we tested the effect of 700 compounds on the CRC cell line HT-29 by using the sulforhodamine B assay and screened out 17 compounds that exhibited high toxicity (indicated by an inhibition rate of ≥75 % when applied at a concentration of 10 µM) against the HT-29 cell line. Next, we investigated the mechanisms underlying the effects of these 17 highly toxic compounds. The results of ferroptosis analysis and electron microscopy showed that compounds 575 and 578 were able to significantly reverse RSL3-induced increase in ferroptosis, while compound 580 had a less pronounced ferroptosis-regulating effect. In subsequent experiments, western blotting showed that compounds 575, 578, and 580, which belong to a class of meroterpene-like compounds that affect ferroptosis, do not induce autophagy or apoptosis in the CRC cell line. Instead, Fe2+ chelation experiments showed that these three compounds can serve as iron chelators by chelating Fe2+ at a 1:1 (chelator: Fe2+) ratio. Specifically, the aldehyde and hydroxyl groups of the benzene ring in these compounds may chelate Fe2+, thus reducing Fe2+ levels in cells and inhibiting ferroptosis. These results indicate that these novel meroterpene-like compounds are potential therapeutic small-molecule candidates for targeting ferroptosis in tumors.
Collapse
Affiliation(s)
- Shiyang Lou
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Yan-Xiang Liu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Chao Xia
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Qiang Zhang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Lu Deng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi 712100, China; Shenzhen Research Institute, Northwest A&F University, Shenzhen, Guangdong 518000, China.
| | - Jiang-Jiang Tang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shanxi 712100, China.
| |
Collapse
|
10
|
Wu XQ, Deng LH, Xue Q, Li X, Li MH, Wang JT. Metformin administration in prevention of colorectal polyps in type 2 diabetes mellitus patients. World J Clin Cases 2024; 12:4206-4216. [PMID: 39015918 PMCID: PMC11235560 DOI: 10.12998/wjcc.v12.i20.4206] [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: 03/10/2024] [Revised: 05/07/2024] [Accepted: 05/29/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Colorectal polyps are frequently observed in patients with type 2 diabetes mellitus (DM), posing a significant risk for colorectal cancer. Metformin, a widely prescribed biguanidine drug for type 2 DM, has been suggested to have potential chemoprophylactic effects against various cancers. AIM To explore the correlation between colorectal polyps and metformin use in type 2 DM patients. METHODS Type 2 DM patients were categorized into polyp and non-polyp groups. Following this, all patients were categorized into the type 2 DM-metformin, type 2 DM-non-metformin, and non-type 2 DM groups. Based on the baseline colonoscopy results, we performed pairwise comparisons of the incidence of colorectal polyps among the three groups. Additionally, we analyzed the relationship between colorectal polyps and the duration of metformin use and between the size and number of polyps and metformin use. Simultaneously, we focused on the specific pathological types of polyps and analyzed their relationship with metformin use. Finally, we compared the incidence of polyps between metformin and non-metformin groups according to the interval colonoscopy results. RESULTS The rate of metformin use in patients with colorectal polyps was 0.502 times that of patients without colorectal polyps [odds ratio (OR) = 0.502, 95% confidence interval (CI): 0.365-0.689; P < 0.001]. The incidence of colorectal polyps did not differ significantly between the type 2 DM-metformin and non-type 2 DM groups (P > 0.05). Furthermore, the correlations between the duration of metformin use and the incidence of colorectal polyps and between the size and number of polyps and metformin use were not statistically significant (P > 0.05). Metformin use did not affect the incidence of colorectal polyps during interval colonoscopy (P > 0.05). CONCLUSION Metformin use and colorectal polyp incidence in type 2 DM patients showed a negative correlation, independent of the hypoglycemic effect of metformin.
Collapse
Affiliation(s)
- Xiao-Qing Wu
- Department of Geriatrics, Peking University People's Hospital, Beijing 100044, China
| | - Li-Hua Deng
- Department of Geriatrics, Peking University People's Hospital, Beijing 100044, China
| | - Qian Xue
- Department of Geriatrics, Peking University People's Hospital, Beijing 100044, China
| | - Xia Li
- Department of Geriatrics, Peking University People's Hospital, Beijing 100044, China
| | - Meng-Han Li
- Department of Geriatrics, Peking University People's Hospital, Beijing 100044, China
| | - Jing-Tong Wang
- Department of Geriatrics, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
11
|
Yu L, Wang H, Wang F, Guo J, Xiao B, Hou Z, Lu Z, Pan Z, Zhou Y, Ye S, Wan D, Lin B, Ou Q, Fang Y. Serum biomarkers REG1A and REG3A combined with the traditional CEA represent a novel nomogram for the screening and risk stratification of colorectal cancer. Clin Transl Oncol 2024:10.1007/s12094-024-03566-6. [PMID: 38965192 DOI: 10.1007/s12094-024-03566-6] [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: 05/04/2024] [Accepted: 06/09/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND To develop and validate a serum protein nomogram for colorectal cancer (CRC) screening. METHODS The serum protein characteristics were extracted from an independent sample containing 30 colorectal cancer and 12 polyp tissues along with their paired samples, and different serum protein expression profiles were validated using RNA microarrays. The prediction model was developed in a training cohort that included 1345 patients clinicopathologically confirmed CRC and 518 normal participants, and data were gathered from November 2011 to January 2017. The lasso logistic regression model was employed for features selection and serum nomogram building. An internal validation cohort containing 576 CRC patients and 222 normal participants was assessed. RESULTS Serum signatures containing 27 secreted proteins were significantly differentially expressed in polyps and CRC compared to paired normal tissue, and REG family proteins were selected as potential predictors. The C-index of the nomogram1 (based on Lasso logistic regression model) which contains REG1A, REG3A, CEA and age was 0.913 (95% CI, 0.899 to 0.928) and was well calibrated. Addition of CA199 to the nomogram failed to show incremental prognostic value, as shown in nomogram2 (based on logistic regression model). Application of the nomogram1 in the independent validation cohort had similar discrimination (C-index, 0.912 [95% CI, 0.890 to 0.934]) and good calibration. The decision curve (DCA) and clinical impact curve (ICI) analysis demonstrated that nomogram1 was clinically useful. CONCLUSIONS This study presents a serum nomogram that included REG1A, REG3A, CEA and age, which can be convenient for screening of colorectal cancer.
Collapse
Affiliation(s)
- Long Yu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Hao Wang
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Fulong Wang
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Jian Guo
- Senboll Biotechnology Co., Ltd., Pingshan Bio-Pharmacy Business Accelerator, Pingshan District, Shenzhen, 518000, Guangdong, China
| | - Binyi Xiao
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhenlin Hou
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhenhai Lu
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhizhong Pan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yaxian Zhou
- Senboll Biotechnology Co., Ltd., Pingshan Bio-Pharmacy Business Accelerator, Pingshan District, Shenzhen, 518000, Guangdong, China
| | - Sibin Ye
- Senboll Biotechnology Co., Ltd., Pingshan Bio-Pharmacy Business Accelerator, Pingshan District, Shenzhen, 518000, Guangdong, China
| | - Desen Wan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Bo Lin
- Department of Thyroid Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Qingjian Ou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| | - Yujing Fang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
12
|
Guo X, Wang R, Chen R, Zhang Z, Wang J, Liu X. Gut microbiota and serum metabolite signatures along the colorectal adenoma-carcinoma sequence: Implications for early detection and intervention. Clin Chim Acta 2024; 560:119732. [PMID: 38772522 DOI: 10.1016/j.cca.2024.119732] [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: 01/25/2024] [Revised: 04/11/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
AIM Our study focuses on the microbial and metabolomic profile changes during the adenoma stage, as adenomas can be considered potential precursors to colorectal cancer through the adenoma-carcinoma sequence. Identifying possible intervention targets at this stage may aid in preventing the progression of colorectal adenoma (CRA) to malignant lesions. Furthermore, we evaluate the efficacy of combined microbial and metabolite biomarkers in detecting CRA. METHODS Fecal metagenomic and serum metabolomic analyses were performed for the discovery of alterations of gut microbiome and metabolites in CRA patients (n = 26), Colorectal cancer (CRC) patients (n = 19), Familial Adenomatous Polyposis (FAP) patients (n = 10), and healthy controls (n = 20). Finally, analyzing the associations between gut microbes and metabolites was performed by a Receiver Operating Characteristic (ROC) curve. RESULTS Our analysis present that CRA patients differ significantly in gut microflora and serum metabolites compared with healthy controls, especially for Lachnospiraceae and Parasutterella. Its main metabolite, butyric acid, concentrations were raised in CRA patients compared with the healthy controls, indicating its role as a promoter of colorectal tumorigenesis. α-Linolenic acid and lysophosphatidylcholine represented the other healthy metabolite for CRA. Combining five microbial and five metabolite biomarkers, we differentiated CRA from CRC with an Area Under the Curve (AUC) of 0.85 out of this performance vastly superior to the specificity recorded by traditional markers CEA and CA199 in such differentiation of these conditions. CONCLUSIONS The study underlines significant microbial and metabolic alterations in CRA with a novel insight into screening and early intervention of its tumorigenesis.
Collapse
Affiliation(s)
- Xiaodong Guo
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200437, China.
| | - Ruoyao Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200437, China
| | - Rui Chen
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200437, China
| | - Zhongxiao Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No.1111, XianXia Road, Shanghai 200336, China.
| | - Jingxia Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200437, China
| | - Xuan Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
13
|
Du H, You L, Wu A, Wang F, Yu J, Chen C. Resolvin D1 Inhibits IL-6-Induced Epithelial-Mesenchymal Transition of Colorectal Cancer Cells by Targeting IL-6/STAT3 Signaling. Cell Biochem Biophys 2024; 82:1453-1461. [PMID: 38740668 DOI: 10.1007/s12013-024-01299-5] [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] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
Colorectal cancer (CRC) has emerged as a prevalent malignancy worldwide, exhibiting the high morbidity and mortality rates. Resolvin D1 (RvD1) can exert anti-inflammation and anti-cancer effects on various diseases. This study is aimed to explore the role of RvD1 in CRC cells. HCT15 and SW480 cells were stimulated with IL-6 in our study. A series of assays such as CCK-8, colony formation, wound healing, Transwell, Western blotting, and immunofluorescence staining were designed and conducted to figure out the role of RvD1 in CRC cells. RvD1 suppressed IL-6-induced SW480 and HCT15 cell proliferation. In addition, RvD1 inhibited IL-6-induced SW480 and HCT15 cell migration, invasion, and EMT process. In mechanism, RvD1 inhibited the activation of IL-6/STAT3 signaling in SW480 and HCT15 cells. Angoline strengthened the inhibitive effect of RvD1 on cell malignancy. RvD1 inhibited cell growth, migration, invasion and EMT process by inactivating IL-6/STAT3 signaling in CRC.
Collapse
Affiliation(s)
- Heng Du
- Department of Gestrointestinal Surgery, Huanggang Central Hospital Affiliated to Yangtze University, Changsha, 438000, China
| | - Lijuan You
- Department of Anesthesiology, Huanggang Central Hospital Affiliated to Yangtze University, Changsha, 438000, China
| | - Anding Wu
- Department of Gestrointestinal Surgery, Huanggang Central Hospital Affiliated to Yangtze University, Changsha, 438000, China
| | - Fei Wang
- Department of Gestrointestinal Surgery, Huanggang Central Hospital Affiliated to Yangtze University, Changsha, 438000, China
| | - Jie Yu
- Department of Gestrointestinal Surgery, Huanggang Central Hospital Affiliated to Yangtze University, Changsha, 438000, China
| | - Chaowu Chen
- Department of Gestrointestinal Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 438000, China.
| |
Collapse
|
14
|
Tang Y, Feng X, Lu Q, Cui C, Yu M, Wen Z, Luan Y, Dong L, Hu Z, Zhang R, Lu C, Liu J, Shinkura R, Hase K, Wang JY. MZB1-mediated IgA secretion suppresses the development and progression of colorectal cancer triggered by gut inflammation. Mucosal Immunol 2024; 17:450-460. [PMID: 38101774 DOI: 10.1016/j.mucimm.2023.12.002] [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: 08/14/2023] [Revised: 11/20/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Colorectal cancer (CRC) ranks among the top causes of mortality globally. Gut inflammation is one crucial risk factor that augments CRC development since patients suffering from inflammatory bowel disease have an increased incidence of CRC. The role of immunoglobulin (Ig)A in maintaining gut homeostasis and preventing inflammation has been well established. Our earlier work demonstrated that the marginal zone and B1 cell-specific protein (MZB1) promotes gut IgA secretion and its absence results in pronounced dextran sulfate sodium salt (DSS)-induced colitis. In the present study, we explored the role of MZB1 in CRC development using the azoxymethane (AOM)/DSS-induced CRC model. We observed an increase in both the number and size of the tumor nodules in Mzb1-/- mice compared with Mzb1+/+ mice. The increase in CRC development and progression in Mzb1-/- mice was associated with reduced intestinal IgA levels, altered gut flora, and more severe gut and systemic inflammation. Oral administration of the monoclonal IgA, W27, alleviated both the gut inflammation and AOM/DSS-induced CRC. Notably, cohousing Mzb1+/+ and Mzb1-/- mice from the 10th day after birth led to similar CRC development. Our findings underscore the pivotal role of MZB1-mediated IgA secretion in suppressing the onset and progression of CRC triggered by gut inflammation. Moreover, our study highlights the profound impact of microbiota composition, modulated by gut IgA levels, on gut inflammation. Nonetheless, establishing a direct correlation between the severity of colitis and subsequent CRC development and the presence or absence of a particular microbiota is challenging.
Collapse
Affiliation(s)
- Yue Tang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaoqian Feng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Lu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chaoqun Cui
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meiping Yu
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Zichao Wen
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yingying Luan
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lulu Dong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ziying Hu
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Runyun Zhang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chunhui Lu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jie Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Reiko Shinkura
- Laboratory of Immunology and Infection Control, Institute of Quantitative Biosciences, the University of Tokyo, Tokyo, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China; Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai 200052, China.
| |
Collapse
|
15
|
Huang Y, Zhang Z, Tong H, Qin W, Li Q, Ma L, Ren Z, Chen W, Zhang Y, Zhong Y, Yao L, Zhou P. Chondroitin polymerizing factor promotes development and progression of colorectal cancer via facilitating transcription of VEGFB. J Cell Mol Med 2024; 28:e18268. [PMID: 38775031 PMCID: PMC11109815 DOI: 10.1111/jcmm.18268] [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: 05/17/2023] [Revised: 02/28/2024] [Accepted: 03/13/2024] [Indexed: 05/24/2024] Open
Abstract
Colorectal cancer (CRC) is a highly prevalent malignancy affecting the digestive system on a global scale. This study aimed to explore the previously unexplored role of CHPF in the progression of CRC. Our results revealed a significant upregulation of CHPF expression in CRC tumour tissues compared to normal tissues, with its levels correlating with tumour malignancy. In vitro experiments using CRC cell lines demonstrated that inhibiting CHPF expression suppressed cell proliferation, colony formation and cell migration, while promoting apoptosis. Conversely, overexpressing CHPF had the opposite effect. Additionally, our xenograft models in mice confirmed the inhibitory impact of CHPF knockdown on CRC progression using various cell models. Mechanistic investigations unveiled that CHPF may enhance VEGFB expression through E2F1-mediated transcription. Functionally, suppressing VEGFB expression successfully mitigated the oncogenic effects induced by CHPF overexpression. Collectively, these findings suggest that CHPF may act as a tumour promoter in CRC, operating in a VEGFB-dependent manner and could be a potential target for therapeutic interventions in CRC treatment.
Collapse
Affiliation(s)
- Yuan Huang
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Zhen Zhang
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Hanxing Tong
- Department of General Surgery, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Wenzheng Qin
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Quanlin Li
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Lili Ma
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Zhong Ren
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Weifeng Chen
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yiqun Zhang
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yunshi Zhong
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Liqing Yao
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Pinghong Zhou
- Shanghai Collaborative Innovation Center of Endoscopy, Endoscopy Center and Endoscopy Research Institute, Zhongshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
16
|
Pei J, Gao Y, Wu A. An inflammation-related subtype classification for analyzing tumor microenvironment and clinical prognosis in colorectal cancer. Front Immunol 2024; 15:1369726. [PMID: 38742117 PMCID: PMC11089903 DOI: 10.3389/fimmu.2024.1369726] [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: 01/31/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Background The inflammatory response plays an essential role in the tumor microenvironment (TME) of colorectal cancer (CRC) by modulating tumor growth, progression, and response to therapy through the recruitment of immune cells, production of cytokines, and activation of signaling pathways. However, the molecular subtypes and risk score prognostic model based on inflammatory response remain to be further explored. Methods Inflammation-related genes were collected from the molecular signature database and molecular subtypes were identified using nonnegative matrix factorization based on the TCGA cohort. We compared the clinicopathological features, immune infiltration, somatic mutation profile, survival prognosis, and drug sensitivity between the subtypes. The risk score model was developed using LASSO and multivariate Cox regression in the TCGA cohort. The above results were independently validated in the GEO cohort. Moreover, we explored the biological functions of the hub gene, receptor interacting protein kinase 2 (RIPK2), leveraging proteomics data, in vivo, and in vitro experiments. Results We identified two inflammation-related subtypes (inflammation-low and inflammation-high) and have excellent internal consistency and stability. Inflammation-high subtype showed higher immune cell infiltration and increased sensitivity to common chemotherapeutic drugs, while inflammation-low subtype may be more suitable for immunotherapy. Besides, the two subtypes differ significantly in pathway enrichment and biological functions. In addition, the 11-gene signature prognostic model constructed from inflammation-related genes showed strong prognostic assessment power and could serve as a novel prognostic marker to predict the survival of CRC patients. Finally, RIPK2 plays a crucial role in promoting malignant proliferation of CRC cell validated by experiment. Conclusions This study provides new insights into the heterogeneity of CRC and provides novel opportunities for treatment development and clinical decision making.
Collapse
Affiliation(s)
| | | | - Aiwen Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
17
|
Steffes S, Kostev K, Schattenberg JM, Heinzow HS, Maschmeier M. Elevated Colon Cancer Rates Linked to Prior Appendicitis: A Retrospective Cohort Study Based on Data from German General Practices. J Clin Med 2024; 13:2342. [PMID: 38673618 PMCID: PMC11050991 DOI: 10.3390/jcm13082342] [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: 03/22/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Background/Objective: The association between appendicitis and colon cancer is not yet fully understood. Previous studies have shown contradictory results. Currently, no population-based data from Germany are available with regard to the incidence of colon cancer following appendicitis. This study investigated the association between appendicitis and the incidence of colon cancer in Germany. Methods: In this retrospective cohort study, the incidence of colon cancer was compared for patients with appendicitis and patients without appendicitis, matched for age, sex, index year, average annual consultation frequency, and comorbidity. The aim of the study was to explore the relationship between appendicitis and the incidence of colon cancer. The evaluation was carried out using logistic regression analyses. Results: The study included 49,790 people with and without appendicitis, with a median age of 41 years. During a follow-up period of up to 15 years, 1.04% of cases with appendicitis and 0.60% of cases without appendicitis were newly diagnosed with colon cancer, with some 36.4% of colon cancer cases diagnosed within the first six months after appendicitis. Regression analyses revealed a significant association between appendicitis and colon cancer, particularly in men and in the age groups 41-50 (HR: 10.30; 95% CI: 1.03-43.82) and 18-30 years (HR: 8.17; 95% CI: 1.03-64.58). Conclusions: The present retrospective cohort study suggests an association between appendicitis and the incidence of colon cancer in Germany. Based on our results, we recommend offering a colonoscopy or at least a stool test within 12 months after appendicitis, especially for 18-50-year-olds and >60-year-olds in good general health.
Collapse
Affiliation(s)
- Susann Steffes
- Department of Internal Medicine I, University Medical Centre of the Johannes Gutenberg-University, 55131 Mainz, Germany;
| | - Karel Kostev
- Epidemiology, IQVIA, 60549 Frankfurt am Main, Germany;
| | - Jörn M. Schattenberg
- Department of Internal Medicine I, University Medical Centre of the Johannes Gutenberg-University, 55131 Mainz, Germany;
- Department of Internal Medicine II, Saarland University Medical Center and Saarland University Faculty of Medicine University, 66421 Homburg, Germany
| | - Hauke S. Heinzow
- Department of Internal Medicine B, University Hospital of Münster, 48149 Münster, Germany;
- Department of Internal Medicine I, Krankenhaus der Barmherzigen Brüder, 54292 Trier, Germany;
| | - Miriam Maschmeier
- Department of Internal Medicine I, Krankenhaus der Barmherzigen Brüder, 54292 Trier, Germany;
| |
Collapse
|
18
|
Xie T, Guo J, Wang W. The Long Noncoding RNA Gall Bladder Cancer-Associated Suppressor of Pyruvate Carboxylase Inhibits the Proliferation, Migration, and Invasion of Colorectal Cancer Cells and Induces Their Apoptosis. Biochem Genet 2024:10.1007/s10528-024-10786-6. [PMID: 38609669 DOI: 10.1007/s10528-024-10786-6] [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: 08/26/2023] [Accepted: 03/15/2024] [Indexed: 04/14/2024]
Abstract
This study aimed to determine the role of the long noncoding RNA (lncRNA) gall bladder cancer-associated suppressor of pyruvate carboxylase (SOD2-1) in the progression of colorectal cancer (CRC). A total of 23 pairs of specimens, including CRC tissues and adjacent normal tissues, were collected, and the expression of lncRNA SOD2-1 (lnc-SOD2-1) was measured. lnc-SOD2-1 function was examined using HCT15 and HCT116 cells. A lnc-SOD2-1 overexpression vector was designed and transfected into both cell lines. MTS and colony formation assays were used to determine cell viability. Flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling assays were performed to measure apoptosis. Cell migration and invasion were evaluated using the Transwell assay. Migration and invasion markers were validated using quantitative reverse transcription-polymerase chain reaction and western blot analysis. The results indicated that the expression of lnc-SOD2-1 was downregulated in CRC tissues. lnc-SOD2-1 overexpression evidently decreased cell viability and led to the formation of fewer cell colonies. lnc-SOD2-1 overexpression induced ~ twofold higher apoptosis than the control group. lnc-SOD2-1 overexpression reduced the proportion of migratory and invasive cells to 50% and 75% of the control group, respectively. lnc-SOD2-1 overexpression significantly decreased the expression of matrix metalloproteinase-2 and -9. In conclusion, lnc-SOD2-1 may act as a tumor suppressor that inhibits the proliferation, migration, and invasion of CRC cells and induces their apoptosis.
Collapse
Affiliation(s)
- Tingting Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianian Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No.250 Changgang East Road, Haizhu District, Guangzhou, 510260, China.
| |
Collapse
|
19
|
Syed RU, Afsar S, Aboshouk NAM, Salem Alanzi S, Abdalla RAH, Khalifa AAS, Enrera JA, Elafandy NM, Abdalla RAH, Ali OHH, Satheesh Kumar G, Alshammari MD. LncRNAs in necroptosis: Deciphering their role in cancer pathogenesis and therapy. Pathol Res Pract 2024; 256:155252. [PMID: 38479121 DOI: 10.1016/j.prp.2024.155252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 04/14/2024]
Abstract
Necroptosis, a controlled type of cell death that is different from apoptosis, has become a key figure in the aetiology of cancer and offers a possible target for treatment. A growing number of biological activities, including necroptosis, have been linked to long noncoding RNAs (lncRNAs), a varied family of RNA molecules with limited capacity to code for proteins. The complex interactions between LncRNAs and important molecular effectors of necroptosis, including mixed lineage kinase domain-like pseudokinase (MLKL) and receptor-interacting protein kinase 3 (RIPK3), will be investigated. We will explore the many methods that LncRNAs use to affect necroptosis, including protein-protein interactions, transcriptional control, and post-transcriptional modification. Additionally, the deregulation of certain LncRNAs in different forms of cancer will be discussed, highlighting their dual function in influencing necroptotic processes as tumour suppressors and oncogenes. The goal of this study is to thoroughly examine the complex role that LncRNAs play in controlling necroptotic pathways and how that regulation affects the onset and spread of cancer. In the necroptosis for cancer treatment, this review will also provide insight into the possible therapeutic uses of targeting LncRNAs. Techniques utilising LncRNA-based medicines show promise in controlling necroptotic pathways to prevent cancer from spreading and improve the effectiveness of treatment.
Collapse
Affiliation(s)
- Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Hail 81442, Saudi Arabia.
| | - S Afsar
- Department of Virology, Sri Venkateswara University, Tirupathi, Andhra Pradesh 517502, India.
| | - Nayla Ahmed Mohammed Aboshouk
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | | | | | - Amna Abakar Suleiman Khalifa
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Jerlyn Apatan Enrera
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Nancy Mohammad Elafandy
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Randa Abdeen Husien Abdalla
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - Omar Hafiz Haj Ali
- Department of Clinical laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 81442, Saudi Arabia
| | - G Satheesh Kumar
- Department of Pharmaceutical Chemistry, College of Pharmacy, Seven Hills College of Pharmacy, Venkataramapuram, Tirupati, India
| | - Maali D Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| |
Collapse
|
20
|
Morabito A, Mercadante E, Muto P, Manzo A, Palumbo G, Sforza V, Montanino A, Sandomenico C, Costanzo R, Esposito G, Totaro G, Cecio RD, Picone C, Porto A, Normanno N, Capasso A, Pinto M, Tracey M, Caropreso G, Pascarella G. Improving the quality of patient care in lung cancer: key factors for successful multidisciplinary team working. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:260-277. [PMID: 38751383 PMCID: PMC11093720 DOI: 10.37349/etat.2024.00217] [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: 06/23/2023] [Accepted: 01/08/2024] [Indexed: 05/18/2024] Open
Abstract
International Guidelines as well as Cancer Associations recommend a multidisciplinary approach to lung cancer care. A multidisciplinary team (MDT) can significantly improve treatment decision-making and patient coordination by putting different physicians and other health professionals "in the same room", who collectively decide upon the best possible treatment. However, this is not a panacea for cancer treatment. The impact of multidisciplinary care (MDC) on patient outcomes is not univocal, while the effective functioning of the MDT depends on many factors. This review presents the available MDT literature with an emphasis on the key factors that characterize high-quality patient care in lung cancer. The study was conducted with a bibliographic search using different electronic databases (PubMed Central, Scopus, Google Scholar, and Google) referring to multidisciplinary cancer care settings. Many key elements appear consolidated, while others emerge as prevalent and actual, especially those related to visible barriers which work across geographic, organizational, and disciplinary boundaries. MDTs must be sustained by strategic management, structured within the entity, and cannot be managed as a separate care process. Furthermore, they need to coordinate with other teams (within and outside the organization) and join with the broad range of services delivered by multiple providers at various points of the cancer journey or within the system, with the vision of integrated care.
Collapse
Affiliation(s)
- Alessandro Morabito
- Thoracic Medical Oncology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Edoardo Mercadante
- Thoracic Surgery, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Paolo Muto
- Radiotherapy, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Anna Manzo
- Thoracic Medical Oncology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Giuliano Palumbo
- Thoracic Medical Oncology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Vincenzo Sforza
- Thoracic Medical Oncology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Agnese Montanino
- Thoracic Medical Oncology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Claudia Sandomenico
- Thoracic Medical Oncology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Raffaele Costanzo
- Thoracic Medical Oncology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Giovanna Esposito
- Thoracic Medical Oncology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Giuseppe Totaro
- Radiotherapy, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Rossella De Cecio
- Pathology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Carmine Picone
- Radiology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Annamaria Porto
- Radiology, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Nicola Normanno
- Cellular Biology and Biotherapy, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Arturo Capasso
- WSB Merito University in Wroclaw, Fabryczna 29-31, 53-609 Wroclaw, Poland
| | - Monica Pinto
- Rehabilitative Medicine Unit, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Maura Tracey
- Rehabilitative Medicine Unit, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| | - Giuseppe Caropreso
- Division of Medical Oncology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Giacomo Pascarella
- Scientific Directorate, Istituto Nazionale Tumori, IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy
| |
Collapse
|
21
|
Lin T, Zhang S, Tang Y, Xiao M, Li M, Gong H, Xie H, Wang Y. ART1 knockdown decreases the IL-6-induced proliferation of colorectal cancer cells. BMC Cancer 2024; 24:354. [PMID: 38504172 PMCID: PMC10953198 DOI: 10.1186/s12885-024-12120-0] [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: 11/23/2023] [Accepted: 03/13/2024] [Indexed: 03/21/2024] Open
Abstract
Colorectal cancer (CRC) is a worldwide health concern. Chronic inflammation is a risk factor for CRC, and interleukin-6 (IL-6) plays a pivotal role in this process. Arginine-specific mono-ADP-ribosyltransferase-1 (ART1) positively regulates inflammatory cytokines. ART1 knockdown reduces the level of glycoprotein 130 (gp130), a key transducer in the IL-6 signalling pathway. However, the relationship between ART1 and IL-6 and the resulting effects on IL-6-induced proliferation in CRC cells remain unclear. The aims of this study were to investigate the effects of ART1 knockdown on IL-6-induced cell proliferation in vitro and use an in vivo murine model to observe the growth of transplanted tumours. The results showed that compared with the control, ART1-sh cancer cells induced by IL-6 exhibited reduced viability, a lower rate of colony formation, less DNA synthesis, decreased protein levels of gp130, c-Myc, cyclin D1, Bcl-xL, and a reduced p-STAT3/STAT3 ratio (P < 0.05). Moreover, mice transplanted with ART1-sh CT26 cells that had high levels of IL-6 displayed tumours with smaller volumes (P < 0.05). ART1 and gp130 were colocalized in CT26, LoVo and HCT116 cells, and their expression was positively correlated in human CRC tissues. Overall, ART1 may serve as a promising regulatory factor for IL-6 signalling and a potential therapeutic target for human CRC.
Collapse
Affiliation(s)
- Ting Lin
- Department of Pathology, Molecular Medicine and Cancer Research Center, Basic Medicine College, Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Shuxian Zhang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Basic Medicine College, Chongqing Medical University, Chongqing, 400016, P.R. China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Yi Tang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Basic Medicine College, Chongqing Medical University, Chongqing, 400016, P.R. China
- Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Ming Xiao
- Department of Pathology, Molecular Medicine and Cancer Research Center, Basic Medicine College, Chongqing Medical University, Chongqing, 400016, P.R. China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Ming Li
- Department of Pathology, Molecular Medicine and Cancer Research Center, Basic Medicine College, Chongqing Medical University, Chongqing, 400016, P.R. China
- Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Hanjuan Gong
- Department of Pathology, Molecular Medicine and Cancer Research Center, Basic Medicine College, Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Hailun Xie
- Department of Pathology, Molecular Medicine and Cancer Research Center, Basic Medicine College, Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Yalan Wang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Basic Medicine College, Chongqing Medical University, Chongqing, 400016, P.R. China.
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, 400016, P.R. China.
- Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P.R. China.
| |
Collapse
|
22
|
Lan D, Wang J, Sun G, Jiang L, Chen Q, Li S, Qu H, Wang Y, Wu B. Abnormal upregulation of NUBP2 contributes to cancer progression in colorectal cancer. Mol Cell Biochem 2024:10.1007/s11010-024-04956-8. [PMID: 38492158 DOI: 10.1007/s11010-024-04956-8] [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: 08/21/2023] [Accepted: 02/03/2024] [Indexed: 03/18/2024]
Abstract
Colorectal cancer (CRC), a digestive tract malignancy with high mortality and morbidity, lacks effective biomarkers for clinical prognosis due to its complex molecular pathogenesis. Nucleotide binding protein 2 (NUBP2) plays a vital role in the assembly of cytosolic Fe/S protein and has been implicated in cancer progression. In this study, we found that NUBP2 was highly expressed in CRC by TCGA database analysis. Subsequently, we verified the expression of NUBP2 in CRC tumor tissues and para-carcinoma tissues using IHC staining, and further investigated its association with clinicopathological parameters. In vitro cell experiments were conducted to assess the role of NUBP2 in CRC by evaluating cell proliferation, migration, and apoptosis upon NUBP2 dysregulation. Furthermore, we established a subcutaneous CRC model to evaluate the impact of NUBP2 on tumor growth in vivo. Additionally, we performed mechanistic exploration using a Human Phospho-Kinase Array-Membrane. Our results showed higher expression of NUBP2 in CRC tissues, which positively correlated with the pathological stage, indicating its involvement in tumor malignancy. Functional studies demonstrated that NUBP2 knockdown reduced cell proliferation, increased apoptosis, and impaired migration ability. Moreover, NUBP2 knockdown inhibited tumor growth in mice. We also observed significant changes in the phosphorylation level of GSK3β upon NUBP2 knockdown or overexpression. Additionally, treatment with CHIR-99021 HCl, an inhibitor of GSK3β, reversed the malignant phenotype induced by NUBP2 overexpression. Overall, this study elucidated the functional role of NUBP2 in CRC progression both in vitro and in vivo, providing insights into the molecular mechanisms underlying CRC and potential implications for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Danfeng Lan
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan, China
| | - Junyu Wang
- Department of General Surgery II, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Guishun Sun
- Department of General Surgery II, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Lixia Jiang
- Department of General Surgery II, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Qiyun Chen
- Department of General Surgery II, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Sha Li
- Department of General Surgery II, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Haiyan Qu
- Department of General Surgery II, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Yibo Wang
- Department of General Surgery II, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China
| | - Bian Wu
- Department of General Surgery II, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jingbi Road, Kunming, 650032, Yunnan, China.
| |
Collapse
|
23
|
Liu J, Liu Y, Zhao Q. Knockdown of ANP32E inhibits colorectal cancer cell growth and glycolysis by regulating the AKT/mTOR pathway. Open Life Sci 2024; 19:20220817. [PMID: 38585643 PMCID: PMC10997116 DOI: 10.1515/biol-2022-0817] [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: 07/31/2023] [Revised: 11/14/2023] [Accepted: 11/23/2023] [Indexed: 04/09/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common tumor, with an increasing number of deaths worldwide each year. Tremendous advances in the diagnosis and treatment of CRC have significantly improved the outcomes for CRC patients. Additionally, accumulating evidence has hinted the relationship between acidic nuclear phosphoprotein 32 family member E (ANP32E) and cancer progression. But the role of ANP32E in CRC remains unclear. In our study, through TCGA database, it was demonstrated that the expression of ANP32E was enhanced in COAD tissues (n = 286). In addition, the mRNA and protein expression of ANP32E was also confirmed to be upregulated in CRC cell lines. Further investigation uncovered that knockdown of ANP32E suppressed cell proliferation and glycolysis, and facilitated cell apoptosis in CRC. Moreover, inhibition of ANP32E inhibited the AKT/mTOR pathway. Through rescue assays, we discovered that the reduced cell proliferation, glycolysis and the enhanced cell apoptosis mediated by ANP32E repression was reversed by SC79 treatment. In summary, ANP32E aggravated the growth and glycolysis of CRC cells by stimulating the AKT/mTOR pathway. This finding suggested that the ANP32E has the potential to be explored as a novel biomarker for CRC treatment.
Collapse
Affiliation(s)
- Jiaojiao Liu
- Department of Clinical Laboratory, Beihua University Affiliated Hospital, No. 12, Jiefang Middle Road, Jilin, Jilin, 132011, China
| | - Yanchao Liu
- Department of Clinical Laboratory, Jilin Gynecology and Obstetrics Hospital, Jilin, Jilin, 130211, China
| | - Qi Zhao
- Department of Clinical Laboratory, Beihua University Affiliated Hospital, No. 12, Jiefang Middle Road, Jilin, Jilin, 132011, China
| |
Collapse
|
24
|
Wang R, Lin X. GPC2 as a diagnostic and prognostic marker regulated progression of colorectal cancer. Arab J Gastroenterol 2024; 25:51-57. [PMID: 38220478 DOI: 10.1016/j.ajg.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/12/2023] [Accepted: 11/27/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND AND STUDY AIMS Glypican 2 (GPC2) is a member of the glypican gene family and is expressed in multiple kinds of cancer. However, the function and mechanism of GPC2 in colorectal cancer remains unclear. In this study, we aimed to identify the role of GPC2 on tumor cell proliferation and survival in colorectal cancer. PATIENTS AND METHODS Ten pairs of colon cancer and matched normal colon tissues were collected in this research. GEPIA was used to analysis the GPC2 gene expression profile in TGCA data base. RT-qPCR and western blot assay were performed to determine the mRNA and protein expressions. CCK-8, Flow cytometry and colon formation assay were applied to evaluate cell viability. IHC staining was performed to evaluate the protein expression in tissues. The function of GPC2 in vivo was verified by an animal model of colon cancer. RESULTS Through the bioinformatics analysis and qRT-PCR validation, we found that GPC2 was upregulated in the colon cancer tissues and cells. GPC2 knockdown suppressed cell proliferation in vitro and in vivo was confirmed by the results of CCK-8, colony formation assays, and tumor xenograft models. Moreover, by the analysis of flow cytometry assay and gain-or-loss function experiments, we discovered that CEP164 was highly associated with the expression state of GPC2, and mediated G2/M-phase arrest in GPC2-downregulated tumor cells. CONCLUSION GPC2 might be a novel oncogenic gene in colorectal cancer, suggesting that it could be a considerable marker for the diagnosis and prognosis of colorectal cancer.
Collapse
Affiliation(s)
- Rugang Wang
- Tumors Ward 3, Harbin Chest Hospital, Harbin 150056, China
| | - Xuan Lin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310000, China.
| |
Collapse
|
25
|
Chen H, Yao J, Shan Z, Wei Y, You S, Li D, Zhang Y. To Assess the Role of microRNA-451 in the Progression and Metastasis of Colorectal Cancer. Appl Biochem Biotechnol 2024; 196:1044-1057. [PMID: 37318687 DOI: 10.1007/s12010-023-04538-2] [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] [Accepted: 04/11/2023] [Indexed: 06/16/2023]
Abstract
The study aimed to determine the expression of miR451 in colorectal cancer (CRC) subjects with CRC cells, and the role of miR451 in colorectal cancer cells. In October 2020, ATC purchased CRC and normal mucosal cell lines of CRC and implanted them in DMEM with 10% fetal serum. The suitability of the HT29 cell line is verified using the STR profile. In an incubator with 5% CO2, enlarged cells were placed at 37 °C. TCGA data was used to select the top 120 patients with a high voice and the lowest 120 patients with a low voice. Cells were collected and coated with Annexin V and PE according to the manufacturer's instructions after 24.0 h. After that, the cells were separated. Cells were also tested using flow cytometry. HCT-120 cells were transplanted into a concentration of 5×105/ml cells in 6-source plates. HCT120 cells in the experimental group were combined with miR451 mimics, miR451 inhibitors, or miR451 miR + SMAD4B for 12 h at 37 °C, and cells were collected 24 h later at 37 °C. The sample was injected with 5 ml of Annexin VFITC and PE. Compared with normal colorectal mucosal cells, CRC cell lines decreased miR451 expression levels (fetal human cells (FHC) and HCoEpiC). Then, the HCT120 cells were transfected with miR451 inhibitors, and 72 h after transfection, say of miR451 was normal. There was a significant decrease in cell function in the miR451mimic groups, but an increase when the miR451 was blocked. The proliferation of cancer cells was prevented and chemotherapy was effective when miR451 was overexpressed. The SMAD4 gene provides instructions for making a protein involved in transmitting chemical signals from the cell surface to the nucleus. The SMAD4B expression was tested by RT-qPCR and Western blotting after 72.0 h of transmission. The mRNA and protein expression of SMAD4B decreased significantly when miR451 was significantly higher than when inhibited, as revealed in the results of this study. Seventy-two hours after transplantation, mRNA levels and SMAD4B proteins were measured in HCT120 cells. In addition, the researchers in this study investigated whether miR451 was associated with SMAD4B-directed control of CRC growth and migration. It was found that SMAD4B is highly expressed in both CRC and para-cancer tissues while using the TCGA database to detect SMAD4B expression. Patients with CRC with SMAD4B have a severe prognosis. MiR451 is sensitive to depressive disorders by targeting SMAD4B, according to these studies. We found that miR451 inhibited cell growth and migration, made CRC cells more readily available in chemotherapy, and did so by targeting SMAD4B. The findings suggest that miR451 and its genetic predisposition, SMAD4B, may help predict the prognosis and course of cancer patients. Treatments that target the miR451/SMAD4B axis may be helpful to people with CRC.
Collapse
Affiliation(s)
- Hong Chen
- Department of General Surgery, Dushu Lake Hospital Affiliated to Soochow University, No 9 of Chongwen Road, Suzhou, Jiangsu Province, 215000, China
| | - Jun Yao
- Department of General Surgery, Dushu Lake Hospital Affiliated to Soochow University, No 9 of Chongwen Road, Suzhou, Jiangsu Province, 215000, China
| | - Zhili Shan
- Department of General Surgery, Dushu Lake Hospital Affiliated to Soochow University, No 9 of Chongwen Road, Suzhou, Jiangsu Province, 215000, China
| | - YiJun Wei
- Department of General Surgery, Dushu Lake Hospital Affiliated to Soochow University, No 9 of Chongwen Road, Suzhou, Jiangsu Province, 215000, China
| | - Shijie You
- Department of General Surgery, Dushu Lake Hospital Affiliated to Soochow University, No 9 of Chongwen Road, Suzhou, Jiangsu Province, 215000, China
| | - Dechun Li
- Department of General Surgery, Dushu Lake Hospital Affiliated to Soochow University, No 9 of Chongwen Road, Suzhou, Jiangsu Province, 215000, China
| | - Yi Zhang
- Department of General Surgery, Dushu Lake Hospital Affiliated to Soochow University, No 9 of Chongwen Road, Suzhou, Jiangsu Province, 215000, China.
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No 899 of Pinghai Road, Suzhou, Jiangsu Province, 215000, China.
| |
Collapse
|
26
|
Cao Y, Li J, Zhang G, Fang H, Du Y, Liang Y. KLF15 transcriptionally activates LINC00689 to inhibit colorectal cancer development. Commun Biol 2024; 7:130. [PMID: 38273088 PMCID: PMC10810960 DOI: 10.1038/s42003-023-05757-3] [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: 04/21/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024] Open
Abstract
Colorectal cancer is a grievous health concern, we have proved long non-coding RNA LINC00689 is considered as a potential diagnosis biomarker for colorectal cancer, and it is necessary to further investigate its upstream and downstream mechanisms. Here, we show that KLF15, a transcription factor, exhibits the reduced expression in colorectal cancer. KLF15 suppresses the proliferative and metastatic capacities of colorectal cancer cells both in vitro and in vivo by transcriptionally activating LINC00689. Subsequently, LINC00689 recruits PTBP1 protein to enhance the stability of LATS2 mRNA in the cytoplasm. This stabilization causes the suppression of the YAP1/β-catenin pathway and its target downstream genes. Our findings highlight a regulatory network involving KLF15, LINC00689, PTBP1, LATS2, and the YAP1/β-catenin pathway in colorectal cancer, shedding light on potential therapeutic targets for colorectal cancer therapy.
Collapse
Affiliation(s)
- Yan Cao
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Jian Li
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Gang Zhang
- Department 2 of Gastrointestinal Surgery, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou People's Hospital, Haikou, 570208, Hainan Province, PR China
| | - Hao Fang
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Yongliang Du
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Yan Liang
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China.
| |
Collapse
|
27
|
Xu S, Zhong F, jiang J, Yao F, Li M, Tang M, Cheng Y, Yang Y, Wen W, Zhang X, Huang B, Wang X. High Expression of SRSF10 Promotes Colorectal Cancer Progression by Aberrant Alternative Splicing of RFC5. Technol Cancer Res Treat 2024; 23:15330338241271906. [PMID: 39110418 PMCID: PMC11307364 DOI: 10.1177/15330338241271906] [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: 03/10/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) remains a global health concern with persistently high incidence and mortality rates. However, the specific pathogenesis of CRC remains poorly understood. This study aims to investigate the role and pathogenesis of serine and arginine rich splicing factor 10 (SRSF10) in colorectal cancer. METHODS Bioinformatics analysis was employed to predict SRSF10 gene expression in CRC patients. Functional experiments involving SRSF10 knockdown and overexpression were conducted using CCK8, transwell, scratch assay, and flow cytometry. Additionally, the PRIdictor website was utilized to predict the SRSF10 interaction site with RFC5. The identification of different transcripts of SRSF10-acting RFC5 pre-mRNA was achieved through agarose gel electrophoresis. RESULT The knockdown of SRSF10 inhibited the proliferation and migration ability of CRC cells, while promoting apoptosis and altering the DNA replication of CRC cells. Conversely, when SRSF10 was highly expressed, it enhanced the proliferation and migration ability of CRC cells and caused changes in the cell cycle of colorectal cancer cells. This study revealed a change in the replicating factor C subunit 5 (RFC5) gene in colorectal cancer cells following SRSF10 knockdown. Furthermore, it was confirmed that SRSF10 increased RFC5 exon2-AS1(S) transcription variants, thereby promoting the development of colorectal cancer through AS1 exclusion to exon 2 of RFC5. CONCLUSION In summary, this study demonstrates that SRSF10 promotes the progression of colorectal cancer by generating an aberrantly spliced exclusion isoform of AS1 within RFC5 exon 2. These findings suggest that SRSF10 could serve as a crucial target for the clinical diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Shuai Xu
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fangmin Zhong
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Junyao jiang
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fangyi Yao
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Meiyong Li
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mengxin Tang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ying Cheng
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yulin Yang
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, No. 461 BaYi Boulevard, Nanchang 330006, Jiangxi, China
| | - Wen Wen
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, No. 461 BaYi Boulevard, Nanchang 330006, Jiangxi, China
| | - Xueru Zhang
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, No. 461 BaYi Boulevard, Nanchang 330006, Jiangxi, China
| | - Bo Huang
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, No. 461 BaYi Boulevard, Nanchang 330006, Jiangxi, China
| | - Xiaozhong Wang
- Department of Clinical Laboratory, Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, No. 461 BaYi Boulevard, Nanchang 330006, Jiangxi, China
| |
Collapse
|
28
|
Mohan S, Hakami MA, Dailah HG, Khalid A, Najmi A, Zoghebi K, Halawi MA, Alotaibi TM. From inflammation to metastasis: The central role of miR-155 in modulating NF-κB in cancer. Pathol Res Pract 2024; 253:154962. [PMID: 38006837 DOI: 10.1016/j.prp.2023.154962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 11/27/2023]
Abstract
Cancer is a multifaceted, complex disease characterized by unchecked cell growth, genetic mutations, and dysregulated signalling pathways. These factors eventually cause evasion of apoptosis, sustained angiogenesis, tissue invasion, and metastasis, which makes it difficult for targeted therapeutic interventions to be effective. MicroRNAs (miRNAs) are essential gene expression regulators linked to several biological processes, including cancer and inflammation. The NF-κB signalling pathway, a critical regulator of inflammatory reactions and oncogenesis, has identified miR-155 as a significant participant in its modulation. An intricate network of transcription factors known as the NF-κB pathway regulates the expression of genes related to inflammation, cell survival, and immunological responses. The NF-κB pathway's dysregulation contributes to many cancer types' development, progression, and therapeutic resistance. In numerous cancer models, the well-studied miRNA miR-155 has been identified as a crucial regulator of NF-κB signalling. The p65 subunit and regulatory molecules like IκB are among the primary targets that miR-155 directly targets to alter NF-κB activity. The molecular processes by which miR-155 affects the NF-κB pathway are discussed in this paper. It also emphasizes the miR-155's direct and indirect interactions with important NF-κB cascade elements to control the expression of NF-κB subunits. We also investigate how miR-155 affects NF-κB downstream effectors in cancer, including inflammatory cytokines and anti-apoptotic proteins.
Collapse
Affiliation(s)
- Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al, Quwayiyah, Shaqra University, Riyadh, Saudi Arabia.
| | - Hamad Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan 45142, Saudi Arabia
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Maryam A Halawi
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | | |
Collapse
|
29
|
Zhang Z, Wang Z, Fan H, Li J, Ding J, Zhou G, Yuan C. The Indispensable Roles of GMDS and GMDS-AS1 in the Advancement of Cancer: Fucosylation, Signal Pathway and Molecular Pathogenesis. Mini Rev Med Chem 2024; 24:1712-1722. [PMID: 38591197 DOI: 10.2174/0113895575285276240324080234] [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: 12/01/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/10/2024]
Abstract
Fucosylation is facilitated by converting GDP-mannose to GDP-4-keto-6-deoxymannose, which GDP-mannose 4,6-dehydratase, a crucial enzyme in the route, carries out. One of the most prevalent glycosylation alterations linked to cancer has reportedly been identified as fucosylation. There is mounting evidence that GMDS is intimately linked to the onset and spread of cancer. Furthermore, the significance of long-chain non-coding RNAs in the development and metastasis of cancer is becoming more well-recognized, and the regulatory mechanism of lncRNAs has emerged as a prominent area of study in the biological sciences. GMDS-AS1, an antisense RNA of GMDS, was discovered to have the potential to be an oncogene. We have acquired and analyzed relevant data to understand better how GMDS-AS1 and its lncRNA work physiologically and in tumorigenesis and progression. Additionally, we have looked into the possible effects of these molecules on cancer treatment approaches and patient outcomes. The physiological roles and putative processes of GMDS and lncRNA GMDS-AS1 throughout the development and progression of tumors have been assembled and examined. We also examined how these chemicals might affect patient prognosis and cancer therapy approaches. GMDS and GMDS-AS1 were determined to be research subjects by searching and gathering pertinent studies using the PubMed system. The analysis of these research articles demonstrated the close relationship between GMDS and GMDS-AS1 and tumorigenesis and the factors that influence them. GMDS plays a vital role in regulating fucosylation. The related antisense gene GMDS-AS1 affects the biological behaviors of cancer cells through multiple pathways, including the key processes of proliferation, migration, invasion, and apoptosis, providing potential biomarkers and therapeutic targets for cancer treatment and prognosis assessment.
Collapse
Affiliation(s)
- Ziyan Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Zhuowei Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Hong Fan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Jiayi Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Jiaqi Ding
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Gang Zhou
- College of Traditional Chinese Medicine, China Three Gorges University, Yichang 443002, China
- Yichang Hospital of Traditional Chinese Medicine, Yichang 443002, China
| | - Chengfu Yuan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| |
Collapse
|
30
|
Cai Z, Wang S, Zhou H, Cao D. Low expression of ZHX3 is associated with progression and poor prognosis in colorectal cancer. Transl Oncol 2024; 39:101829. [PMID: 37979559 PMCID: PMC10656720 DOI: 10.1016/j.tranon.2023.101829] [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: 08/02/2023] [Revised: 10/31/2023] [Accepted: 11/10/2023] [Indexed: 11/20/2023] Open
Abstract
Accumulating studies suggest that ZHX3, the member of ZHX family, is involved in a variety of biological functions such as development and differentiation. Recently, ZHX3 may also be involved in the progression of several cancer types including renal cancer, gastric cancer, liver cancer and breast cancer. However, the potential role of ZHX3 in colorectal cancer (CRC) is still unknown. In this study, we analyzed the protein levels of ZHX3 by immunohistochemistry and evaluated its relationship with the clinicopathological features and prognosis in 286 CRC patients. In vitro cell proliferation assay, plate colony formation assay and xenograft model in nude mice were applied to evaluate CRC cell proliferative ability. Our results showed that the expression of ZHX3 was significantly downregulated in CRC tissues compared with paired adjacent nontumor tissues. Furthermore, the ZHX3 expression was found to have a strong correlation with tumor size, tumor invasion depth and TNM stage. Kaplan-Meier analysis demonstrated that low ZHX3 expression was related to a poorer overall survival and disease-free survival in CRC patients. In addition, cox's proportional hazards analysis indicated that low ZHX3 expression was an independent prognostic indicator of poor prognosis. Functionally, reduced expression of ZHX3 promotes the proliferation of CRC cells both in vitro and in vivo. Conversely, overexpression of ZHX3 inhibited the growth of CRC cells, indicated that ZHX3 was significantly correlated with CRC progression. Our results indicate for the first time that ZHX3 may be a potential marker of cancer prognosis and CRC recurrence.
Collapse
Affiliation(s)
- Zhai Cai
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Songsheng Wang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huabin Zhou
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ding Cao
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
31
|
Du W, Xia G, Chen L, Geng L, Xu R, Han Q, Ying X, Yu H. Clinical value of SLC12A9 for diagnosis and prognosis in colorectal cancer. Aging (Albany NY) 2023; 15:15419-15433. [PMID: 38157260 PMCID: PMC10781446 DOI: 10.18632/aging.205360] [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: 08/29/2023] [Accepted: 11/09/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE The goal of the study is to assess the clinical value and the potential mechanism of SLC12A9 combing transcriptome and single cell sequencing data. METHODS In this study, the expression level and the receiver operating characteristic curve analysis of SLC12A9 in CRC and normal tissue were analyzed in multiple data cohort. The standardized mean difference (SMD) calculation and the summary receiver operating characteristic (SROC) analysis were performed further to detect its diagnostic ability and expression level. KM survival analysis was performed to assess the prognosis value of SLC12A9. The expression level of SLC12A9 in different clinical characteristics was analyzed to explore the clinical value. Single cell data was studied to reveal the potential mechanism of SLC12A9. The correlation analysis of immunoinfiltration was performed to detect the potential immune cell related to SLC12A9. The nomogram was drawn to assess the probable mortality rate of CRC patient. RESULTS We found that SLC12A9 was significantly up-regulated with the moderate diagnostic value in CRC. Patients with overexpressed SLC12A9 had a worse prognosis. SLC12A9 was related to Age, Pathologic N stage, Pathologic M stage, Lymphatic invasion and Pathologic stage (p < 0.05). The 1, 3 and 5-year survival rates of patient named TCGA-G4-6309 are 0.959, 0.897 and 0.827. PCR also showed that SLC12A9 was overexpressed in CRC comparing with normal tissue. CONCLUSION In conclusion, our study comprehensively analyzed the clinical value of SLC12A9 and its potential mechanism, as well as immune cell infiltration, which may accelerate the diagnosis and improve the prognosis of CRC.
Collapse
Affiliation(s)
- Wang Du
- Department of General Surgery, Fuyang Hospital of Anhui Medical University, Fuyang, Anhui 236000, China
| | - Guozhi Xia
- Department of General Surgery, Fuyang Hospital of Anhui Medical University, Fuyang, Anhui 236000, China
| | - Liang Chen
- Department of Hepatobiliary and Pancreatic Surgery, Conversion therapy center for Hepatobiliary and Pancreatic Tumors, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314041, China
| | - Lingjun Geng
- Department of General Surgery, Fuyang Hospital of Anhui Medical University, Fuyang, Anhui 236000, China
| | - Rubin Xu
- Department of General Surgery, Fuyang Hospital of Anhui Medical University, Fuyang, Anhui 236000, China
| | - Qingqing Han
- Department of General Surgery, Fuyang Hospital of Anhui Medical University, Fuyang, Anhui 236000, China
| | - Xiaomei Ying
- Suzhou Hospital of Anhui Medical University, Suzhou, Anhui 234000, China
| | - Hongzhu Yu
- Department of General Surgery, Fuyang Hospital of Anhui Medical University, Fuyang, Anhui 236000, China
| |
Collapse
|
32
|
Santos MD, Barros I, Brandão P, Lacerda L. Amino Acid Profiles in the Biological Fluids and Tumor Tissue of CRC Patients. Cancers (Basel) 2023; 16:69. [PMID: 38201497 PMCID: PMC10778074 DOI: 10.3390/cancers16010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Amino acids are the building blocks of proteins and essential players in pathways such as the citric acid and urea cycle, purine and pyrimidine biosynthesis, and redox cell signaling. Therefore, it is unsurprising that these molecules have a significant role in cancer metabolism and its metabolic plasticity. As one of the most prevalent malign diseases, colorectal cancer needs biomarkers for its early detection, prognostic, and prediction of response to therapy. However, the available biomarkers for this disease must be more powerful and present several drawbacks, such as high costs and complex laboratory procedures. Metabolomics has gathered substantial attention in the past two decades as a screening platform to study new metabolites, partly due to the development of techniques, such as mass spectrometry or liquid chromatography, which have become standard practice in diagnostic procedures for other diseases. Extensive metabolomic studies have been performed in colorectal cancer (CRC) patients in the past years, and several exciting results concerning amino acid metabolism have been found. This review aims to gather and present findings concerning alterations in the amino acid plasma pool of colorectal cancer patients.
Collapse
Affiliation(s)
- Marisa Domingues Santos
- Colorectal Unit, Hospital de Santo António, Centro Hospitalar Universitário de Santo António, 4050-651 Porto, Portugal;
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - Ivo Barros
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
| | - Pedro Brandão
- Colorectal Unit, Hospital de Santo António, Centro Hospitalar Universitário de Santo António, 4050-651 Porto, Portugal;
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - Lúcia Lacerda
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
- Genetic Laboratory Service, Centro de Genética Médica Jacinto de Magalhães, Centro Hospitalar Universitário de Santo António, 4050-651 Porto, Portugal
| |
Collapse
|
33
|
Liu H, Wu X, Wang D, Li Q, Zhang X, Xu L. Unveiling the role of miR-137-3p/miR-296-5p/SERPINA3 signaling in colorectal cancer progression: integrative analysis of gene expression profiles and in vitro studies. BMC Med Genomics 2023; 16:327. [PMID: 38087342 PMCID: PMC10714458 DOI: 10.1186/s12920-023-01763-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a prevalent malignancy worldwide, with increasing incidence and mortality rates. Although treatment options have improved, CRC remains a leading cause of death due to metastasis. Early intervention can significantly improve patient outcomes, making it crucial to understand the molecular mechanisms underlying CRC metastasis. In this study, we performed bioinformatics analysis to identify potential genes associated with CRC metastasis. METHODS We downloaded and integrated gene expression datasets (GSE89393, GSE100243, and GSE144259) from GEO database. Differential expression analysis was conducted, followed by Gene Ontology (GO) functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The hub gene SERPINA3 was selected for further in vitro functional studies. Additionally, the role of miR-137-3p/miR-296-5p/ Serpin family A member 3 (SERPINA3) in CRC cell function was investigated using in vitro assays. RESULTS Analysis of the gene expression datasets revealed differentially expressed genes (DEGs) associated with CRC metastasis. GO analysis showed enrichment in biological processes such as blood coagulation regulation and wound healing. Cellular component analysis highlighted extracellular matrix components and secretory granules. Molecular function analysis identified activities such as serine-type endopeptidase inhibition and lipoprotein receptor binding. KEGG analysis revealed involvement in pathways related to complement and coagulation cascades, cholesterol metabolism, and immune responses. The common DEGs among the datasets were further investigated. We identified SERPINA3 as a hub gene associated with CRC metastasis. SERPINA3 exerted enhanced effects on migration, proliferation and epithelial-mesenchymal transition (EMT) and inhibitory effects on caspase-3/-9 activities in HT29 and SW620 cells. MiR-137-3p overexpression increased activities of caspase-3/-9, decreased migration and proliferation, and also repressed EMT in HT29 cells, which were obviously attenuated by SERPINA3 enforced overexpression. Consistently, SERPINA3 enforced overexpression also largely reversed miR-296-5p mimics-induced increased in activities of caspase-3/-9, decrease in migration, proliferation and EMT in HT29 cells. CONCLUSION Through bioinformatics analysis, we identified potential genes associated with CRC metastasis. The functional studies focusing on SERPINA3/miR-137-3p/miR-296-5p further consolidated its role in regulating CRC progression. Our findings provide insights into novel mechanisms underlying CRC metastasis and might contribute to the development of effective treatment strategies. However, the role of SERPINA3/miR-137-3p/miR-296-5p signaling in CRC still requires further investigation.
Collapse
Affiliation(s)
- Huimin Liu
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, China
| | - Xingxing Wu
- Department of Pediatric Surgery, The Second People's Hospital of Lianyungang, Lianyungang, China
| | - Dandan Wang
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, China
| | - Quanxi Li
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, China
| | - Xin Zhang
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, China
| | - Liang Xu
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang, China.
| |
Collapse
|
34
|
Ma Y, Wang Z, Sun J, Tang J, Zhou J, Dong M. Investigating the Diagnostic and Therapeutic Potential of SREBF2-Related Lipid Metabolism Genes in Colon Cancer. Onco Targets Ther 2023; 16:1027-1042. [PMID: 38107762 PMCID: PMC10723182 DOI: 10.2147/ott.s428150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/08/2023] [Indexed: 12/19/2023] Open
Abstract
Purpose Colon cancer is one of the leading causes of death worldwide, and screening of effective molecular markers for the diagnosis is prioritised for prevention and treatment. This study aimed to investigate the diagnostic and predictive potential of genes related to the lipid metabolism pathway, regulated by a protein called sterol-regulatory element-binding transcription Factor 2 (SREBF2), for colon cancer and patient outcomes. Methods We used machine-learning algorithms to identify key genes associated with SREBF2 in colon cancer based on a public database. A nomogram was created to assess the diagnostic value of these genes and validated in the Cancer Genome Atlas. We also analysed the relationship between these genes and the immune microenvironment of colon tumours, as well as the correlation between gene expression and clinicopathological characteristics and prognosis in the China Medical University (CMU) clinical cohort. Results Three genes, 7-dehydrocholesterol reductase (DHCR7), hydroxysteroid 11-beta dehydrogenase 2 (HSD11B2), and Ral guanine nucleotide dissociation stimulator-like 1 (RGL1), were identified as hub genes related to SREBF2 and colon cancer. Using the TCGA dataset, receiver operating characteristic curve analysis showed the area under the curve values of 0.943, 0.976, and 0.868 for DHCR7, HSD11B2, and RGL1, respectively. In the CMU cohort, SREBF2 and DHCR7 expression levels were correlated with TNM stage and tumour invasion depth (P < 0.05), and high DHCR7 expression was related to poor prognosis of colon cancer (P < 0.05). Furthermore, DHCR7 gene expression was positively correlated with the abundance of M0 and M1 macrophages and inversely correlated with the abundance of M2 macrophages, suggesting that the immune microenvironment may play a role in colon cancer surveillance. There was a correlation between SREBF2 and DHCR7 expression across cancers in the TCGA database. Conclusion This study highlights the potential of DHCR7 as a diagnostic marker and therapeutic target for colon cancer.
Collapse
Affiliation(s)
- Yuteng Ma
- Department of Gastrointestinal Surgery, First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Jian Sun
- Department of Gastrointestinal Surgery, First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Jingtong Tang
- Department of Gastrointestinal Surgery, First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Jianping Zhou
- Department of Gastrointestinal Surgery, First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Ming Dong
- Department of Gastrointestinal Surgery, First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| |
Collapse
|
35
|
Wang H, Wang W, Wang Z, Li X. Transcriptomic correlates of cell cycle checkpoints with distinct prognosis, molecular characteristics, immunological regulation, and therapeutic response in colorectal adenocarcinoma. Front Immunol 2023; 14:1291859. [PMID: 38143740 PMCID: PMC10749195 DOI: 10.3389/fimmu.2023.1291859] [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: 09/10/2023] [Accepted: 11/22/2023] [Indexed: 12/26/2023] Open
Abstract
Backgrounds Colorectal adenocarcinoma (COAD), accounting for the most common subtype of colorectal cancer (CRC), is a kind of malignant digestive tumor. Some cell cycle checkpoints (CCCs) have been found to contribute to CRC progression, whereas the functional roles of a lot of CCCs, especially the integrated role of checkpoint mechanism in the cell cycle, remain unclear. Materials and methods The Genomic Data Commons (GDC) The Cancer Genome Atlas (TCGA) COAD cohort was retrieved as the training dataset, and GSE24551 and GSE29623 were downloaded from Gene Expression Omnibus (GEO) as the validation datasets. A total of 209 CCC-related genes were derived from the Gene Ontology Consortium and were subsequently enrolled in the univariate, multivariate, and least absolute shrinkage and selection operator (LASSO) Cox regression analyses, finally defining a CCC signature. Cell proliferation and Transwell assay analyses were utilized to evaluate the functional roles of signature-related CCCs. The underlying CCC signature, molecular characteristics, immune-related features, and therapeutic response were finally estimated. The Genomics of Drug Sensitivity in Cancer (GDSC) database was employed for the evaluation of chemotherapeutic responses. Results The aberrant gene expression of CCCs greatly contributed to COAD development and progression. Univariate Cox regression analysis identified 27 CCC-related genes significantly affecting the overall survival (OS) of COAD patients; subsequently, LASSO analysis determined a novel CCC signature. Noticeably, CDK5RAP2, MAD1L1, NBN, RGCC, and ZNF207 were first identified to be correlated with the prognosis of COAD, and it was proven that all of them were significantly correlated with the proliferation and invasion of HCT116 and SW480 cells. In TCGA COAD cohort, CCC signature robustly stratified COAD patients into high and low CCC score groups (median OS: 57.24 months vs. unreached, p< 0.0001), simultaneously, with the good AUC values for OS prediction at 1, 2, and 3 years were 0.74, 0.78, and 0.77. Furthermore, the prognostic capacity of the CCC signature was verified in the GSE24551 and GSE29623 datasets, and the CCC signature was independent of clinical features. Moreover, a higher CCC score always indicated worse OS, regardless of clinical features, histological subtypes, or molecular subgroups. Intriguingly, functional enrichment analysis confirmed the CCC score was markedly associated with extracellular, matrix and immune (chemokine)-related signaling, cell cycle-related signaling, and metabolisms. Impressively, a higher CCC score was positively correlated with a majority of chemokines, receptors, immunostimulators, and anticancer immunity, indicating a relatively immune-promoting microenvironment. In addition, GSE173839, GSE25066, GSE41998, and GSE194040 dataset analyses of the underlying CCC signature suggested that durvalumab with olaparib and paclitaxel, taxane-anthracycline chemotherapy, neoadjuvant cyclophosphamide/doxorubicin with ixabepilone or paclitaxel, and immunotherapeutic strategies might be suitable for COAD patients with higher CCC score. Eventually, the GDSC database analysis showed that lower CCC scores were likely to be more sensitive to 5-fluorouracil, bosutinib, gemcitabine, gefitinib, methotrexate, mitomycin C, and temozolomide, while patients with higher CCC score seemed to have a higher level of sensitivity to bortezomib and elesclomol. Conclusion The novel CCC signature exhibited a good ability for prognosis prediction for COAD patients, and the CCC score was found to be highly correlated with molecular features, immune-related characteristics, and therapeutic responses, which would greatly promote clinical management and precision medicine for COAD.
Collapse
Affiliation(s)
- Heng Wang
- Department of Colorectal Surgery, Shanghai Yangpu Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Wei Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhen Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xu Li
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
36
|
Ding W, Wang JX, Wu JZ, Liu AC, Jiang LL, Zhang HC, Meng Y, Liu BY, Peng GJ, Lou EZ, Mao Q, Zhou H, Tang DL, Chen X, Liu JB, Shi XP. Targeting proteasomal deubiquitinases USP14 and UCHL5 with b-AP15 reduces 5-fluorouracil resistance in colorectal cancer cells. Acta Pharmacol Sin 2023; 44:2537-2548. [PMID: 37528233 PMCID: PMC10692219 DOI: 10.1038/s41401-023-01136-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 07/09/2023] [Indexed: 08/03/2023] Open
Abstract
5-Fluorouracil (5-FU) is the first-line treatment for colorectal cancer (CRC) patients, but the development of acquired resistance to 5-FU remains a big challenge. Deubiquitinases play a key role in the protein degradation pathway, which is involved in cancer development and chemotherapy resistance. In this study, we investigated the effects of targeted inhibition of the proteasomal deubiquitinases USP14 and UCHL5 on the development of CRC and resistance to 5-FU. By analyzing GEO datasets, we found that the mRNA expression levels of USP14 and UCHL5 in CRC tissues were significantly increased, and negatively correlated with the survival of CRC patients. Knockdown of both USP14 and UCHL5 led to increased 5-FU sensitivity in 5-FU-resistant CRC cell lines (RKO-R and HCT-15R), whereas overexpression of USP14 and UCHL5 in 5-FU-sensitive CRC cells decreased 5-FU sensitivity. B-AP15, a specific inhibitor of USP14 and UCHL5, (1-5 μM) dose-dependently inhibited the viability of RKO, RKO-R, HCT-15, and HCT-15R cells. Furthermore, treatment with b-AP15 reduced the malignant phenotype of CRC cells including cell proliferation and migration, and induced cell death in both 5-FU-sensitive and 5-FU-resistant CRC cells by impairing proteasome function and increasing reactive oxygen species (ROS) production. In addition, b-AP15 inhibited the activation of NF-κB pathway, suppressing cell proliferation. In 5-FU-sensitive and 5-FU-resistant CRC xenografts nude mice, administration of b-AP15 (8 mg·kg-1·d-1, intraperitoneal injection) effectively suppressed the growth of both types of tumors. These results demonstrate that USP14 and UCHL5 play an important role in the development of CRC and resistance to 5-FU. Targeting USP14 and UCHL5 with b-AP15 may represent a promising therapeutic strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Wa Ding
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Jin-Xiang Wang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Precision Medicine Center, Department of Biobank, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jun-Zheng Wu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Ao-Chu Liu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Li-Ling Jiang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Hai-Chuan Zhang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Yi Meng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Bing-Yuan Liu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Guan-Jie Peng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - En-Zhe Lou
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Qiong Mao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Huan Zhou
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Dao-Lin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xin Chen
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China.
| | - Jin-Bao Liu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China.
| | - Xian-Ping Shi
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China.
| |
Collapse
|
37
|
Ogbodo UC, Enejoh OA, Okonkwo CH, Gnanasekar P, Gachanja PW, Osata S, Atanda HC, Iwuchukwu EA, Achilonu I, Awe OI. Computational identification of potential inhibitors targeting cdk1 in colorectal cancer. Front Chem 2023; 11:1264808. [PMID: 38099190 PMCID: PMC10720044 DOI: 10.3389/fchem.2023.1264808] [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: 07/21/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction: Despite improved treatment options, colorectal cancer (CRC) remains a huge public health concern with a significant impact on affected individuals. Cell cycle dysregulation and overexpression of certain regulators and checkpoint activators are important recurring events in the progression of cancer. Cyclin-dependent kinase 1 (CDK1), a key regulator of the cell cycle component central to the uncontrolled proliferation of malignant cells, has been reportedly implicated in CRC. This study aimed to identify CDK1 inhibitors with potential for clinical drug research in CRC. Methods: Ten thousand (10,000) naturally occurring compounds were evaluated for their inhibitory efficacies against CDK1 through molecular docking studies. The stability of the lead compounds in complex with CDK1 was evaluated using molecular dynamics simulation for one thousand (1,000) nanoseconds. The top-scoring candidates' ADME characteristics and drug-likeness were profiled using SwissADME. Results: Four hit compounds, namely, spiraeoside, robinetin, 6-hydroxyluteolin, and quercetagetin were identified from molecular docking analysis to possess the least binding scores. Molecular dynamics simulation revealed that robinetin and 6-hydroxyluteolin complexes were stable within the binding pocket of the CDK1 protein. Discussion: The findings from this study provide insight into novel candidates with specific inhibitory CDK1 activities that can be further investigated through animal testing, clinical trials, and drug development research for CRC treatment.
Collapse
Affiliation(s)
| | - Ojochenemi A. Enejoh
- Genomics and Bioinformatics Department, National Biotechnology Development Agency, Abuja, Nigeria
| | - Chinelo H. Okonkwo
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Nigeria
| | | | - Pauline W. Gachanja
- Department of Biochemistry and Biotechnology, Pwani University, Kilifi, Kenya
| | - Shamim Osata
- Department of Biochemistry, University of Nairobi, Nairobi, Kenya
| | - Halimat C. Atanda
- Biotechnology Department, Federal University of Technology, Akure, Nigeria
| | - Emmanuel A. Iwuchukwu
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, Faculty of Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Ikechukwu Achilonu
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, Faculty of Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Olaitan I. Awe
- Department of Computer Science, University of Ibadan, Ibadan, Nigeria
- African Society for Bioinformatics and Computational Biology, Cape Town, South Africa
| |
Collapse
|
38
|
Luo Y, Xie Y, Wu D, Wang B, Lu H, Wang Z, Quan Y, Han B. AL360181.1 promotes proliferation and invasion in colon cancer and is one of ten m6A-related lncRNAs that predict overall survival. PeerJ 2023; 11:e16123. [PMID: 37953780 PMCID: PMC10638913 DOI: 10.7717/peerj.16123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/27/2023] [Indexed: 11/14/2023] Open
Abstract
Background N6-methyladenosine (m6A) exerted a pivotal role in colon cancer. Nevertheless, the long non-coding RNAs (lncRNAs) associated with this process have yet to be elucidated. Methods The open-access data used for analysis was downloaded from The Cancer Genome Atlas (TCGA) database for analysis, employing the R software for computational evaluations. The RNA level of specific molecules was assessed using the quantitative real-time PCR. CCK8, colony formation and transwell assay were used to evaluate the proliferation, invasion and migration ability of colon cancer cells. Results Here, we identified the m6A regulators from TCGA data and subsequently pinpointed lncRNAs with a -Cor- > 0.3 and P < 0.05, categorizing them as m6A-associated lncRNAs. Moreover, we formulated a prognosis signature rooted in ten m6A-related lncRNAs, consisting of AL360181.1, PCAT6, SNHG26, AC016876.1, AC104667.2, AL114730.3, LINC02257, AC147067.1, AP006621.3 and AC009237.14. This signature exhibited notable predictive accuracy in gauging patient survival. Immune-related evaluations revealed varied immune cell infiltration patterns across different risk groups, with our findings suggesting superior immunotherapy response in low-risk patients. Biological enrichment analysis indicated that the high-risk patients had a higher activity of multiple carcinogenic pathways, including glycolysis. The previously unreported lncRNA, AL360181.1, displayed a connection to glycolytic activity and diminished survival rates, warranting further investigation. The result indicated that AL360181.1 was correlated with more aggressive clinical characteristics. Immune infiltration assessments found AL360181.1 to have a positive correlation with Tcm infiltration, but an inverse relationship with entities like Th2 cells, T cells, neutrophils and macrophages. Biological enrichment analysis indicated that the pathways of WNT/β-catenin, pancreas beta cells, hedgehog signaling and some metabolism pathways were upregulated in high AL360181.1 patients. In vitro experiments showed that AL360181.1 was upregulated in the colon cancer cells. Moreover, AL360181.1 significantly promotes the proliferation, invasion and migration of colon cancer cells. Conclusions Our results can provide direction for future studies on m6A-related lncRNA in colon cancer.
Collapse
Affiliation(s)
- Yi Luo
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yayun Xie
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dejun Wu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Bingyi Wang
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Helei Lu
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiqiang Wang
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingjun Quan
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Han
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Zheng Y, Zhou R, Cai J, Yang N, Wen Z, Zhang Z, Sun H, Huang G, Guan Y, Huang N, Shi M, Liao Y, Bin J, Liao W. Matrix Stiffness Triggers Lipid Metabolic Cross-talk between Tumor and Stromal Cells to Mediate Bevacizumab Resistance in Colorectal Cancer Liver Metastases. Cancer Res 2023; 83:3577-3592. [PMID: 37610655 PMCID: PMC10618741 DOI: 10.1158/0008-5472.can-23-0025] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/06/2023] [Accepted: 08/21/2023] [Indexed: 08/24/2023]
Abstract
Bevacizumab is an anti-VEGF monoclonal antibody that plays an important role in the combination treatment of advanced colorectal cancer. However, resistance remains a major hurdle limiting bevacizumab efficacy, highlighting the importance of identifying a mechanism of antiangiogenic therapy resistance. Here, we investigated biophysical properties of the extracellular matrix (ECM) related to metabolic processes and acquired resistance to bevacizumab. Evaluation of paired pre- and posttreatment samples of liver metastases from 20 colorectal cancer patients treated with combination bevacizumab therapy, including 10 responders and 10 nonresponders, indicated that ECM deposition in liver metastases and a highly activated fatty acid oxidation (FAO) pathway were elevated in nonresponders after antiangiogenic therapy compared with responders. In mouse models of liver metastatic colorectal cancer (mCRC), anti-VEGF increased ECM deposition and FAO in colorectal cancer cells, and treatment with the FAO inhibitor etomoxir enhanced the efficacy of antiangiogenic therapy. Hepatic stellate cells (HSC) were essential for matrix stiffness-mediated FAO in colon cancer cells. Matrix stiffness activated lipolysis in HSCs via the focal adhesion kinase (FAK)/yes-associated protein (YAP) pathway, and free fatty acids secreted by HSCs were absorbed as metabolic substrates and activated FAO in colon cancer cells. Suppressing HSC lipolysis using FAK and YAP inhibition enhanced the efficacy of anti-VEGF therapy. Together, these results indicate that bevacizumab-induced ECM remodeling triggers lipid metabolic cross-talk between colon cancer cells and HSCs. This metabolic mechanism of bevacizumab resistance mediated by the physical tumor microenvironment represents a potential therapeutic target for reversing drug resistance. SIGNIFICANCE Extracellular matrix stiffening drives bevacizumab resistance by stimulating hepatic stellate cells to provide fuel for mCRC cells in the liver, indicating a potential metabolism-based therapeutic strategy for overcoming resistance.
Collapse
Affiliation(s)
- Yannan Zheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Rui Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianan Cai
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Nanyan Yang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhaowei Wen
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhihua Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Huiying Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Genjie Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yijin Guan
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianping Bin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
40
|
Xie Y, Li P, Sun D, Qi Q, Ma S, Zhao Y, Zhang S, Wang T, Wang J, Li S, Gong T, Xu H, Xiong M, Li G, You C, Luo Z, Li J, Wang C, Du L. DNA Methylation-Based Testing in Peripheral Blood Mononuclear Cells Enables Accurate and Early Detection of Colorectal Cancer. Cancer Res 2023; 83:3636-3649. [PMID: 37602818 PMCID: PMC10618739 DOI: 10.1158/0008-5472.can-22-3402] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/15/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023]
Abstract
An effective blood-based method for the diagnosis of colorectal cancer has not yet been developed. Molecular alterations of immune cells occur early in tumorigenesis, providing the theoretical underpinning for early cancer diagnosis based on immune cell profiling. Therefore, we aimed to develop an effective detection method based on peripheral blood mononuclear cells (PBMC) to improve the diagnosis of colorectal cancer. Analysis of the genome-wide methylation landscape of PBMCs from patients with colorectal cancer and healthy controls by microarray, pyrosequencing, and targeted bisulfite sequencing revealed five DNA methylation markers for colorectal cancer diagnosis, especially early-stage colorectal cancer. A single-tube multiple methylation-specific quantitative PCR assay (multi-msqPCR) for simultaneous detection of five methylation markers was established, which allowed quantitative analysis of samples with as little as 0.1% PBMC DNA and had better discriminative performance than single-molecule detection. Then, a colorectal cancer diagnostic model (CDM) based on methylation markers and the multi-msqPCR method was constructed that achieved high accuracy for early-stage colorectal cancer (AUC = 0.91; sensitivity = 81.18%; specificity = 89.39%), which was improved compared with CEA (AUC = 0.79). The CDM also enabled a high degree of discrimination for advanced adenoma cases (AUC = 0.85; sensitivity = 63.04%). Follow-up data also demonstrated that the CDM could identify colorectal cancer potential up to 2 years before currently used diagnostic methods. In conclusion, the approach constructed in this study based on PBMC-derived DNA methylation markers and a multi-msqPCR method is a promising and easily implementable diagnostic method for early-stage colorectal cancer. SIGNIFICANCE Development of a diagnostic model for early colorectal cancer based on epigenetic analysis of PBMCs supports the utility of altered DNA methylation in immune cells for cancer diagnosis.
Collapse
Affiliation(s)
- Yan Xie
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Dong Sun
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Qiuchen Qi
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
- Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, P.R. China
| | - Suhong Ma
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Yinghui Zhao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Shujun Zhang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Tiantian Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Jing Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Shijun Li
- Department of Clinical Laboratory, The First Hospital of Dalian Medical University, Dalian, P.R. China
| | - Tingting Gong
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, P.R. China
| | - Huiting Xu
- Department of Clinical Laboratory Medicine, Affiliated Tumor Hospital of Nantong University, Jiangsu, P.R. China
- Medical School of Nantong University, Nantong, P.R. China
| | - Mengqiu Xiong
- Department of Clinical Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, P.R. China
| | - Guanghua Li
- Department of Clinical Laboratory, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, P.R. China
| | - Chongge You
- Laboratory Medicine Center, Lanzhou University Second Hospital, The Second Clinical Medical College of Lanzhou University, Lanzhou, P.R. China
| | - Zhaofan Luo
- Department of Clinical Laboratory, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, P.R. China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
- Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, P.R. China
- Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, P.R. China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, P.R. China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Innovation Technology in Laboratory Medicine, Jinan, P.R. China
| |
Collapse
|
41
|
Wang Z, Chen K, Zhang K, He K, Zhang D, Guo X, Huang T, Hu J, Zhou X, Nie S. Agrocybe cylindracea fucoglucogalactan induced lysosome-mediated apoptosis of colorectal cancer cell through H3K27ac-regulated cathepsin D. Carbohydr Polym 2023; 319:121208. [PMID: 37567726 DOI: 10.1016/j.carbpol.2023.121208] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 08/13/2023]
Abstract
Inducing lysosomal dysfunction is emerging as a promising means for cancer therapy. Agrocybe cylindracea fucoglucogalactan (ACP) is a bioactive ingredient with anti-tumor activity, while its mechanism remains obscure. Herein, we found that ACP visibly inhibited the proliferation of colorectal cancer cells, and the IC50 value on HCT-116 cells (HT29 cells) was 490 μg/mL (786.4 μg/mL) at 24 h. RNA-seq showed that ACP regulated mitochondria, lysosome and apoptosis-related pathways. Further experiments proved that ACP indeed promoted apoptosis and lysosomal dysfunction of HCT-116 cells. Moreover, ChIP-seq revealed that ACP increased histone-H3-lysine-27 acetylation (H3K27ac) on CTSD (cathepsin D) promoter in HCT-116 cells, thus facilitating the binding of transcription factor EB (TFEB), and resulted in ascension of CTSD expression. Additionally, ACP triggered mitochondrial-mediated apoptosis by decreasing mitochondrial membrane potential and increasing pro-apoptotic protein levels. Notably, Pepstatin A (CTSD inhibitor) availably alleviated ACP-induced apoptosis. Taken together, our results indicated that ACP induced lysosome-mitochondria mediated apoptosis via H3K27ac-regulated CTSD in HCT-116 cells. This study indicates that ACP has anti-cancer potential in the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Ziwei Wang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Kunying Chen
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Ke Zhang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Kaihong He
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Duoduo Zhang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Xiaohan Guo
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Tongwen Huang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Jielun Hu
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China
| | - Xingtao Zhou
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China.
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Nanchang University, Nanchang 330047, China.
| |
Collapse
|
42
|
Ma Y, Zhu H, Jiang X, Zhou Z, Zhou Y, Tian Y, Zhang H, Sun M, Tu L, Lu J, Niu Y, Liu H, Liu Y, Chen P. Biological Evaluation of 8-Methoxy-2,5-dimethyl-5H-indolo[2,3-b] Quinoline as a Potential Antitumor Agent via PI3K/AKT/mTOR Signaling. Int J Mol Sci 2023; 24:15142. [PMID: 37894822 PMCID: PMC10606936 DOI: 10.3390/ijms242015142] [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: 09/05/2023] [Revised: 09/28/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Chemotherapy is commonly used clinically to treat colorectal cancer, but it is usually prone to drug resistance, so novel drugs need to be developed continuously to treat colorectal cancer. Neocryptolepine derivatives have attracted a lot of attention because of their good cytotoxic activity; however, cytotoxicity studies on colorectal cancer cells are scarce. In this study, the cytotoxicity of 8-methoxy-2,5-dimethyl-5H-indolo[2,3-b] quinoline (MMNC) in colorectal cells was evaluated. The results showed that MMNC inhibits the proliferation of HCT116 and Caco-2 cells, blocks the cell cycle in the G2/M phase, decreases the cell mitochondrial membrane potential and induces apoptosis. In addition, the results of western blot experiments suggest that MMNC exerts cytotoxicity by inhibiting the expression of PI3K/AKT/mTOR signaling pathway-related proteins. Based on these results, MMNC is a promising lead compound for anticancer activity in the treatment of human colorectal cancer.
Collapse
Affiliation(s)
- Yunhao Ma
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Hongmei Zhu
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Xinrong Jiang
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Zhongkun Zhou
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Yong Zhou
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Yanan Tian
- Faculty of Applied Science, Macao Polytechnic University, Macao, China; (Y.T.); (H.L.)
| | - Hao Zhang
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Mengze Sun
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Lixue Tu
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Juan Lu
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Yuqing Niu
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Huanxiang Liu
- Faculty of Applied Science, Macao Polytechnic University, Macao, China; (Y.T.); (H.L.)
| | - Yingqian Liu
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| | - Peng Chen
- School of Pharmacy, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; (Y.M.); (H.Z.); (X.J.); (Z.Z.); (Y.Z.); (H.Z.); (M.S.); (L.T.); (J.L.); (Y.N.)
| |
Collapse
|
43
|
Mehryab F, Taghizadeh F, Goshtasbi N, Merati F, Rabbani S, Haeri A. Exosomes as cutting-edge therapeutics in various biomedical applications: An update on engineering, delivery, and preclinical studies. Biochimie 2023; 213:139-167. [PMID: 37207937 DOI: 10.1016/j.biochi.2023.05.010] [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: 12/24/2022] [Revised: 04/29/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Exosomes are cell-derived nanovesicles, circulating in different body fluids, and acting as an intercellular mechanism. They can be purified from culture media of different cell types and carry an enriched content of various protein and nucleic acid molecules originating from their parental cells. It was indicated that the exosomal cargo can mediate immune responses via many signaling pathways. Over recent years, the therapeutic effects of various exosome types were broadly investigated in many preclinical studies. Herein, we present an update on recent preclinical studies on exosomes as therapeutic and/or delivery agents for various applications. The exosome origin, structural modifications, natural or loaded active ingredients, size, and research outcomes were summarized for various diseases. Overall, the present article provides an overview of the latest exosome research interests and developments to clear the way for the clinical study design and application.
Collapse
Affiliation(s)
- Fatemeh Mehryab
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Taghizadeh
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nazanin Goshtasbi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Merati
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Rabbani
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Pham H, Dixon E. Integration of Next-Generation Sequencing in the Surgical Management of Colorectal Liver Metastasis. Ann Surg Oncol 2023; 30:6815-6823. [PMID: 37316745 DOI: 10.1245/s10434-023-13750-7] [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: 05/27/2022] [Accepted: 06/04/2023] [Indexed: 06/16/2023]
Abstract
Hepatic resection remains the treatment of choice for colorectal liver metastases. The advancement of surgical technique and use of perioperative systemic therapy has expanded the number and complexity of patients eligible for surgical resection. In recent years, investigation into gene mutations, such as RAS/RAF pathway, have led to targeted therapies that have significantly improved outcomes. Next-generation sequencing allows analysis of large number of genes that may have potential prognostic relevance in the clinical setting. This review summarizes the current applications of next-generation sequencing technology in metastatic colorectal cancer, focusing on its prognostic implications on patient management.
Collapse
Affiliation(s)
- Helen Pham
- Department of Surgery, Faculty of Medicine, University of Calgary, Foothills Medical Centre, Calgary, AB, Canada.
| | - Elijah Dixon
- Department of Surgery, Faculty of Medicine, University of Calgary, Foothills Medical Centre, Calgary, AB, Canada
| |
Collapse
|
45
|
Soyluoglu S, Ozdemir Gunay B. Contribution of Metabolic Parameters and Pericolic Fat Stranding on Preoperative 18F-FDG PET/CT in Predicting Post-operative Histopathology and Outcome in Colorectal Cancer. Nucl Med Mol Imaging 2023; 57:223-234. [PMID: 37720878 PMCID: PMC10504194 DOI: 10.1007/s13139-023-00808-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 09/19/2023] Open
Abstract
Purpose We aimed to investigate the additional value of preoperative PET/CT and reveal relationships between metabolic parameters, pericolic fat stranding finding, postoperative histopathology, and overall survival in colorectal cancer (CRC). Methods CRC patients who underwent preoperative PET/CT between January 2017-December 2021 were analyzed. Lymph nodes, organ metastases, and metabolic parameters were evaluated from PET/CT. The pericolic fat stranding was evaluated from CT component. Relationships between these factors and postoperative histopathological findings were statistically analyzed. Survival analyses were performed. Results Ninety-one patients (59 males, 32 females) were included in the study. All tumors showed high FDG uptake (mean SUVmax 19.5 ± 9.9). SUVmax of the tumor differed significantly at T3 and T4 stages (p = 0.041). A significant correlation was found between MTV, TLG values and the differentiation degree (p = 0.005, 0.003, respectively). PET/CT predicted the N stage with a high accuracy rate (80%). PET/CT found additional metastases that changed treatment decisions in one-third of patients. A relationship was found between tumor length, surgical margin, lymphovascular invasion and pericolic fat stranding. In multivariate analysis, differentiation degree (HR = 26.1, 95%CI 1.672-408.467), MTV (HR = 0.3, 95%CI 0.071-0.841), TLG (HR = 3.5, 95%CI 1.065-11.193), and lymphovascular invasion (HR = 0.2, 95%CI 0.026-0.853, p = 0.033) were independent factors affecting overall survival. Conclusion Preoperative PET/CT contributes to CRC management by detecting additional metastases as well as predicting prognosis and postoperative findings such as T stage, N stage and tumor differentiation. The SUVmax may differentiate between T3 and T4 tumor. Reporting of pericolic fat stranding may contribute to the estimation of lymphatic invasion and positive surgical margin.
Collapse
Affiliation(s)
- Selin Soyluoglu
- Faculty of Medicine, Department of Nuclear Medicine, Trakya University, Balkan Yerleskesi Trakya Üniversitesi Tıp Fakultesi Nukleer Tıp AD, Edirne, 22030 Turkey
| | - Busra Ozdemir Gunay
- Faculty of Medicine, Department of Nuclear Medicine, Trakya University, Balkan Yerleskesi Trakya Üniversitesi Tıp Fakultesi Nukleer Tıp AD, Edirne, 22030 Turkey
| |
Collapse
|
46
|
Ghasemi-Kebria F, Jafari-Delouie N, Semnani S, Fazel A, Etemadi A, Norouzi A, Khandoozi R, Besharat S, Shokouhifar N, Mirkarimi H, Sedaghat S, Mansoury M, Mehrjerdian M, Weiderpass E, Roshandel G, Bray F, Malekzadeh R. Colorectal cancer incidence trends in Golestan, Iran: An age-period-cohort analysis 2004-2018. Cancer Epidemiol 2023; 86:102415. [PMID: 37442047 DOI: 10.1016/j.canep.2023.102415] [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: 05/11/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND We investigated the effects of factors including age, birth year (cohort) and diagnosis year (period) on colorectal cancer (CRC) incidence trends in Golestan, Northeast of Iran, 2004-2018. METHODS We obtained data on incidence cases of CRC from the Golestan Population-based Cancer Registry by sex and area of residence (urban/rural). Age-standardized incidence rates (ASRs) were calculated using the World standard population and presented per 100,000 person-years. We calculated the estimated annual percentage change (EAPC) with 95 % confidence intervals (95 % CI) fitted age-period-cohort (APC) models to assess non-linear period and cohort effects as incidence rate ratios (IRRs). RESULTS Overall, 2839 new cases of CRC (ASR = 13.7) were registered in the GPCR over 2004-2018. Our findings suggested significantly increasing trends in CRC incidence rates from 2004 to 2018 (EAPC = 3.7; 95%CI: 0.4, 7.1), with the greatest changes occurring in rural women (EAPC= 4.7; 95%CI: 0.4, 9.2). We observed a strong cohort effect with a consistent increase in the IRR across successive birth cohorts, starting with the oldest birth cohort (1924) (IRR= 0.1 versus the reference birth cohort of 1955) through to the most recent cohort born in 1983 (IRR= 1.9). The largest cohort effects were found among rural females (IRR = 0.0, and IRR = 2.5 for the oldest and the youngest birth cohorts vs. the reference birth cohort, respectively). CONCLUSION The increasing trends in CRC rates in Golestan are largely driven by generational changes in exposure to underlying risk factors. Further investigations are warranted to deliver effective prevention strategies for the control of CRC in Golestan.
Collapse
Affiliation(s)
- Fatemeh Ghasemi-Kebria
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Nastaran Jafari-Delouie
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shahryar Semnani
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abdolreza Fazel
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Arash Etemadi
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Alireza Norouzi
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Reza Khandoozi
- Cancer Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sima Besharat
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Nesa Shokouhifar
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Honyehsadat Mirkarimi
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Mohsen Mansoury
- Office of the Statistics and Information Technology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahshid Mehrjerdian
- Department of Pathology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Elisabete Weiderpass
- Office of the Director, International Agency for Research on Cancer (IARC), Lyon, France
| | - Gholamreza Roshandel
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Freddie Bray
- Cancer Surveillance Branch, International Agency for Research on Cancer (IARC), World Health Organization (WHO), Lyon, France.
| | - Reza Malekzadeh
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
47
|
Ren G, Li H, Hong D, Hu F, Jin R, Wu S, Sun W, Jin H, Zhao L, Zhang X, Liu D, Huang C, Huang H. LINC00955 suppresses colorectal cancer growth by acting as a molecular scaffold of TRIM25 and Sp1 to Inhibit DNMT3B-mediated methylation of the PHIP promoter. BMC Cancer 2023; 23:898. [PMID: 37742010 PMCID: PMC10518100 DOI: 10.1186/s12885-023-11403-2] [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: 04/02/2023] [Accepted: 09/14/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND Long non-coding RNAs play an important role in the development of colorectal cancer (CRC), while many CRC-related lncRNAs have not yet been identified. METHODS The relationship between the expression of LINC00955 (Long Intergenic Non-protein Coding RNA 955) and the prognosis of colorectal cancer patients was analyzed using the sequencing results of the TCGA database. LINC00955 expression levels were measured using qRT-PCR. The anti-proliferative activity of LINC00955 was evaluated using CRC cell lines in vitro and xenograft models in nude mice in vivo. The interaction of TRIM25-Sp1-DNMT3B-PHIP-CDK2 was analyzed by western blotting, protein degradation experiment, luciferase, RNA-IP, RNA pull-down assays and immunohistochemically analysis. The biological roles of LINC00955, tripartite motif containing 25 (TRIM25), Sp1 transcription factor (Sp1), DNA methyltransferase 3 beta (DNMT3B), pleckstrin homology domain interacting protein (PHIP), cyclin dependent kinase 2 (CDK2) in colorectal cancer cells were analyzed using ATP assays, Soft agar experiments and EdU assays. RESULTS The present study showed that LINC00955 is downregulated in CRC tissues, and such downregulation is associated with poor prognosis of CRC patients. We found that LINC00955 can inhibit CRC cell growth both in vitro and in vivo. Evaluation of its mechanism of action showed that LINC00955 acts as a scaffold molecule that directly promotes the binding of TRIM25 to Sp1, and promotes ubiquitination and degradation of Sp1, thereby attenuating transcription and expression of DNMT3B. DNMT3B inhibition results in hypomethylation of the PHIP promoter, in turn increasing PHIP transcription and promoting ubiquitination and degradation of CDK2, ultimately leading to G0/G1 growth arrest and inhibition of CRC cell growth. CONCLUSIONS These findings indicate that downregulation of LINC00955 in CRC cells promotes tumor growth through the TRIM25/Sp1/DNMT3B/PHIP/CDK2 regulatory axis, suggesting that LINC00955 may be a potential target for the therapy of CRC.
Collapse
Affiliation(s)
- Ganglin Ren
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Jiaxing Center for Disease Control and Prevention, Jiaxing, 314050, Zhejiang, China
| | - Hongyan Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Dan Hong
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fangyu Hu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Rongjia Jin
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Shuang Wu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Wenhao Sun
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Honglei Jin
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Lingling Zhao
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiaodong Zhang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Dongxiang Liu
- Center for Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Chuanshu Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
48
|
Macis D, Briata IM, D’Ecclesiis O, Johansson H, Aristarco V, Buttiron Webber T, Oppezzi M, Gandini S, Bonanni B, DeCensi A. Inflammatory and Metabolic Biomarker Assessment in a Randomized Presurgical Trial of Curcumin and Anthocyanin Supplements in Patients with Colorectal Adenomas. Nutrients 2023; 15:3894. [PMID: 37764678 PMCID: PMC10537228 DOI: 10.3390/nu15183894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Colorectal cancer prevention is crucial for public health, given its high mortality rates, particularly in young adults. The early detection and treatment of precancerous lesions is key to preventing carcinogenesis progression. Natural compounds like curcumin and anthocyanins show promise in impeding adenomatous polyp progression in preclinical models. We conducted a randomized, double-blind, placebo-controlled, phase II presurgical trial in 35 patients with adenomatous polyps to explore the biological effects of curcumin and anthocyanins on circulating biomarkers of inflammation and metabolism. No significant difference in biomarker changes by treatment arm was observed. However, the network analysis before treatment revealed inverse correlations between adiponectin and BMI and glycemia, as well as direct links between inflammatory biomarkers and leptin and BMI. In addition, a considerable inverse relationship between adiponectin and grade of dysplasia was detected after treatment (corr = -0.45). Finally, a significant increase in IL-6 at the end of treatment in subjects with high-grade dysplasia was also observed (p = 0.02). The combined treatment of anthocyanins and curcumin did not result in the direct modulation of circulating biomarkers of inflammation and metabolism, but revealed a complex modulation of inflammatory and metabolic biomarkers of colon carcinogenesis.
Collapse
Affiliation(s)
- Debora Macis
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.M.); (H.J.); (V.A.); (B.B.)
| | - Irene Maria Briata
- Division of Medical Oncology, E.O. Galliera Hospital, 16128 Genoa, Italy; (I.M.B.); (T.B.W.)
| | - Oriana D’Ecclesiis
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (O.D.); (S.G.)
| | - Harriet Johansson
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.M.); (H.J.); (V.A.); (B.B.)
| | - Valentina Aristarco
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.M.); (H.J.); (V.A.); (B.B.)
| | - Tania Buttiron Webber
- Division of Medical Oncology, E.O. Galliera Hospital, 16128 Genoa, Italy; (I.M.B.); (T.B.W.)
| | - Massimo Oppezzi
- Division of Gastroenterology, E.O. Galliera Hospital, 16128 Genoa, Italy;
| | - Sara Gandini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (O.D.); (S.G.)
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.M.); (H.J.); (V.A.); (B.B.)
| | - Andrea DeCensi
- Division of Medical Oncology, E.O. Galliera Hospital, 16128 Genoa, Italy; (I.M.B.); (T.B.W.)
| |
Collapse
|
49
|
Di Z, Xu G, Ding Z, Li C, Song J, Huang G, Zheng J, Zhang X, Xiong B. Identification and validation of a novel prognosis model based on m5C-related long non-coding RNAs in colorectal cancer. Cancer Cell Int 2023; 23:196. [PMID: 37670275 PMCID: PMC10481501 DOI: 10.1186/s12935-023-03025-2] [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: 02/23/2023] [Accepted: 08/08/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND The prognosis of tumor patients can be assessed by measuring the levels of lncRNAs (long non-coding RNAs), which play a role in controlling the methylation of the RNA. Prognosis in individuals with colorectal adenocarcinoma (CRC) is strongly linked to lncRNA expression, making it imperative to find lncRNAs that are associated with RNA methylation with strong prognostic value. METHODS In this study, by analyzing TCGA dataset, we were able to develop a risk model for lncRNAs that are associated with m5C with prognostic significance by employing LASSO regression and univariate Cox proportional analysis. There were a number of methods employed to ensure the model was accurate, including multivariate and univariate Cox regression analysis, Kaplan analysis, and receiver operating characteristic curve analysis. The principal component analysis, GSEA and GSVA analysis were used for risk model analysis. The CIBERSORT instrument and the TIMER database were used to evaluate the link between the immune cells that infiltrate tumors and the risk model. In vitro experiments were also performed to validate the predicted m5C-related significant lncRNAs. RESULTS The m5c regulators were differentially expressed in colorectal cancer and normal tissue. Based on the screening criteria and LASSO regression, 11 m5c-related lncRNAs were identified for developing the prognostic risk model. Multivariate and univariate Cox regression analysis showed the risk score is a crucial prognostic factor in CRC patients. The 1-year, 3-year, and 5-year AUC curves showed the risk score was higher than those identified for other clinicopathological characteristics. A nomogram using the risk score as a quantitative tool was developed for predicting patients' outcomes in clinical settings. In addition, the risk profile of m5C-associated lncRNAs can discriminate between tumor immune cells' characteristics in CRC. Mutation patterns and chemotherapy were analyzed between high- and low- risk groups of CRC patients. Moreover, TNFRSF10A-AS1 was chosen for the in vitro verification of the m5C-connected lncRNA to demonstrate impressive effects on the proliferation, migration and invasion of CRC cells. CONCLUSION A risk model including the prognostic value of 11 m5C-associated lncRNAs proves to be a useful prognostic tool for CRC and improves the care of patients suffering from CRC based on these findings.
Collapse
Affiliation(s)
- Ziyang Di
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
| | - Gaoran Xu
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
| | - Zheyu Ding
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
| | - Chengxin Li
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
| | - Jialin Song
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
| | - Guoquan Huang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
| | - Jinsen Zheng
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
| | - Xinyao Zhang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
| | - Bin Xiong
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.
| |
Collapse
|
50
|
Wang X, Li X, Zhang L, An L, Guo L, Huang L, Gao W. Recent progress in plant-derived polysaccharides with prebiotic potential for intestinal health by targeting gut microbiota: a review. Crit Rev Food Sci Nutr 2023:1-30. [PMID: 37651130 DOI: 10.1080/10408398.2023.2248631] [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: 09/01/2023]
Abstract
Natural products of plant origin are of high interest and widely used, especially in the food industry, due to their low toxicity and wide range of bioactive properties. Compared to other plant components, the safety of polysaccharides has been generally recognized. As dietary fibers, plant-derived polysaccharides are mostly degraded in the intestine by polysaccharide-degrading enzymes secreted by gut microbiota, and have potential prebiotic activity in both non-disease and disease states, which should not be overlooked, especially in terms of their involvement in the treatment of intestinal diseases and the promotion of intestinal health. This review elucidates the regulatory effects of plant-derived polysaccharides on gut microbiota and summarizes the mechanisms involved in targeting gut microbiota for the treatment of intestinal diseases. Further, the structure-activity relationships between different structural types of plant-derived polysaccharides and the occurrence of their prebiotic activity are further explored. Finally, the practical applications of plant-derived polysaccharides in food production and food packaging are summarized and discussed, providing important references for expanding the application of plant-derived polysaccharides in the food industry or developing functional dietary supplements.
Collapse
Affiliation(s)
- Xiaozhen Wang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Xia Li
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Luyao Zhang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Lingzhuo An
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, China
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|