1
|
Chang H, Zheng N, Zhu X. Comprehensive analysis identifies a lactylation-related signature for predicting prognosis and guiding therapies in colon adenocarcinoma. Gene 2024; 939:149191. [PMID: 39724993 DOI: 10.1016/j.gene.2024.149191] [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: 08/15/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
PURPOSE This study aimed to identify a lactylation-related gene signature for predicting prognosis and guiding therapies in colon adenocarcinoma (COAD). We seek to address the challenges in COAD prognostication due to tumor heterogeneity and variable treatment responses. METHODS The study employed integrative bioinformatics analyses on multi-omics data from public databases, including gene expression profiles, clinical data, and lactylation-related genes (LRGs). The least absolute shrinkage and selection operator (LASSO) regression analysis and Cox risk model were applied to develop a prognostic signature. The predictive capabilities of the signature were assessed in four independent COAD cohorts (GSE39582, GSE71187, GSE75500, and GSE17536). Functional enrichment, immune infiltrations, and scRNA-seq analysis were performed to investigate biological processes and the tumor microenvironment (TME). Additionally, functional assays were performed to assess the impact of gene knockdown on COAD cell behavior. RESULTS A 3-gene signature (SUSD5, FABP4, CALB2) was identified, demonstrating robust predictive performance for clinical outcomes in COAD patients across multiple cohorts. The signature revealed involvement in critical cancer-related biological processes and showed potential in guiding therapeutic decisions. The bulk RNA-seq and scRNA-seq analysis suggested that LRGs modulates the TME, particularly immune cell populations like mast cells. Knockdown of CALB2 significantly suppressed COAD cell proliferation, invasion, and migration. CONCLUSION This comprehensive analysis identified a lactylation-related signature with significant prognostic and therapeutic implications for COAD. The findings highlight the importance of lactylation in COAD biology and offer novel insights for developing personalized treatment strategies, potentially improving patient outcomes in this prevalent malignancy.
Collapse
Affiliation(s)
- Huan Chang
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Hunan Changsha 410013, China
| | - Ning Zheng
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Hunan Changsha 410013, China
| | - Xiaocheng Zhu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Hunan Changsha 410008, China.
| |
Collapse
|
2
|
Deng RZ, Zheng X, Lu ZL, Yuan M, Meng QC, Wu T, Tian Y. Effect of colorectal cancer stem cells on the development and metastasis of colorectal cancer. World J Gastrointest Oncol 2024; 16:4354-4368. [PMID: 39554751 PMCID: PMC11551631 DOI: 10.4251/wjgo.v16.i11.4354] [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: 07/23/2024] [Revised: 08/24/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
The relevant mechanism of tumor-associated macrophages (TAMs) in the treatment of colorectal cancer patients with immune checkpoint inhibitors (ICIs) is discussed, and the application prospects of TAMs in reversing the treatment tolerance of ICIs are discussed to provide a reference for related studies. As a class of drugs widely used in clinical tumor immunotherapy, ICIs can act on regulatory molecules on cells that play an inhibitory role - immune checkpoints - and kill tumors in the form of an immune response by activating a variety of immune cells in the immune system. The sensitivity of patients with different types of colorectal cancer to ICI treatment varies greatly. The phenotype and function of TAMs in the colorectal cancer microenvironment are closely related to the efficacy of ICIs. ICIs can regulate the phenotypic function of TAMs, and TAMs can also affect the tolerance of colorectal cancer to ICI therapy. TAMs play an important role in ICI resistance, and making full use of this target as a therapeutic strategy is expected to improve the immunotherapy efficacy and prognosis of patients with colorectal cancer.
Collapse
Affiliation(s)
- Run-Zhi Deng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, Fujian Province, China
| | - Xin Zheng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, Fujian Province, China
| | - Zhong-Lei Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, Fujian Province, China
| | - Ming Yuan
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Qi-Chang Meng
- Department of General Surgery, Peking University First Hospital, Beijing 100034, China
| | - Tao Wu
- Department of General Surgery, West China Hospital of Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Yu Tian
- Department of Thoracic Surgery, Yancheng No. 1 People’s Hospital, Affiliated Hospital of Nanjing University Medical School, The First People’s Hospital of Yancheng, Yancheng 224000, Jiangsu Province, China
| |
Collapse
|
3
|
Li GS, Tang YX, Zhang W, Li JD, Huang HQ, Liu J, Fu ZW, He RQ, Kong JL, Zhou HF, Chen G. MMP12 is a Potential Predictive and Prognostic Biomarker of Various Cancers Including Lung Adenocarcinoma. Cancer Control 2024; 31:10732748241235468. [PMID: 38410859 PMCID: PMC10898301 DOI: 10.1177/10732748241235468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/01/2023] [Accepted: 01/09/2024] [Indexed: 02/28/2024] Open
Abstract
OBJECTIVE This study sought to explore the clinical value of matrix metalloproteinases 12 (MMP12) in multiple cancers, including lung adenocarcinoma (LUAD). METHODS Using >10,000 samples, this retrospective study demonstrated the first pan-cancer analysis of MMP12. The expression of MMP12 between cancer groups and their control groups was analyzed using Wilcoxon rank-sum tests. The clinical significance of MMP12 expression in multiple cancers was assessed using receiver operating characteristic curves, Kaplan-Meier curves, and univariate Cox analysis. A further LUAD-related analysis based on 4565 multi-center and in-house samples was performed to verify the findings regarding MMP12 in pan-cancer analysis partly. RESULTS MMP12 mRNA is highly expressed in 13 cancers compared to their controls, and the MMP12 protein level is elevated in some of these cancers (e.g., colon adenocarcinoma) (P < .05). MMP12 expression makes it feasible to distinguish 21 cancer tissues from normal tissues (AUC = 0.86). A high MMP12 expression is a prognosis risk factor in eight cancers, such as adrenocortical carcinoma (hazard ratio >1, P < .05). The elevated MMP12 expression is also a prognosis protective factor in breast-invasive carcinoma and colon adenocarcinoma (hazard ratio <1, P < .05). Some pan-cancer findings regarding MMP12 are verified in LUAD-MMP12 expression is upregulated in LUAD at both the mRNA and protein levels (P < .05), has the potential to distinguish LUAD with considerable accuracy (AUC = .91), and plays a risk prognosis factor for patients with the disease (P < .05). CONCLUSIONS MMP12 is highly expressed in most cancers and may serve as a novel biomarker for the prediction and prognosis of numerous cancers.
Collapse
Affiliation(s)
- Guo-Sheng Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Yu-Xing Tang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Wei Zhang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Jian-Di Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - He-Qing Huang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Jun Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Zong-Wang Fu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Jin-Liang Kong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Hua-Fu Zhou
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, P. R. China
| |
Collapse
|
4
|
Zhang W, Li GS, Gan XY, Huang ZG, He RQ, Huang H, Li DM, Tang YL, Tang D, Zou W, Liu J, Dang YW, Chen G, Zhou HF, Kong JL, Lu HP. MMP12 serves as an immune cell-related marker of disease status and prognosis in lung squamous cell carcinoma. PeerJ 2023; 11:e15598. [PMID: 37601247 PMCID: PMC10439720 DOI: 10.7717/peerj.15598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Background Worldwide, lung squamous cell carcinoma (LUSC) has wreaked havoc on humanity. Matrix metallopeptidase 12 (MMP12) plays an essential role in a variety of cancers. This study aimed to reveal the expression, clinical significance, and potential molecular mechanisms of MMP12 in LUSC. Methods There were 2,738 messenger RNA (mRNA) samples from several multicenter databases used to detect MMP12 expression in LUSC, and 125 tissue samples were validated by immunohistochemistry (IHC) experiments. Receiver operator characteristic (ROC) curves, Kaplan-Meier curves, and univariate and multivariate Cox regression analyses were used to assess the clinical value of MMP12 in LUSC. The potential molecular mechanisms of MMP12 were explored by gene enrichment analysis and immune correlation analysis. Furthermore, single-cell sequencing was used to determine the distribution of MMP12 in multiple tumor microenvironment cells. Results MMP12 was significantly overexpressed at the mRNA level (p < 0.05, SMD = 3.13, 95% CI [2.51-3.75]), which was verified at the protein level (p < 0.001) by internal IHC experiments. MMP12 expression could be used to differentiate LUSC samples from normal samples, and overexpression of MMP12 itself implied a worse clinical prognosis and higher levels of immune cell infiltration in LUSC patients. MMP12 was involved in cancer development and progression through two immune-related signaling pathways. The high expression of MMP12 in LUSC might act as an antigen-presenting cell-associated tumor neoantigen and activate the body's immune response. Conclusions MMP12 expression is upregulated in LUSC and high expression of MMP12 serves as a risk factor for LUSC patients. MMP12 may be involved in cancer development by participating in immune-related signaling pathways and elevating the level of immune cell infiltration.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Guo-Sheng Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiang-Yu Gan
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Guang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Hong Huang
- Department of Respiratory and Critical Care, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Dong-Ming Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yu-Lu Tang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Deng Tang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Wen Zou
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jun Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yi-Wu Dang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Hua-Fu Zhou
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jin-Liang Kong
- Department of Respiratory and Critical Care, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Hui-ping Lu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
5
|
Rodger EJ, Gimenez G, Ajithkumar P, Stockwell PA, Almomani S, Bowden SA, Leichter AL, Ahn A, Pattison S, McCall JL, Schmeier S, Frizelle FA, Eccles MR, Purcell RV, Chatterjee A. An epigenetic signature of advanced colorectal cancer metastasis. iScience 2023; 26:106986. [PMID: 37378317 PMCID: PMC10291510 DOI: 10.1016/j.isci.2023.106986] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/12/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Colorectal cancer (CRC) is a leading cause of morbidity and mortality worldwide. The majority of CRC deaths are caused by tumor metastasis, even following treatment. There is strong evidence for epigenetic changes, such as DNA methylation, accompanying CRC metastasis and poorer patient survival. Earlier detection and a better understanding of molecular drivers for CRC metastasis are of critical clinical importance. Here, we identify a signature of advanced CRC metastasis by performing whole genome-scale DNA methylation and full transcriptome analyses of paired primary cancers and liver metastases from CRC patients. We observed striking methylation differences between primary and metastatic pairs. A subset of loci showed coordinated methylation-expression changes, suggesting these are potentially epigenetic drivers that control the expression of critical genes in the metastatic cascade. The identification of CRC epigenomic markers of metastasis has the potential to enable better outcome prediction and lead to the discovery of new therapeutic targets.
Collapse
Affiliation(s)
- Euan J. Rodger
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Gregory Gimenez
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | | | - Peter A. Stockwell
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Suzan Almomani
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Sarah A. Bowden
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Anna L. Leichter
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Antonio Ahn
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Sharon Pattison
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - John L. McCall
- Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | | | - Frank A. Frizelle
- Department of Surgery, University of Otago, Christchurch, New Zealand
| | - Michael R. Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Rachel V. Purcell
- Department of Surgery, University of Otago, Christchurch, New Zealand
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
- Honorary Professor, School of Health Sciences and Technology, UPES University, India
| |
Collapse
|
6
|
Ren S, Cao W, Ma J, Li H, Xia Y, Zhao J. Correlation evaluation between cancer microenvironment related genes and prognosis based on intelligent medical internet of things. Front Genet 2023; 14:1132242. [PMID: 36845384 PMCID: PMC9947234 DOI: 10.3389/fgene.2023.1132242] [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: 12/27/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
The study of tumor microenvironment plays an important role in the treatment of cancer patients. In this paper, intelligent medical Internet of Things technology was used to analyze cancer tumor microenvironment-related genes. Through experiments designed and analyzed cancer-related genes, this study concluded that in cervical cancer, patients with high expression of P16 gene had a shorter life cycle and a survival rate of 35%. In addition, through investigation and interview, it was found that patients with positive expression of P16 and Twist genes had a higher recurrence rate than patients with negative expression of both genes; high expression of FDFT1, AKR1C1, and ALOX12 in colon cancer is associated with short survival; high expressions of HMGCR and CARS1 is associated with longer survival; overexpression of NDUFA12, FD6, VEZT, GDF3, PDE5A, GALNTL6, OPMR1, and AOAH in thyroid cancer is associated with shortened survival; high expressions of NR2C1, FN1, IPCEF1, and ELMO1 is associated with prolonged survival. Among the genes associated with the prognosis of liver cancer, the genes associated with shorter survival period are AGO2, DCPS, IFIT5, LARP1, NCBP2, NUDT10, and NUDT16; the genes associated with longevity are EIF4E3, EIF4G3, METTL1, NCBP1, NSUN2, NUDT11, NUDT4, and WDR4. Depending on the prognostic role of genes in different cancers, they can influence patients to achieve the effect of reducing patients' symptoms. In the process of disease analysis of cancer patients, this paper uses bioinformation technology and Internet of things technology to promote the development of medical intelligence.
Collapse
Affiliation(s)
- Shoulei Ren
- Oncology Department, Yangguangronghe Hospital, Weifang, Shandong, China
| | - Wenli Cao
- Oncology Department, Yangguangronghe Hospital, Weifang, Shandong, China
| | - Jianzeng Ma
- Oncology Department, Yangguangronghe Hospital, Weifang, Shandong, China
| | - Hongchun Li
- Nerosurgery Department, Yangguangronghe Hospital, Weifang, Shandong, China
| | - Yutao Xia
- Oncology Department, Yangguangronghe Hospital, Weifang, Shandong, China
| | - Jianwen Zhao
- Oncology Department, Yangguangronghe Hospital, Weifang, Shandong, China,*Correspondence: Jianwen Zhao,
| |
Collapse
|
7
|
Wang H, Tang Y, Wang M, Zhao J, Ding C, Yang X, Han P, Liu P. Low expression of MEOX2 is associated with poor survival in patients with breast cancer. Biomark Med 2022; 16:1161-1170. [PMID: 36625258 DOI: 10.2217/bmm-2022-0468] [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: 01/11/2023] Open
Abstract
Aim: To investigate associations of MEOX2 expression with clinicopathological features and survival of breast cancer patients. Materials & methods: We used a breast cancer tissue microarray for immunohistochemistry. Associations between MEOX2 expression and clinicopathological features were analyzed using the χ-square test. Survival analysis was determined using a Kaplan-Meier curve. Multivariate Cox regression was used to determine associations of MEOX2 expression with overall survival. Results: We found that 74.1% of patients (100/135) had expression of MEOX2 at varying levels. MEOX2 was associated with histological grade and negatively correlated with Ki67 expression. Lower MEOX2 expression was significantly associated with decreased overall survival (p = 0.0011). Conclusion: MEOX2 expression could be a novel diagnostic and prognostic biomarker of breast cancer.
Collapse
Affiliation(s)
- Huxia Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.,Department of Mammary, Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Yanan Tang
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Meixia Wang
- Department of Health Examination, Shenmu Hospital, Yulin, 719300, China
| | - Jing Zhao
- Department of Mammary, Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Caixia Ding
- Department of Pathology, Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Xiaomin Yang
- Department of Mammary, Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Pihua Han
- Department of Mammary, Shaanxi Provincial Cancer Hospital, Xi'an, 710061, China
| | - Peijun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
8
|
Wang L, Chen X, Zhang H, Hong L, Wang J, Shao L, Chen G, Wu J. Comprehensive analysis of transient receptor potential channels-related signature for prognosis, tumor immune microenvironment, and treatment response of colorectal cancer. Front Immunol 2022; 13:1014834. [PMID: 36389750 PMCID: PMC9642045 DOI: 10.3389/fimmu.2022.1014834] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/23/2022] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Transient receptor potential channels (TRPC) play critical regulatory functions in cancer occurrence and progression. However, knowledge on its role in colorectal cancer (CRC) is limited. In addition, neoadjuvant treatment and immune checkpoint inhibitors (ICIs) have increasing roles in CRC management, but not all patients benefit from them. In this study, a TRPC related signature (TRPCRS) was constructed for prognosis, tumor immune microenvironment (TIME), and treatment response of CRC. METHODS Data on CRC gene expression and clinical features were retrospectively collected from TCGA and GEO databases. Twenty-eight TRPC regulators (TRPCR) were retrieved using gene set enrichment analysis. Different TRPCR expression patterns were identified using non-negative matrix factorization for consensus clustering, and a TRPCRS was established using LASSO. The potential value of TRPCRS was assessed using functional enrichment analysis, tumor immune analysis, tumor somatic mutation analysis, and response to preoperative chemoradiotherapy or ICIs. Moreover, an external validation was conducted using rectal cancer samples that received preoperative chemoradiotherapy at Fujian Cancer Hospital (FJCH) via qRT-PCR. RESULTS Among 834 CRC samples in the TCGA and meta-GEO cohorts, two TRPCR expression patterns were identified, which were associated with various immune infiltrations. In addition, 266 intersected genes from 5564 differentially expressed genes (DEGs) between two TRPC subtypes, 4605 DEGs between tumor tissue and adjacent non-tumor tissue (all FDR< 0.05, adjusted P< 0.001), and 1329 prognostic related genes (P< 0.05) were identified to establish the TRPCRS, which was confirmed in the TCGA cohort, two cohorts from GEO, and one qRT-PCR cohort from FJCH. According to the current signature, the high-TRPC score group had higher expressions of PD-1, PD-L1, and CTLA4, lower TIDE score, and improved response to anti-PD-1 treatment with better predictive ability. Compared to the high-TRPC score group, the low-TRPC score group comprised an immunosuppressive phenotype with increased infiltration of neutrophils and activated MAPK signaling pathway, but was more sensitive to preoperative chemoradiotherapy and associated with improved prognosis. CONCLUSIONS The current TRPCRS predicted the prognosis of CRC, evaluated the TIME in CRC, and anticipated the response to immune therapy and neoadjuvant treatment.
Collapse
Affiliation(s)
- Lei Wang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xingte Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hejun Zhang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Liang Hong
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jianchao Wang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Lingdong Shao
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Gang Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Junxin Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
9
|
The Underlying Roles of Exosome-Associated PIGR in Fatty Acid Metabolism and Immune Signaling in Colorectal Cancer. JOURNAL OF ONCOLOGY 2022; 2022:4675683. [PMID: 36157233 PMCID: PMC9499750 DOI: 10.1155/2022/4675683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022]
Abstract
The polymeric immunoglobulin receptor (PIGR), an exosome-associated glycoprotein, plays an important role in the occurrence and development of different tumors. This study aimed to investigate whether PIGR is essential for colorectal cancer (CRC). Comprehensive bioinformatics analysis and immunohistochemistry (IHC) revealed that expression of PIGR was significantly decreased in CRC patients. Upregulated PIGR displayed favorable prognostic values in CRC patients. Several algorithms, such as TISIDB and TIMER, were used to evaluate the roles of PIGR expression in the regulation of immune response in CRC. Moreover, GSEA enrichment analysis indicated the underlying role of PIGR in the regulation of fatty acid metabolism in CRC. Taken together, our findings might provide a new potential prognostic and immune-associated biomarker for CRC and supply a new destination for PIGR-related immunotherapy in clinical treatment.
Collapse
|
10
|
Hu J, He Y, Liao K, Yang Q, Xu Y, Cao G, Wang X. Identification of inflammatory factor-related genes associated with the prognostic and immune cell infiltration in colorectal cancer patients. Genes Dis 2022. [PMID: 37492736 PMCID: PMC10363590 DOI: 10.1016/j.gendis.2022.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This study aims to identify the inflammatory factor-related genes which help to predict the prognosis of patients with colorectal cancer. GSEA (Gene Set Enrichment Analysis) was used to acquire inflammation-related genes and the corresponding expression information was collected from TCGA database to determine the DEGs (differentially-expressed genes) in CRC patients. We conducted enrichment analysis and PPI (protein-protein interaction) of these DEGs. Besides, key genes that are both differentially-expressed and prognosis-related were screened out, which were used to establish the prognostic model. We obtained 79 DEGs and 19 prognostic genes, 10 prognostic-related differential genes were eventually screened. These genes were used to construct the prognostic model. We also identified that the immune infiltration score of macrophages between different risk groups was significantly different and similar distinction was witnessed in immune function score of APC (antigen-presenting cell) co-stimulation and type I IFN (interferon) response.
Collapse
|
11
|
TNFAIP6 Promotes Gastric Carcinoma Cell Invasion via Upregulating PTX3 and Activating the Wnt/ β-Catenin Signaling Pathway. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:5697034. [PMID: 35854776 PMCID: PMC9279067 DOI: 10.1155/2022/5697034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 11/25/2022]
Abstract
Tumor metastasis is a fundamental cause of the poor prognosis of gastric carcinoma (GC). In order to study the problems affecting metastasis and recurrence of gastric cancer, the paper expose that TNF alpha induced protein 6 (TNFAIP6) is aberrantly overexpressed in GC, and patients with high-TNFAIP6 levels exhibited inferior overall survival. Mechanistically, overexpression of TNFAIP6 raised β-catenin ectopic nuclear distribution and activated the Wnt/β-catenin signal pathway. The experimental results show that TNFAIP6 facilitates the aggressive potential of GC cells through modulating PTX3 expression.
Collapse
|
12
|
Identification of FABP7 as a Potential Biomarker for Predicting Prognosis and Antiangiogenic Drug Efficacy of Glioma. DISEASE MARKERS 2022; 2022:2091791. [PMID: 35783014 PMCID: PMC9249527 DOI: 10.1155/2022/2091791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022]
Abstract
Objective Glioma is a common malignant tumor of the central nervous system with extremely poor prognosis. An efficient molecular marker for diagnosis and treatment is urgently needed. Fatty acid binding protein 7(FABP7), which regulates intracellular lipid metabolism, is highly expressed in nervous system tumors, but its prognostic value remains undetermined. The present study investigated the relationship between FABP7 expression and prognosis in glioma patients by bioinformatics analysis, as well as immunohistochemically evaluating the effect of FABP7 expression on the efficacy of antiangiogenic drugs. Methods Gene expression and clinical data on patients with glioma were collected from the China Glioma Genome Atlas (CGGA) database, The Cancer Genome Atlas (TCGA), and the Gene Expression Omnibus (GEO) databases. Levels of FABP7 expression and their association with the clinicopathologic characteristics of glioma patients were analyzed in the CGGA database. The relationships between FABP7 expression and clinical findings, such as survival and prognostic, were determined and used for nomogram construction. Mechanisms of action of FABP7 were assessed using GSEA software. FABP7 expression in the tissues of glioma patients treated with apatinib was evaluated immunohistochemically. Results FABP7 was highly expressed in glioma samples, with higher FABP7 expression associated with poorer patient prognosis and more advanced clinicopathological features. Bioinformatics analysis, including survival, receiver operating characteristic curve, and univariate and multivariate Cox analyses, showed that FABP7 was independently prognostic of outcomes in glioma patients. GSEA analysis showed that FABP7 was associated with angiogenesis, with FABP7 having correlation coefficients > 0.4 with seven factors in the angiogenic pathway, POSTN, TIMP1, PDGFA, FGFR1, S100A4, COL5A2, and STC1, and the expression of these factors related to patient prognosis. Immunohistochemistry showed that FABP7 expression was higher in glioma patients with poor survival after apatinib treatment. Conclusions High FABP7 expression is associated with poor prognosis in glioma patients. FABP7, which is important for glioma angiogenesis, may serve as an independent prognostic predictor in glioma patients.
Collapse
|
13
|
Feng W, Zhang Y, Liu W, Wang X, Lei T, Yuan Y, Chen Z, Song W. A Prognostic Model Using Immune-Related Genes for Colorectal Cancer. Front Cell Dev Biol 2022; 10:813043. [PMID: 35252182 PMCID: PMC8893267 DOI: 10.3389/fcell.2022.813043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/04/2022] [Indexed: 11/29/2022] Open
Abstract
There is evidence suggesting that immune genes play pivotal roles in the development and progression of colorectal cancer (CRC). Colorectal carcinoma patient data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) were randomly classified into a training set, a test set, and an external validation set. Differentially expressed gene (DEG) analyses, univariate Cox regression, and the least absolute shrinkage and selection operator (LASSO) were used to identify survival-associated immune genes and develop a prognosis model. Receiver operating characteristic (ROC) analysis and principal component analysis (PCA) were used to evaluate the discrimination of the risk models. The model genes predicted were verified using the Human Protein Atlas (HPA) databases, colorectal cell lines, and fresh CRC and adjacent tissues. To understand the relationship between IRGs and immune invasion and the TME, we analyzed the content of immune cells and scored the TME using CIBERSORT and ESTIMATE algorithms. Finally, we predicted the potential sensitive chemotherapeutic drugs in different risk score groups by the Genomics of Drug Sensitivity in Cancer (GDSC). A total of 491 IRGs were screened, and 14 IRGs were identified to be significantly related to overall survival (OS) and applied to construct an immune-related gene (IRG) prognostic signature (IRGSig) for CRC patients. Calibration plots showed that nomograms have powerful predictive ability. PCA and ROC analysis further verified the predictive value of this fourteen-gene prognostic model in three independent databases. Furthermore, we discovered that the tumor microenvironment changed significantly during the tumor development process, from early to middle to late stage, which may be an essential factor for tumor deterioration. Finally, we selected six commonly used chemotherapeutic drugs that have the potential to be useful in the treatment of CRC. Altogether, immune genes were used to construct a prognosis model for CRC patients, and a variety of methods were used to test the accuracy of this model. In addition, we explored the immune mechanisms of CRC through immune cell infiltration and TME in CRC. Furthermore, we assessed the therapeutic sensitivity of many commonly used chemotherapeutic medicines in individuals with varying risk factors. Finally, the immune risk model and immune mechanism of CRC were thoroughly investigated in this paper.
Collapse
Affiliation(s)
- Wei Feng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongxin Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenwei Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaofeng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tianxiang Lei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yujie Yuan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zehong Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wu Song
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Marques-Magalhães Â, Cruz T, Costa ÂM, Estêvão D, Rios E, Canão PA, Velho S, Carneiro F, Oliveira MJ, Cardoso AP. Decellularized Colorectal Cancer Matrices as Bioactive Scaffolds for Studying Tumor-Stroma Interactions. Cancers (Basel) 2022; 14:cancers14020359. [PMID: 35053521 PMCID: PMC8773780 DOI: 10.3390/cancers14020359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/02/2022] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
More than a physical structure providing support to tissues, the extracellular matrix (ECM) is a complex and dynamic network of macromolecules that modulates the behavior of both cancer cells and associated stromal cells of the tumor microenvironment (TME). Over the last few years, several efforts have been made to develop new models that accurately mimic the interconnections within the TME and specifically the biomechanical and biomolecular complexity of the tumor ECM. Particularly in colorectal cancer, the ECM is highly remodeled and disorganized and constitutes a key component that affects cancer hallmarks, such as cell differentiation, proliferation, angiogenesis, invasion and metastasis. Therefore, several scaffolds produced from natural and/or synthetic polymers and ceramics have been used in 3D biomimetic strategies for colorectal cancer research. Nevertheless, decellularized ECM from colorectal tumors is a unique model that offers the maintenance of native ECM architecture and molecular composition. This review will focus on innovative and advanced 3D-based models of decellularized ECM as high-throughput strategies in colorectal cancer research that potentially fill some of the gaps between in vitro 2D and in vivo models. Our aim is to highlight the need for strategies that accurately mimic the TME for precision medicine and for studying the pathophysiology of the disease.
Collapse
Affiliation(s)
- Ângela Marques-Magalhães
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Tânia Cruz
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Ângela Margarida Costa
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Diogo Estêvão
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Elisabete Rios
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- IPATIMUP-Institute of Pathology and Molecular Immunology, University of Porto, 4200-135 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário São João, 4200-319 Porto, Portugal
| | - Pedro Amoroso Canão
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário São João, 4200-319 Porto, Portugal
| | - Sérgia Velho
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- IPATIMUP-Institute of Pathology and Molecular Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Fátima Carneiro
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- IPATIMUP-Institute of Pathology and Molecular Immunology, University of Porto, 4200-135 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário São João, 4200-319 Porto, Portugal
| | - Maria José Oliveira
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
| | - Ana Patrícia Cardoso
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- Correspondence: ; Tel.: +351-22-607-4900
| |
Collapse
|