1
|
Zheng X, Zhang X, Li D, Wang Z, Zhang J, Li J, Li Y. Integrative bioinformatics and experimental analyses identify U2SURP as a novel lactylation-related prognostic signature in esophageal carcinoma. Immunol Res 2025; 73:45. [PMID: 39900790 DOI: 10.1007/s12026-024-09589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/27/2024] [Indexed: 02/05/2025]
Abstract
The lactylation modification has been implicated in several cancer types; however, the role of lactylation modification-related genes in esophageal carcinoma (EC) remains underexplored. Utilizing a set of 16 lactylation modification-related genes, cohorts of patients with EC were stratified into two distinct clusters, characterized by significant disparities in both survival outcomes and the immune microenvironment. An extensive bioinformatics analysis unveiled 382 differentially expressed genes (DEGs) between these two clusters. A subsequent univariate Cox regression analysis identified 24 DEGs specifically associated with lactylation, forming the basis of a constructed lactylation-related score. The resultant lactylation-related score exhibited notable predictive efficacy for survival and other clinicopathological traits, which was validated through calibration curves, Kaplan-Meier survival curves and the Wilcoxon test. Moreover, the lactylation-related score displayed a close correlation with immune cell infiltration in EC. Notable differential expressions of immune checkpoints and regulators were observed between groups stratified by low and high lactylation scores, with the latter exhibiting a more favorable response to anti-PD-1/PD-L1 therapy. Furthermore, the expression profile of U2 snRNP associated SURP domain containing (U2SURP), a constituent of the lactylation-related score, underwent both ex vivo and in vitro validation. The expression of U2SURP was significantly associated with lactylation levels, histological grade and tumor stage. Notably, knockdown of U2SURP expression inhibited the lactylation levels, immune genes IL-1A and IL-1B, proliferation, migration and invasion of EC cells. In conclusion, the lactylation-related score developed in the present study showed promise in predicting the prognosis and immunotherapeutic responses among patients with EC. Moreover, the identification of U2SUPR as a novel oncogene in EC suggests its potential as a prospective therapeutic target for EC treatment.
Collapse
Affiliation(s)
- Xuan Zheng
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China
| | - Xiaoru Zhang
- Nuclear Medicine Laboratory, Tangshan People's Hospital, Tangshan, 063001, China
| | - Dan Li
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China
| | - Zhuo Wang
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China
| | - Jun Zhang
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China
| | - Jingwu Li
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China.
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China.
| | - Yufeng Li
- The Cancer Institute, Tangshan People's Hospital, Tangshan, 063001, China.
- Hebei Key Laboratory of Molecular Oncology, Tangshan, 063001, China.
| |
Collapse
|
2
|
Wang N, Chen Y, Lin J, Lin Y, Song H, Huang W, Shen L, Chen F, Liu F, Wang J, Qiu Y, Shi B, Li L, Lin L, Pan L, He B. Identification of novel serum lipid metabolism potential markers and metabolic pathways for oral cancer: a population-based study. BMC Cancer 2025; 25:177. [PMID: 39885458 PMCID: PMC11783747 DOI: 10.1186/s12885-025-13561-x] [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: 04/15/2024] [Accepted: 01/20/2025] [Indexed: 02/01/2025] Open
Abstract
OBJECTIVE This study aims to identify potential lipid biomarkers and metabolic pathways associated with oral cancer (OC). Then to establish and evaluate disease classification models capable of distinguishing OC patients from healthy controls. METHODS A total of 41 OC patients and 41 controls were recruited from a hospital in Southeast China to examine the serum lipidomics by Ultra-high Performance Liquid Chromatography Q Exactive Mass Spectrometry (UHPLC-QE-MS). RESULTS The total serum lipid profile showed that triglycerides accounted for the highest proportion of total metabolites, reaching 35.90% of the total. A total of 74 different metabolites were screened (12 up-regulated and 62 down-regulated), mainly enriched in the glycerophospholipid metabolism pathway. The three most significant changes in lipid metabolites were phosphatidylcholine (PC(18:3e/17:2)), acylcarnitine (ACar(14:2)), and glucuronosyldiacylglycerol (GlcADG(14:1/14:1)). The disease classification model, constructed using a KNN algorithm with 13 metabolites selected through LASSO screening, achieved the best performance, with an AUC of 0.978 (0.955-1.000). CONCLUSION Lipid metabolic biomarkers identified in this study exhibit potential as candidate biomarkers for OC diagnosis. Further validation through prospective studies is required to confirm their clinical utility in early detection.
Collapse
Affiliation(s)
- Na Wang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fujian, 350108, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fujian, China
| | - Yujia Chen
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fujian, 350108, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fujian, China
| | - Jianli Lin
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fujian, 350108, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fujian, China
| | - Yulan Lin
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fujian, 350108, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fujian, China
| | - Haoyuan Song
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fujian, 350108, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fujian, China
| | - Weihai Huang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fujian, 350108, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fujian, China
| | - Liling Shen
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fujian, 350108, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fujian, China
| | - Fa Chen
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fujian, 350108, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fujian, China
| | - Fengqiong Liu
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fujian, 350108, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fujian, China
| | - Jing Wang
- Laboratory Center, School of Public Health, The Major Subject of Environment and Health of Fujian Key Universities, Fujian Medical University, Fujian, China
| | - Yu Qiu
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Bin Shi
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ling Li
- International Nursing School, Hainan Medical University, Hainan, China
| | - Lisong Lin
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lizhen Pan
- Department of Nursing, the First Affiliated Hospital of Fujian Medical University, 88 Jiaotong Road, Fujian, 350004, China.
| | - Baochang He
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fujian, 350108, China.
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fujian, China.
| |
Collapse
|
3
|
Lin RY, Chen RP, Lin FQ. Crotonylation-Related Prognostic Model of Esophageal Squamous Cell Carcinoma Based on Transcriptome Analysis and Single-Cell Sequencing Analysis. Int J Gen Med 2025; 18:415-436. [PMID: 39895826 PMCID: PMC11784406 DOI: 10.2147/ijgm.s493800] [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: 10/04/2024] [Accepted: 01/21/2025] [Indexed: 02/04/2025] Open
Abstract
Background Crotonylation is an emerging lysine acylation modification implicated in various diseases, yet its role in esophageal squamous cell cancer (ESCC) is unexplored. This study aimed to investigate the role of crotonylation-related genes (CRGs) in ESCC using bioinformatics approaches. Methods We included three ESCC datasets and 24 CRGs. Differentially expressed genes (DEGs) from TCGA-ESCA were intersected with key module genes related to CRGs to identify candidate genes. Univariate and LASSO regression analyses were conducted to select prognostic genes, which were then used to construct risk models. Independent prognostic analysis and nomogram construction followed. Functional enrichment and immune infiltration analyses were performed using the prognostic genes. Single-cell analysis was conducted to assess cell communication and pseudotemporal dynamics in key cells. Results Intersection of 1529 DEGs with 1,048 key module genes yielded 55 candidate genes. OSM, FABP3, MICB, and FAM189A2 were identified as prognostic genes. These genes were used to classify ESCA patients into different risk groups and construct a nomogram. FABP3 and FAM189A2 were enriched in neuroactive ligand-receptor interaction and ribosome terms. MICB and FABP3 showed strong positive correlations with natural killer T (NKT) cells, while FAM189A2 negatively correlated with gamma delta T (γδT) cells. Single-cell analysis identified mast cells and neutrophils as key cells, differentiating into seven and three states, respectively. Conclusion Four genes (OSM, FABP3, MICB, and FAM189A2) were identified as prognostic crotonylation-related genes in ESCC, potentially involved in its pathogenesis. OSM was negatively correlated with ESCC, while FABP3 and MICB were positively correlated.
Collapse
Affiliation(s)
- Ruo-Yang Lin
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Ren-Pin Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Fu-Qiang Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| |
Collapse
|
4
|
Ma X, He Y, Lv D, Chen X, Hong Z, Chai Y, Liu Y. Optimization of metabolomics pretreatment method of cholangiocarcinoma cells based on ultrahigh performance liquid chromatography coupled with mass spectrometry. J Pharm Biomed Anal 2025; 252:116508. [PMID: 39426275 DOI: 10.1016/j.jpba.2024.116508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024]
Abstract
Metabolomics intends to maximize the quantity of available metabolites for the global metabolome, which largely depends on sample pretreatment protocols. However, there are few studies that comprehensively examined the effects of extraction and reconstitution solvents on metabolome coverage of adherent mammalian cells. In this study, the human cholangiocarcinoma TFK-1 cells were chosen as a cell model, and eight extraction solvents and five reconstitution solvents were used for the pretreatment based on ultrahigh performance liquid chromatography coupled with mass spectrometry (UPLC/MS). The coverage, reproducibility, and stability of the data were norms to evaluate the effectiveness of different extraction solvents and reconstitution solvents. Based on the number of metabolites, the mean Euclidean distance (EDMEAN) in the principal component analysis (PCA) 3D score plots and the relative standard deviation (RSD) distribution of metabolites, it was demonstrated that MeOH-CHCl3-H2O (8:1:1, v/v/v) was the optimal extraction solvent and MeOH-H2O (1:1, v/v) or H2O was superior to other reconstitution solvents for RP column analysis, and the extraction solvent MeOH-ACN-H2O (2:2:1, v/v/v) and the reconstitution solvents ACN-H2O (4:1, v/v) or MeOH-H2O (1:1, v/v) provide the best performance for HILIC column analysis. The optimized pretreatment methods explored in this study expand the coverage of polar and non-polar metabolites and improve the reproducibility and stability of the metabolic data, which can be applied to UPLC/MS-based global metabolomics study on cholangiocarcinoma cells, potentially providing better extraction solvents and reconstitution solvents for other adherent mammalian cells with similar chemical and physical properties.
Collapse
Affiliation(s)
- Xiaoyu Ma
- Department of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai 200433, China; School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China
| | - Yongping He
- Department of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Department of Pharmacy, Chongzuo People's Hospital, Chongzuo 532200, China
| | - Diya Lv
- Department of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Xiaofei Chen
- Department of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai 200433, China
| | - Zhanying Hong
- Department of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai 200433, China.
| | - Yifeng Chai
- Department of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai 200433, China.
| | - Yue Liu
- Department of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai 200433, China.
| |
Collapse
|
5
|
Shahpari M, Hashemi M, Younesirad T, Hasanzadeh A, Mosanne MM, Ahmadifard M. The functional roles of competitive endogenous RNA (ceRNA) networks in apoptosis in human cancers: The circRNA/miRNA/mRNA regulatory axis and cell signaling pathways. Heliyon 2024; 10:e37089. [PMID: 39524849 PMCID: PMC11546195 DOI: 10.1016/j.heliyon.2024.e37089] [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: 08/18/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024] Open
Abstract
Circular RNAs are noncoding RNAs with circular conformation mainly due to backsplicing event. CircRNAs can potentially impact cell biological processes by interacting with cell signaling pathways. Numerous circRNAs have been found to be aberrantly expressed in a variety of cancers. These RNAs can act as ceRNA (competitive endogenous RNA) by sponging certain miRNAs to form circRNA/miRNA/mRNA networks. Dysregulation of ceRNA networks may lead to dysfunctions in various cell pathways, which modulate apoptosis-associated genes and ultimately result in cancer progression. Since disruption of apoptosis is one of the leading causes of cancer development, one approach for cancer treatment is to drive cells toward apoptosis. In this review, we present a summary of studies on the role of ceRNA networks in cellular signaling pathways that regulate apoptosis; these networks are suggested to be potential biomarkers for cancer treatment.
Collapse
Affiliation(s)
| | | | - Tayebeh Younesirad
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Aida Hasanzadeh
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad mahdi Mosanne
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohamadreza Ahmadifard
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
6
|
D'Mello R, Hüttmann N, Minic Z, V Berezovski M. Untargeted metabolomic profiling of small extracellular vesicles reveals potential new biomarkers for triple negative breast cancer. Metabolomics 2024; 20:123. [PMID: 39487276 DOI: 10.1007/s11306-024-02191-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024]
Abstract
INTRODUCTION Breast Cancer (BC) is one of the most diagnosed malignancies among women and the second leading cause of cancer related death in North America. Triple Negative BC (TNBC), one of the most severe subtypes of BC, is extremely aggressive and has a higher chance of occurrence in women under 50 years of age. Due to a lack of regular mammographic testing in women under 50, many individuals with TNBC are diagnosed late which can decrease their survival rate. Currently, liquid biopsy is being investigated as a potentially less-invasive alternative to traditional breast tissue biopsy, but this approach is not completely reliable. Blood contains extracellular vesicles (EVs), which carry biomolecular cargo and play a role in BC progression and metastasis. Examination of small EVs could potentially yield metabolite biomarkers for early BC diagnosis. OBJECTIVE We aim to study metabolites in small EVs to find biomarkers for BC diagnosis. METHODS In this work, an untargeted nano-LC MS/MS metabolomics approach was used to analyze metabolites from small EVs derived from metastatic MDA-MB-231 and compare it with a non-cancerous MCF10A cell line. RESULTS Two metabolites, LysoPC 22:6/0:0 and N-acetyl-L-Phenylalanine, unique to sEVs of MDA-MB-231, were identified, validated, and proposed as potential BC biomarkers. CONCLUSION Metabolites from sEVs may be used for BC diagnosis.
Collapse
Affiliation(s)
- Rochelle D'Mello
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
| | - Nico Hüttmann
- John L. Holmes Biological Mass Spectrometry Facility, University of Ottawa, Ottawa, Canada
| | - Zoran Minic
- John L. Holmes Biological Mass Spectrometry Facility, University of Ottawa, Ottawa, Canada.
| | - Maxim V Berezovski
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada.
- John L. Holmes Biological Mass Spectrometry Facility, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
7
|
Zhang J, Guo J, Gu B, Wang F, Li Y, Shang L, Jiang W, Ma J, Wu W. Shikonin Induces Autophagy and Apoptosis in Esophageal Cancer EC9706 Cells by Regulating the AMPK/mTOR/ULK Axis. Anal Cell Pathol (Amst) 2024; 2024:7752299. [PMID: 39502521 PMCID: PMC11537739 DOI: 10.1155/2024/7752299] [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: 03/16/2024] [Revised: 08/29/2024] [Accepted: 09/18/2024] [Indexed: 11/08/2024] Open
Abstract
Shikonin is a plant medicine extracted from Lithospermum, which dominate influential antioxidant and antitumor effect. Here, we report that shikonin was capable of inducing human esophageal cancer EC9706 cell apoptosis and autophagy, in a time- and dose-dependent manner. Shikonin exposure repressed cell viability and migration and invasion capabilities and caused EC9706 cell autophagy and apoptosis by activating the AMPK/mTOR/ULK axis. Autophagy inhibition secured EC9706 cells against shikonin-induced autophagy and apoptosis and reversed the upregulation of AMPK and ULK phosphorylation and downregulation of mTOR phosphorylation provoked by shikonin. In summary, shikonin instigates EC9706 cell apoptosis and autophagy using the target AMPK/mTOR/ULK signal pathway axis, which provides a potential new target to treat human esophageal cancer.
Collapse
Affiliation(s)
- Junli Zhang
- Bengbu Third People's Hospital Affiliated to Bengbu Medical University, Bengbu 233030, Anhui, China
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Jiayi Guo
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Biao Gu
- Bengbu Third People's Hospital Affiliated to Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Fen Wang
- Bengbu Third People's Hospital Affiliated to Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Yi Li
- Bengbu Third People's Hospital Affiliated to Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Ling Shang
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Wendi Jiang
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Junrao Ma
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu 233030, Anhui, China
| | - Wenjuan Wu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, Anhui, China
| |
Collapse
|
8
|
Deng J, Wu S, Huang Y, Deng Y, Yu K. Esophageal cancer risk is influenced by genetically determined blood metabolites. Medicine (Baltimore) 2024; 103:e40122. [PMID: 39470544 PMCID: PMC11521038 DOI: 10.1097/md.0000000000040122] [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: 05/24/2024] [Revised: 07/13/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
It remains unclear what causes esophageal cancer (EC), but blood metabolites have been connected to it. Our study performed a Mendelian randomization (MR) analysis to assess the causality from genetically proxied 1400 blood metabolites to EC level. A two-sample MR analysis was employed to evaluate the causal relationship between 1400 blood metabolites and EC. Initially, the EC genome-wide association study (GWAS) data (from Jiang L et al) were examined, leading to the identification of certain metabolites. Subsequently, another set of EC GWAS data from FINNGEN was utilized to validate the findings. Causality was primarily determined through inverse variance weighting, with additional support from the MR-Egger, weighted median, and MR-PRESSO models. Heterogeneity was assessed using the MR Cochran Q test. The MR-Egger intercept and MR-PRESSO global methods were employed to detect multicollinearity. In this study, Bonferroni corrected P value was used for significance threshold. We found 2 metabolites with overlaps, which are lipids. Docosatrienoate (22:3n3) was found to be causally associated with a decreased risk of EC, as evidenced by the EC GWAS data (from Jiang et al) (odds ratio [OR] = 0.620, 95% confidence interval [CI] = 0.390-0.986, P = .044) and the EC GWAS data (from FINNGEN) (OR = 0.77, 95% CI = 0.6-0.99, P = .042), these results were consistent across both data sets. Another overlapping metabolite, glycosyl-N-(2-hydroxyneuramoyl)-sphingosine, was associated with the risk of ES, with EC GWAS data (from Jiang L et al) (OR = 1.536, 95% CI = 1.000-2.360, P = .049), while EC GWAS data (from FINNGEN) (OR = 0.733, 95% CI = 0.574-0.937, P = .013), the 2 data had opposite conclusions. The findings of this study indicate a potential association between lipid metabolites (Docosatrienoate (22:3n3) and glycosyl-N-(2-hydroxynervonoyl)-sphingosine (d18:1/24:1 (2OH))) and the risk of esophageal carcinogenesis.
Collapse
Affiliation(s)
- Jieyin Deng
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
| | - Silin Wu
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
- School of Clinical Medicine, North Sichuan Medical College, Sichuan, China
| | - Ye Huang
- Department of Nursing, Nursing School, Chengdu Medical College, Chengdu, China
| | - Yi Deng
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
| | - Ke Yu
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
| |
Collapse
|
9
|
Jiang M, Yu H. Ginsenoside 20(S)-Rg3 Hinders Esophageal Squamous Cell Carcinoma Cells Malignant Behaviors by miR-210-3p/B4GALT5 Axis. Cell Biochem Biophys 2024:10.1007/s12013-024-01566-5. [PMID: 39422791 DOI: 10.1007/s12013-024-01566-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 10/19/2024]
Abstract
Ginsenoside 20(S)-Rg3 (20(S)-Rg3) belongs to a natural chemical with an anti-tumor function, but its potential function and underlying mechanism in esophageal squamous cell carcinoma (ESCC) are unknown. Several reports have manifested that microRNA (miRNA) miR-210-3p functions as a tumor repressor in tumors, but its biofunction in ESCC remains obscure. Herein, the role and interaction of 20(S)-Rg3 and miR-210-3p in ESCC cells were investigated. We performed a series of functional experiments to validate that 20(S)-Rg3 notably restrained ESCC cell proliferation and migration while promoting cell apoptosis. Besides, miR-210-3p was found to be lowly expressed in ESCC cells. Overexpressing miR-210-3p suppressed the malignant behaviors of ESCC cells. More importantly, 20(S)-Rg3 could upregulate miR-210-3p expression in ESCC cells. MiR-210-3p knockdown offset the inhibitive impacts of 20(S)-Rg3 treatment on ESCC cell growth and migration. Furthermore, through luciferase reporter assay, beta-1,4-galactosyltransferase 5 (B4GALT5) was certified to be targeted by miR-210-3p. B4GALT5 upregulation neutralized the suppressive function of 20(S)-Rg3 on ESCC progression. Overall, 20(S)-Rg3 attenuated malignant behaviors of ESCC cells by modulating miR-210-3p/B4GALT5 axis, indicating 20(S)-Rg3 has therapeutic potential for ESCC.
Collapse
Affiliation(s)
- Min Jiang
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China.
| |
Collapse
|
10
|
Shirima CA, Bleotu C, Spandidos DA, El-Naggar AK, Pircalabioru GG, Michalopoulos I. Epithelial‑derived head and neck squamous tumourigenesis (Review). Oncol Rep 2024; 52:141. [PMID: 39219259 PMCID: PMC11358675 DOI: 10.3892/or.2024.8800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs), a heterogeneous group of cancers that arise from the mucosal epithelia cells in the head and neck areas, present great challenges in diagnosis, treatment and prognosis due to their complex aetiology and various clinical manifestations. Several factors, including smoking, alcohol consumption, oncogenic genes, growth factors, Epstein‑Barr virus and human papillomavirus infections can contribute to HNSCC development. The unpredictable tumour microenvironment adds to the complexity of managing HNSCC. Despite significant advances in therapies, the prediction of outcome after treatment for patients with HNSCC remains poor, and the 5‑year overall survival rate is low due to late diagnosis. Early detection greatly increases the chances of successful treatment. The present review aimed to bring together the latest findings related to the molecular mechanisms of HNSCC carcinogenesis and progression. Comprehensive genomic, transcriptomic, metabolomic, microbiome and proteomic analyses allow researchers to identify important biological markers such as genetic alterations, gene expression signatures and protein markers that drive HNSCC tumours. These biomarkers associated with the stages of initiation, progression and metastasis of cancer are useful in the management of patients with cancer in order to improve their life expectancy and quality of life.
Collapse
Affiliation(s)
- Charles Adolfu Shirima
- Centre of Systems Biology, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Faculty of Biology, University of Bucharest, 050663 Bucharest, Romania
| | - Coralia Bleotu
- Cellular and Molecular Pathology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Adel K. El-Naggar
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | | | - Ioannis Michalopoulos
- Centre of Systems Biology, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
11
|
Chen J, Ying K, Sun J, Wang Y, Ji M, Sun Y. NEDD4L affects KLF5 stability through ubiquitination to control ferroptosis and radiotherapy resistance in oesophageal squamous cell carcinoma. J Cell Mol Med 2024; 28:e70062. [PMID: 39317954 PMCID: PMC11422174 DOI: 10.1111/jcmm.70062] [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: 04/11/2024] [Revised: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024] Open
Abstract
Oesophageal squamous cell carcinoma (ESCC) contributes to high mortality. Modulating ferroptosis may reverse resistance to radiotherapy. This article was to explore the ubiquitination modification of KLF5 and its effect on ferroptosis in ESCC. KLF5 was under-expressed by shRNA plasmids in the cells and ROS levels were analysed by flow cytometry, ferroptotic gene expression was detected by qRT-PCR, MDA and GSH levels were determined by ELISA, cell morphology was observed by transmission electron microscopy, and Fe ion levels were analysed by immunofluorescence. Cells were treated with Ferrostatin-1 and NAC and analysed for cell proliferation by colony formation assay, cell migration and invasion by Transwell assays, and apoptosis by flow cytometry. DNA damage in cells was also analysed using comet assay, EdU doping assay, γH2AX fluorescence, DNA-PKcs and PCR. NEDD4L and KLF5 binding was analysed by immunoprecipitation. Changes in ferroptosis, DNA damage and resistance were analysed in cells with both silencing NEDD4L and KLF5. Changes in tumour resistance to radiation were analysed in mice underexpressing NEDD4L and KLF5. Low expression of KLF5 significantly promotes cellular lipid peroxidation levels, with decreased expression of SOD and GPX4, and increased expression of ACSL4. Concurrently, MDA levels deplete GSH, and cells exhibit typical ferroptotic morphology with increased Fe2+ content. KLF5 inhibition results in enhanced cellular clonogenicity, migration and invasion activities, reduced apoptosis, increased tail DNA, nuclear EdU incorporation, nuclear γH2AX foci and elevated expression of DNA-PKcs, LIG4, RAD9B and BMI1. Ferrostatin-1 and NAC reverse these effects. NEDD4L ubiquitination modifies and degrades KLF5, with NEDD4L/KLF5 inhibition mitigating cellular ferroptosis and DNA damage, thereby promoting radiosensitivity both in vitro and in vivo. NEDD4L increases radiosensitivity by accelerating cellular ferroptosis via ubiquitination modification of KLF5.
Collapse
Affiliation(s)
- Jinjin Chen
- Department of OncologyThe First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical UniversityYanchengJiangsuChina
| | - Kaijun Ying
- Department of Thoracic SurgeryThe First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical UniversityYanchengJiangsuChina
| | - Jian Sun
- Department of Thoracic SurgeryThe First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical UniversityYanchengJiangsuChina
| | - Yao Wang
- Department of Thoracic SurgeryThe First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical UniversityYanchengJiangsuChina
| | - Mingming Ji
- Department of Thoracic SurgeryThe First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical UniversityYanchengJiangsuChina
| | - Yunhao Sun
- Department of Thoracic SurgeryThe First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical UniversityYanchengJiangsuChina
| |
Collapse
|
12
|
Yue J, Fang H, Chen S, Gu L, Ren G. Causal role of blood metabolites in HER-positive and HER-negative breast cancer: a Mendelian randomization (MR) study. Aging (Albany NY) 2024; 16:11626-11655. [PMID: 39103210 PMCID: PMC11346783 DOI: 10.18632/aging.206042] [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: 01/16/2024] [Accepted: 07/05/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Previous studies provide evidence that in vivo metabolites are associated with breast cancer (BC). However, the causal relationship between blood metabolites and BC remains unclear. METHOD Comprehensive two-sample Mendelian randomization analysis was conducted to determine the causal association between 1400 publicly available genetic data on metabolic factors and human epidermal growth factor receptor positive (HER+) BC or HER- BC in this study. RESULT Epiandrosterone sulfate levels (OR = 1.07, 95% CI = 1.02 ~ 1.10, p = 0.0013), 5alpha-androstan-3beta,17beta-diol monosulfate (2) levels (OR = 1.07, 95% CI = 1.03 ~ 1.12, p = 0.0012), glycohyocholate levels (OR = 0.85, 95% CI = 0.77 ~ 0.93, p = 0.0007) and etiocholanolone glucuronide levels (OR = 1.12, 95% CI = 1.05 ~ 1.20, p = 0.0013) were causally correlated with HER+ BC. 5 metabolites were causally correlated with HER- BC: Vanillic acid glycine levels (OR = 1.14, 95% CI = 1.06 ~ 1.22, p = 0.0003), Thyroxine levels (OR = 1.26, 95% CI = 1.11 ~ 1.44, p = 0.0004), 1-palmitoyl-2-linoleoyl-GPI (16:0/18:2) levels (OR = 0.86, 95% CI = 0.79 ~ 0.94, p = 0.0010), N-acetylphenylalanine levels (OR = 1.12, 95% CI = 1.05 ~ 1.19, p = 0.0007) and Glucose-to-mannose ratio (OR = 1.15, 95% CI = 1.06 ~ 1.24, p = 0.0008). Two common causally related metabolites were identified: Gamma-glutamyl glutamate and X-12849 levels. CONCLUSIONS Our study has respectively demonstrated the connection between blood metabolites and HER+ or HER- BC by genetic means, thereby offering opportunities for therapeutic targets.
Collapse
Affiliation(s)
- Jian Yue
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast Surgery, Gaozhou People’s Hospital, Gaozhou, China
| | - Huiying Fang
- Department of Breast Cancer Center, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, China
| | - Sheng Chen
- Department of Breast Surgery, Gaozhou People’s Hospital, Gaozhou, China
| | - Lei Gu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Liu MJ, Zhao Y, Li QT, Lei XY, He KY, Guo JR, Yang JY, Yan ZH, Wu DH, Zhang L, Jian YP, Xu ZX. HMGA1 promotes the progression of esophageal squamous cell carcinoma by elevating TKT-mediated upregulation of pentose phosphate pathway. Cell Death Dis 2024; 15:541. [PMID: 39080260 PMCID: PMC11289123 DOI: 10.1038/s41419-024-06933-x] [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: 08/27/2023] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) possesses a poor prognosis and treatment outcome. Dysregulated metabolism contributes to unrestricted growth of multiple cancers. However, abnormal metabolism, such as highly activated pentose phosphate pathway (PPP) in the progression of ESCC remains largely unknown. Herein, we report that high-mobility group AT-hook 1 (HMGA1), a structural transcriptional factor involved in chromatin remodeling, promoted the development of ESCC by upregulating the PPP. We found that HMGA1 was highly expressed in ESCC. Elevated HMGA1 promoted the malignant phenotype of ESCC cells. Conditional knockout of HMGA1 markedly reduced 4-nitroquinoline-1-oxide (4NQO)-induced esophageal tumorigenesis in mice. Through the metabolomic analysis and the validation assay, we found that HMGA1 upregulated the non-oxidative PPP. With the transcriptome sequencing, we identified that HMGA1 upregulated the expression of transketolase (TKT), which catalyzes the reversible reaction in non-oxidative PPP to exchange metabolites with glycolytic pathway. HMGA1 knockdown suppressed the PPP by downregulating TKT, resulting in the reduction of nucleotides in ESCC cells. Overexpression of HMGA1 upregulated PPP and promoted the survival of ESCC cells by activating TKT. We further characterized that HMGA1 promoted the transcription of TKT by interacting with and enhancing the binding of transcription factor SP1 to the promoter of TKT. Therapeutics targeting TKT with an inhibitor, oxythiamine, reduced HMGA1-induced ESCC cell proliferation and tumor growth. Together, in this study, we identified a new role of HMGA1 in ESCCs by upregulating TKT-mediated activation of PPP. Our results provided a new insight into the role of HMGA1/TKT/PPP in ESCC tumorigenesis and targeted therapy.
Collapse
Affiliation(s)
- Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yuan Zhao
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Qiu-Tong Li
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Zhen-Hua Yan
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| |
Collapse
|
14
|
Balonov I, Mattis M, Jarmusch S, Koletzko B, Heinrich K, Neumann J, Werner J, Angele MK, Heiliger C, Jacob S. Metabolomic profiling of upper GI malignancies in blood and tissue: a systematic review and meta-analysis. J Cancer Res Clin Oncol 2024; 150:331. [PMID: 38951269 PMCID: PMC11217139 DOI: 10.1007/s00432-024-05857-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024]
Abstract
OBJECTIVE To conduct a systematic review and meta-analysis of case-control and cohort human studies evaluating metabolite markers identified using high-throughput metabolomics techniques on esophageal cancer (EC), cancer of the gastroesophageal junction (GEJ), and gastric cancer (GC) in blood and tissue. BACKGROUND Upper gastrointestinal cancers (UGC), predominantly EC, GEJ, and GC, are malignant tumour types with high morbidity and mortality rates. Numerous studies have focused on metabolomic profiling of UGC in recent years. In this systematic review and meta-analysis, we have provided a collective summary of previous findings on metabolites and metabolomic profiling associated with EC, GEJ and GC. METHODS Following the PRISMA procedure, a systematic search of four databases (Embase, PubMed, MEDLINE, and Web of Science) for molecular epidemiologic studies on the metabolomic profiles of EC, GEJ and GC was conducted and registered at PROSPERO (CRD42023486631). The Newcastle-Ottawa Scale (NOS) was used to benchmark the risk of bias for case-controlled and cohort studies. QUADOMICS, an adaptation of the QUADAS-2 (Quality Assessment of Diagnostic Accuracy) tool, was used to rate diagnostic accuracy studies. Original articles comparing metabolite patterns between patients with and without UGC were included. Two investigators independently completed title and abstract screening, data extraction, and quality evaluation. Meta-analysis was conducted whenever possible. We used a random effects model to investigate the association between metabolite levels and UGC. RESULTS A total of 66 original studies involving 7267 patients that met the required criteria were included for review. 169 metabolites were differentially distributed in patients with UGC compared to healthy patients among 44 GC, 9 GEJ, and 25 EC studies including metabolites involved in glycolysis, anaerobic respiration, tricarboxylic acid cycle, and lipid metabolism. Phosphatidylcholines, eicosanoids, and adenosine triphosphate were among the most frequently reported lipids and metabolites of cellular respiration, while BCAA, lysine, and asparagine were among the most commonly reported amino acids. Previously identified lipid metabolites included saturated and unsaturated free fatty acids and ketones. However, the key findings across studies have been inconsistent, possibly due to limited sample sizes and the majority being hospital-based case-control analyses lacking an independent replication group. CONCLUSION Thus far, metabolomic studies have provided new opportunities for screening, etiological factors, and biomarkers for UGC, supporting the potential of applying metabolomic profiling in early cancer diagnosis. According to the results of our meta-analysis especially BCAA and TMAO as well as certain phosphatidylcholines should be implicated into the diagnostic procedure of patients with UGC. We envision that metabolomics will significantly enhance our understanding of the carcinogenesis and progression process of UGC and may eventually facilitate precise oncological and patient-tailored management of UGC.
Collapse
Affiliation(s)
- Ilja Balonov
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Minca Mattis
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Stefanie Jarmusch
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Dr. Von Hauner Children's Hospital, Ludwig-Maximilians-University Munich Medical Center, Lindwurmstraße 4, 80337, Munich, Germany
| | - Kathrin Heinrich
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Jens Neumann
- Institute of Pathology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Jens Werner
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Martin K Angele
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Christian Heiliger
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Sven Jacob
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Ludwig-Maximilians-University (LMU) Munich, Marchioninistr. 15, 81377, Munich, Germany.
| |
Collapse
|
15
|
He Y, Yu Q, Ma X, Lv D, Wang H, Qiu W, Chen XF, Jiao Y, Liu Y. A metabolomics approach reveals metabolic disturbance of human cholangiocarcinoma cells after parthenolide treatment. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118075. [PMID: 38513779 DOI: 10.1016/j.jep.2024.118075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 03/09/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tanacetum parthenium (L.) Schultz-Bip, commonly known as feverfew, has been traditionally used to treat fever, migraines, rheumatoid arthritis, and cancer. Parthenolide (PTL), the main bioactive ingredient isolated from the shoots of feverfew, is a sesquiterpene lactone with anti-inflammatory and antitumor properties. Previous studies showed that PTL exerts anticancer activity in various cancers, including hepatoma, cholangiocarcinoma, acute myeloid leukemia, breast, prostate, and colorectal cancer. However, the metabolic mechanism underlying the anticancer effect of PTL remains poorly understood. AIM OF THE STUDY To explore the anticancer activity and underlying mechanism of PTL in human cholangiocarcinoma cells. MATERIAL AND METHODS In this investigation, the effects and mechanisms of PTL on human cholangiocarcinoma cells were investigated via a liquid chromatography/mass spectrometry (LC/MS)-based metabolomics approach. First, cell proliferation and apoptosis were evaluated using cell counting kit-8 (CCK-8), flow cytometry analysis, and western blotting. Then, LC/MS-based metabolic profiling along with orthogonal partial least-squares discriminant analysis (OPLS-DA) has been constructed to distinguish the metabolic changes between the negative control group and the PTL-treated group in TFK1 cells. Next, enzyme-linked immunosorbent assay (ELISA) was applied to investigate the changes of metabolic enzymes associated with significantly alerted metabolites. Finally, the metabolic network related to key metabolic enzymes, metabolites, and metabolic pathways was established using MetaboAnalyst 5.0 and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Database. RESULTS PTL treatment could induce the proliferation inhibition and apoptosis of TFK1 in a concentration-dependent manner. Forty-three potential biomarkers associated with the antitumor effect of PTL were identified, which primarily related to glutamine and glutamate metabolism, alanine, aspartate and glutamate metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine metabolism, arginine biosynthesis, arginine and proline metabolism, glutathione metabolism, nicotinate and nicotinamide metabolism, pyrimidine metabolism, fatty acid metabolism, phospholipid catabolism, and sphingolipid metabolism. Pathway analysis of upstream and downstream metabolites, we found three key metabolic enzymes, including glutaminase (GLS), γ-glutamyl transpeptidase (GGT), and carnitine palmitoyltransferase 1 (CPT1), which mainly involved in glutamine and glutamate metabolism, glutathione metabolism, and fatty acid metabolism. The changes of metabolic enzymes associated with significantly alerted metabolites were consistent with the levels of metabolites, and the metabolic network related to key metabolic enzymes, metabolites, and metabolic pathways was established. PTL may exert its antitumor effect against cholangiocarcinoma by disturbing metabolic pathways. Furthermore, we selected two positive control agents that are considered as first-line chemotherapy standards in cholangiocarcinoma therapy to verify the reliability and accuracy of our metabolomic study on PTL. CONCLUSION This research enhanced our comprehension of the metabolic profiling and mechanism of PTL treatment on cholangiocarcinoma cells, which provided some references for further research into the anti-cancer mechanisms of other drugs.
Collapse
Affiliation(s)
- Yongping He
- School of Pharmacy, Guangxi Medical University, Guangxi, Nanning, 530021, China; School of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai, 200433, China; Department of Pharmacy, The People's Hospital of Chongzuo, Guangxi, Chongzuo, 532200, China
| | - Qianxue Yu
- School of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Xiaoyu Ma
- School of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Diya Lv
- School of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Hui Wang
- School of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai, 200433, China
| | - Weian Qiu
- School of Pharmacy, Guangxi Medical University, Guangxi, Nanning, 530021, China
| | - Xiao Fei Chen
- School of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai, 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai, 200433, China
| | - Yang Jiao
- School of Pharmacy, Guangxi Medical University, Guangxi, Nanning, 530021, China.
| | - Yue Liu
- School of Pharmacy, Naval Medical University (Second Military Medical University), Shanghai, 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai, 200433, China.
| |
Collapse
|
16
|
Liu M, Tian H, Wang M, Guo C, Xu R, Li F, Liu A, Yang H, Duan L, Shen L, Wu Q, Liu Z, Liu Y, Liu F, Pan Y, Hu Z, Chen H, Cai H, He Z, Ke Y. Construction and validation of serum Metabolic Risk Score for early warning of malignancy in esophagus. iScience 2024; 27:109965. [PMID: 38832013 PMCID: PMC11144720 DOI: 10.1016/j.isci.2024.109965] [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: 11/11/2023] [Revised: 03/20/2024] [Accepted: 05/09/2024] [Indexed: 06/05/2024] Open
Abstract
Using noninvasive biomarkers to identify high-risk individuals prior to endoscopic examination is crucial for optimization of screening strategies for esophageal squamous cell carcinoma (ESCC). We conducted a nested case-control study based on two community-based screening cohorts to evaluate the warning value of serum metabolites for esophageal malignancy. The serum samples were collected at enrollment when the cases had not been diagnosed. We identified 74 differential metabolites and two prominent perturbed metabolic pathways, and constructed Metabolic Risk Score (MRS) based on 22 selected metabolic predictors. The MRS generated an area under the receiver operating characteristics curve (AUC) of 0.815. The model performed well for the within-1-year interval (AUC: 0.868) and 1-to-5-year interval (AUC: 0.845) from blood draw to diagnosis, but showed limited ability in predicting long-term cases (>5 years). In summary, the MRS could serve as a potential early warning and risk stratification tool for establishing a precision strategy of ESCC screening.
Collapse
Affiliation(s)
- Mengfei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hongrui Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Minmin Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
- Department of Global Health, School of Public Health, Peking University, Beijing 100191, China
| | - Chuanhai Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ruiping Xu
- Anyang Cancer Hospital, Anyang 455000, China
| | - Fenglei Li
- Hua County People’s Hospital, Anyang 456400, China
| | - Anxiang Liu
- Endoscopy Center, Anyang Cancer Hospital, Anyang 455000, China
| | - Haijun Yang
- Department of Pathology, Anyang Cancer Hospital, Anyang 455000, China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Qi Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Endoscopy Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhen Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ying Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fangfang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yaqi Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhe Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Huanyu Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hong Cai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhonghu He
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yang Ke
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
17
|
Xie R, Lin J, Li W, Chen H, Zhang J, Zhong M, Xue J, Mo C, Chen L, Zhu Y, Chen X, Xu S. Homogentisic acid metabolism inhibits papillary thyroid carcinoma proliferation through ROS and p21-induced cell cycle arrest. Life Sci 2024; 347:122682. [PMID: 38702025 DOI: 10.1016/j.lfs.2024.122682] [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: 02/28/2024] [Revised: 04/09/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Thyroid cancer is one of the most common primary endocrine malignancies worldwide, and papillary thyroid carcinoma (PTC) is the predominant histological type observed therein. Although PTC has been studied extensively, our understanding of the altered metabolism and metabolic profile of PTC tumors is limited. We identified that the content of metabolite homogentisic acid (HGA) in PTC tissues was lower than that in adjacent non-cancerous tissues. We evaluated the potential of HGA as a novel molecular marker in the diagnosis of PTC tumors, as well as its ability to indicate the degree of malignancy. Studies have further shown that HGA contributes to reactive oxygen species (ROS) associated oxidative stress, leading to toxicity and inhibition of proliferation. In addition, HGA caused an increase in p21 expression levels in PTC cells and induced G1 arrest. Moreover, we found that the low HGA content in PTC tumors was due to the low expression levels of tyrosine aminotransferase (TAT) and p-hydroxyphenylpyruvate hydroxylase (HPD), which catalyze the conversion of tyrosine to HGA. The low expression levels of TAT and HPD are strongly associated with a higher probability of PTC tumor invasion and metastasis. Our study demonstrates that HGA could be used to diagnose PTC and provides mechanisms linking altered HGA levels to the biological behavior of PTC tumors.
Collapse
Affiliation(s)
- Ruiwang Xie
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Junyu Lin
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Weiwei Li
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Huaying Chen
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Junsi Zhang
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Minjie Zhong
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Jiajie Xue
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Caiqin Mo
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Ling Chen
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Youzhi Zhu
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Xiangjin Chen
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Sunwang Xu
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Fujian Provincial Key Laboratory of Precision Medicine for Cancer, Fuzhou, China.
| |
Collapse
|
18
|
Zhao YX, Zhao HP, Zhao MY, Yu Y, Qi X, Wang JH, Lv J. Latest insights into the global epidemiological features, screening, early diagnosis and prognosis prediction of esophageal squamous cell carcinoma. World J Gastroenterol 2024; 30:2638-2656. [PMID: 38855150 PMCID: PMC11154680 DOI: 10.3748/wjg.v30.i20.2638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/27/2024] Open
Abstract
As a highly invasive carcinoma, esophageal cancer (EC) was the eighth most prevalent malignancy and the sixth leading cause of cancer-related death worldwide in 2020. Esophageal squamous cell carcinoma (ESCC) is the major histological subtype of EC, and its incidence and mortality rates are decreasing globally. Due to the lack of specific early symptoms, ESCC patients are usually diagnosed with advanced-stage disease with a poor prognosis, and the incidence and mortality rates are still high in many countries, especially in China. Therefore, enormous challenges still exist in the management of ESCC, and novel strategies are urgently needed to further decrease the incidence and mortality rates of ESCC. Although the key molecular mechanisms underlying ESCC pathogenesis have not been fully elucidated, certain promising biomarkers are being investigated to facilitate clinical decision-making. With the advent and advancement of high-throughput technologies, such as genomics, proteomics and metabolomics, valuable biomarkers with high sensitivity, specificity and stability could be identified for ESCC. Herein, we aimed to determine the epidemiological features of ESCC in different regions of the world, especially in China, and focused on novel molecular biomarkers associated with ESCC screening, early diagnosis and prognosis prediction.
Collapse
Affiliation(s)
- Yi-Xin Zhao
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - He-Ping Zhao
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - Meng-Yao Zhao
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - Yan Yu
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - Xi Qi
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - Ji-Han Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, Shaanxi Province, China
| | - Jing Lv
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| |
Collapse
|
19
|
Xu J, Jiang W, Hu T, Long Y, Shen Y. NEDD4 and NEDD4L: Ubiquitin Ligases Closely Related to Digestive Diseases. Biomolecules 2024; 14:577. [PMID: 38785984 PMCID: PMC11117611 DOI: 10.3390/biom14050577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Protein ubiquitination is an enzymatic cascade reaction and serves as an important protein post-translational modification (PTM) that is involved in the vast majority of cellular life activities. The key enzyme in the ubiquitination process is E3 ubiquitin ligase (E3), which catalyzes the binding of ubiquitin (Ub) to the protein substrate and influences substrate specificity. In recent years, the relationship between the subfamily of neuron-expressed developmental downregulation 4 (NEDD4), which belongs to the E3 ligase system, and digestive diseases has drawn widespread attention. Numerous studies have shown that NEDD4 and NEDD4L of the NEDD4 family can regulate the digestive function, as well as a series of related physiological and pathological processes, by controlling the subsequent degradation of proteins such as PTEN, c-Myc, and P21, along with substrate ubiquitination. In this article, we reviewed the appropriate functions of NEDD4 and NEDD4L in digestive diseases including cell proliferation, invasion, metastasis, chemotherapeutic drug resistance, and multiple signaling pathways, based on the currently available research evidence for the purpose of providing new ideas for the prevention and treatment of digestive diseases.
Collapse
Affiliation(s)
| | | | | | | | - Yueming Shen
- Department of Digestive Diseases, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, 161 Shaoshan Road, Changsha 410000, China; (J.X.); (W.J.); (T.H.); (Y.L.)
| |
Collapse
|
20
|
Tan Z, Luan S, Wang X, Jiao W, Jiang P. Mechanism study of lncRNA RMRP regulating esophageal squamous cell carcinoma through miR-580-3p/ATP13A3 axis. Discov Oncol 2024; 15:150. [PMID: 38722543 PMCID: PMC11082096 DOI: 10.1007/s12672-024-00990-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
OBJECTIVE It is well-known that lncRNAs regulate energy metabolism in tumors. This study focused on the action of RMRP on esophageal squamous cell carcinoma (ESCC) cell proliferation, apoptosis, and glycolysis. METHODS In the resected ESCC tissues and adjacent tissues from patients, RMRP/miR-580-3p/ATP13A3 expressions were evaluated. ESCC cell proliferation rates and apoptotic rates were measured by CCK-8 and flow cytometry, respectively. Apoptosis related markers were examined by Western blot. Moreover, glucose uptake, lactic acid, and ATP were measured by commercial kits, whereas HK2 and PKM2 were evaluated by Western blot to study ESCC cell glycolysis. Finally, the editing program of RMRP/miR-580-3p/ATP13A3 was translated by luciferase reporter assay and RIP analysis. RESULTS RMRP and ATP13A3 were induced, while miR-580-3p was reduced in their expression in ESCC tissues. Silencing RMRP reduced proliferation, glycolysis, and anti-apoptosis ability of ESCC cells. RMRP sequestered miR-580-3p to target ATP13A3. Silenced ATP13A3 or overexpressed miR-580-3p rescued overexpressed RMRP-mediated promotion of proliferation, glycolysis, and anti-apoptosis of ESCC cells. CONCLUSION RMRP accelerates ESCC progression through the miR-580-3p/ATP13A3 axis, renewing a reference for lncRNA-based therapies for tumors.
Collapse
Affiliation(s)
- ZiRui Tan
- The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Chang'an District, Shijiazhuang City, 050000, Hebei Province, China
| | - ShengJie Luan
- Department of Tumor Chemoradiotherapy, Central Hospital of Qinghe County, Xingtai City, 054800, Hebei Province, China
| | - XiaoPeng Wang
- Department of Tumor Chemoradiotherapy, Central Hospital of Qinghe County, Xingtai City, 054800, Hebei Province, China
| | - WenPeng Jiao
- The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Chang'an District, Shijiazhuang City, 050000, Hebei Province, China
| | - Pu Jiang
- The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Chang'an District, Shijiazhuang City, 050000, Hebei Province, China.
| |
Collapse
|
21
|
Lin C, Tian Q, Guo S, Xie D, Cai Y, Wang Z, Chu H, Qiu S, Tang S, Zhang A. Metabolomics for Clinical Biomarker Discovery and Therapeutic Target Identification. Molecules 2024; 29:2198. [PMID: 38792060 PMCID: PMC11124072 DOI: 10.3390/molecules29102198] [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: 03/13/2024] [Revised: 04/10/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
As links between genotype and phenotype, small-molecule metabolites are attractive biomarkers for disease diagnosis, prognosis, classification, drug screening and treatment, insight into understanding disease pathology and identifying potential targets. Metabolomics technology is crucial for discovering targets of small-molecule metabolites involved in disease phenotype. Mass spectrometry-based metabolomics has implemented in applications in various fields including target discovery, explanation of disease mechanisms and compound screening. It is used to analyze the physiological or pathological states of the organism by investigating the changes in endogenous small-molecule metabolites and associated metabolism from complex metabolic pathways in biological samples. The present review provides a critical update of high-throughput functional metabolomics techniques and diverse applications, and recommends the use of mass spectrometry-based metabolomics for discovering small-molecule metabolite signatures that provide valuable insights into metabolic targets. We also recommend using mass spectrometry-based metabolomics as a powerful tool for identifying and understanding metabolic patterns, metabolic targets and for efficacy evaluation of herbal medicine.
Collapse
Affiliation(s)
- Chunsheng Lin
- Graduate School and Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.L.); (S.G.); (Y.C.); (Z.W.)
| | - Qianqian Tian
- Faculty of Social Sciences, The University of Hong Kong, Hong Kong 999077, China;
| | - Sifan Guo
- Graduate School and Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.L.); (S.G.); (Y.C.); (Z.W.)
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Dandan Xie
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Ying Cai
- Graduate School and Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.L.); (S.G.); (Y.C.); (Z.W.)
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Zhibo Wang
- Graduate School and Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.L.); (S.G.); (Y.C.); (Z.W.)
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Hang Chu
- Department of Biomedical Sciences, Beijing City University, Beijing 100193, China;
| | - Shi Qiu
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Songqi Tang
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| | - Aihua Zhang
- Graduate School and Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.L.); (S.G.); (Y.C.); (Z.W.)
- International Advanced Functional Omics Platform, Scientific Experiment Center, International Joint Research Center on Traditional Chinese and Modern Medicine, Hainan Engineering Research Center for Biological Sample Resources of Major Diseases (First Affiliated Hospital of Hainan Medical University), Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; (D.X.); (S.Q.); (S.T.)
| |
Collapse
|
22
|
Kaźmierczak-Siedlecka K, Muszyński D, Styburski D, Makarewicz J, Sobocki BK, Ulasiński P, Połom K, Stachowska E, Skonieczna-Żydecka K, Kalinowski L. Untargeted metabolomics in gastric and colorectal cancer patients - preliminary results. Front Cell Infect Microbiol 2024; 14:1394038. [PMID: 38774628 PMCID: PMC11106370 DOI: 10.3389/fcimb.2024.1394038] [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: 02/29/2024] [Accepted: 04/08/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction Recent years, microbiota-associated aspects have been analysed in multiple disorders regarding cancers. Existing evidence pints that gut microorganisms might take part in tumour origin and therapy efficacy. Nevertheless, to date, data on faecal metabolomics in cancer patients is still strongly limited. Therefore, we aimed to analyse gut untargeted metabolome in gastrointestinal cancer patients (i.e., gastric and colorectal cancer). Patients and methods There were 12 patients with either gastric (n=4) or colorectal cancer (n=8) enrolled and 8 analysed (n=4 each). Stool samples were collected prior to anti-cancer treatments. Untargeted metabolomics analyses were conducted by means of mass spectrometry. Results A plethora of metabolites in cancer patients we analysed were noted, with higher homogenity in case of gastric cancer patients. We found that the level of Deoxyguanosine,m/z 266.091,[M-H]-, Uridine,m/z 245.075,[M+H]+, Deoxyguanosine,m/z 268.104,[M]+, 3-Indoleacetic acid,m/z 176.07,[M+H]+, Indoxyl,m/z 132.031,[M-H]-, L-Phenylalanine,m/z 164.073,[M-H]-, L-Methionine,m/z 150.058,[M+NH4]+, was significantly higher in colorectal cancer patients and Ethyl hydrogen malonate,m/z 133.031,[M+H]+ in gastric cancer. Conclusion The overall insights into untargeted metabolomics showed that most often higher levels of analysed metabolites were detected in colorectal cancer patients compared to gastric cancer patients. The link between gut metabolome and both local and distal metastasis might exist, however it requires confirmation in further multi-centre studies regarding larger sample size.
Collapse
Affiliation(s)
- Karolina Kaźmierczak-Siedlecka
- Department of Medical Laboratory Diagnostics – Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdansk, Poland
| | - Damian Muszyński
- Scientific Circle of Studies Regarding Personalized Medicine Associated with Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
| | | | - Jakub Makarewicz
- Scientific Circle of Studies Regarding Personalized Medicine Associated with Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
| | - Bartosz Kamil Sobocki
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdansk, Poland
| | - Paweł Ulasiński
- Unit of Surgery with Unit of Surgery with Unit of Oncological Surgery, Specialist Hospital in Koscierzyna, Koscierzyna, Poland
| | - Karol Połom
- Academy of Medical and Social Applied Sciences, Elbląg, Poland
- Department of Surgical Oncology, Medical University of Gdansk, Gdansk, Poland
- Department of Gastrointestinal Surgical Oncology, Greater Poland Cancer Centre, Poznan, Poland
| | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics – Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdansk, Poland
- BioTechMed Centre/Department of Mechanics of Materials and Structures, Gdansk University of Technology, Gdansk, Poland
| |
Collapse
|
23
|
Jin X, Liu L, Liu D, Wu J, Wang C, Wang S, Wang F, Yu G, Jin X, Xue YW, Jiang D, Ni Y, Yang X, Wang MS, Wang ZW, Orlov YL, Jia W, Melino G, Liu JB, Chen WL. Unveiling the methionine cycle: a key metabolic signature and NR4A2 as a methionine-responsive oncogene in esophageal squamous cell carcinoma. Cell Death Differ 2024; 31:558-573. [PMID: 38570607 PMCID: PMC11094133 DOI: 10.1038/s41418-024-01285-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 04/05/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a deadly malignancy with notable metabolic reprogramming, yet the pivotal metabolic feature driving ESCC progression remains elusive. Here, we show that methionine cycle exhibits robust activation in ESCC and is reversely associated with patient survival. ESCC cells readily harness exogenous methionine to generate S-adenosyl-methionine (SAM), thus promoting cell proliferation. Mechanistically, methionine augments METTL3-mediated RNA m6A methylation through SAM and revises gene expression. Integrative omics analysis highlights the potent influence of methionine/SAM on NR4A2 expression in a tumor-specific manner, mediated by the IGF2BP2-dependent stabilization of methylated NR4A2 mRNA. We demonstrate that NR4A2 facilitates ESCC growth and negatively impacts patient survival. We further identify celecoxib as an effective inhibitor of NR4A2, offering promise as a new anti-ESCC agent. In summary, our findings underscore the active methionine cycle as a critical metabolic characteristic in ESCC, and pinpoint NR4A2 as a novel methionine-responsive oncogene, thereby presenting a compelling target potentially superior to methionine restriction.
Collapse
Affiliation(s)
- Xing Jin
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai, 200032, China
| | - Lei Liu
- Department of Thoracic Surgery, The Affiliated Tumor Hospital of Nantong University, Nantong, 226300, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Dan Liu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai, 200032, China
| | - Jia Wu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai, 200032, China
| | - Congcong Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai, 200032, China
| | - Siliang Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai, 200032, China
| | - Fengying Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai, 200032, China
| | - Guanzhen Yu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Laboratory of Digital Health and Artificial Intelligence, Zhejiang Digital Content Research Institute, Shaoxing, 312000, China
| | - Xiaoxia Jin
- Department of Pathology, The Affiliated Tumor Hospital of Nantong University, Nantong, 226300, China
| | - Yu-Wen Xue
- Pathology department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Dan Jiang
- Pathology department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yan Ni
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310029, China
| | - Xi Yang
- Department of Oncology, Shanxi Provincial Hospital of Traditional Chinese Medicine, Shanxi, 030001, China
| | - Ming-Song Wang
- Department of Thoracic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhi-Wei Wang
- Department of Breast, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yuriy L Orlov
- The Digital Health Institute, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119991, Russia
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090, Novosibirsk, Russia
- Life Sciences Department, Novosibirsk State University, Novosibirsk, 630090, Russia
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, 690922, Russia
- Agrarian and Technological Institute, Peoples' Friendship University of Russia, Moscow, 117198, Russia
| | - Wei Jia
- Department of Pharmacology and Pharmacy, Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Ji-Bin Liu
- Cancer Institute, The Affiliated Tumor Hospital of Nantong University, Nantong, 226361, China
| | - Wen-Lian Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- Shanghai Frontiers Science Center of Disease and Syndrome Biology of Inflammatory Cancer Transformation, Shanghai, 200032, China.
| |
Collapse
|
24
|
Yang H, Shi Y, Lin A, Qi C, Liu Z, Cheng Q, Miao K, Zhang J, Luo P. PESSA: A web tool for pathway enrichment score-based survival analysis in cancer. PLoS Comput Biol 2024; 20:e1012024. [PMID: 38717988 PMCID: PMC11078417 DOI: 10.1371/journal.pcbi.1012024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 03/26/2024] [Indexed: 05/12/2024] Open
Abstract
The activation levels of biologically significant gene sets are emerging tumor molecular markers and play an irreplaceable role in the tumor research field; however, web-based tools for prognostic analyses using it as a tumor molecular marker remain scarce. We developed a web-based tool PESSA for survival analysis using gene set activation levels. All data analyses were implemented via R. Activation levels of The Molecular Signatures Database (MSigDB) gene sets were assessed using the single sample gene set enrichment analysis (ssGSEA) method based on data from the Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA), The European Genome-phenome Archive (EGA) and supplementary tables of articles. PESSA was used to perform median and optimal cut-off dichotomous grouping of ssGSEA scores for each dataset, relying on the survival and survminer packages for survival analysis and visualisation. PESSA is an open-access web tool for visualizing the results of tumor prognostic analyses using gene set activation levels. A total of 238 datasets from the GEO, TCGA, EGA, and supplementary tables of articles; covering 51 cancer types and 13 survival outcome types; and 13,434 tumor-related gene sets are obtained from MSigDB for pre-grouping. Users can obtain the results, including Kaplan-Meier analyses based on the median and optimal cut-off values and accompanying visualization plots and the Cox regression analyses of dichotomous and continuous variables, by selecting the gene set markers of interest. PESSA (https://smuonco.shinyapps.io/PESSA/ OR http://robinl-lab.com/PESSA) is a large-scale web-based tumor survival analysis tool covering a large amount of data that creatively uses predefined gene set activation levels as molecular markers of tumors.
Collapse
Affiliation(s)
- Hong Yang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Haizhu District, Guangzhou, Guangdong, China
- The First School of Clinical Medicine, Southern Medical University, Baiyun District, Guangzhou, Guangdong, China
| | - Ying Shi
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Haizhu District, Guangzhou, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Baiyun District, Guangzhou, Guangdong, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Haizhu District, Guangzhou, Guangdong, China
| | - Chang Qi
- Institute of Logic and Computation, TU Wien, Austria
| | - Zaoqu Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Kai Miao
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Haizhu District, Guangzhou, Guangdong, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Haizhu District, Guangzhou, Guangdong, China
| |
Collapse
|
25
|
Cicalini I, Chiarelli AM, Chiacchiaretta P, Perpetuini D, Rosa C, Mastrodicasa D, d'Annibale M, Trebeschi S, Serafini FL, Cocco G, Narciso M, Corvino A, Cinalli S, Genovesi D, Lanuti P, Valentinuzzi S, Pieragostino D, Brocco D, Beets-Tan RGH, Tinari N, Sensi SL, Stuppia L, Del Boccio P, Caulo M, Delli Pizzi A. Multi-omics staging of locally advanced rectal cancer predicts treatment response: a pilot study. LA RADIOLOGIA MEDICA 2024; 129:712-726. [PMID: 38538828 PMCID: PMC11088547 DOI: 10.1007/s11547-024-01811-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 03/13/2024] [Indexed: 05/12/2024]
Abstract
Treatment response assessment of rectal cancer patients is a critical component of personalized cancer care and it allows to identify suitable candidates for organ-preserving strategies. This pilot study employed a novel multi-omics approach combining MRI-based radiomic features and untargeted metabolomics to infer treatment response at staging. The metabolic signature highlighted how tumor cell viability is predictively down-regulated, while the response to oxidative stress was up-regulated in responder patients, showing significantly reduced oxoproline values at baseline compared to non-responder patients (p-value < 10-4). Tumors with a high degree of texture homogeneity, as assessed by radiomics, were more likely to achieve a major pathological response (p-value < 10-3). A machine learning classifier was implemented to summarize the multi-omics information and discriminate responders and non-responders. Combining all available radiomic and metabolomic features, the classifier delivered an AUC of 0.864 (± 0.083, p-value < 10-3) with a best-point sensitivity of 90.9% and a specificity of 81.8%. Our results suggest that a multi-omics approach, integrating radiomics and metabolomic data, can enhance the predictive value of standard MRI and could help to avoid unnecessary surgical treatments and their associated long-term complications.
Collapse
Affiliation(s)
- Ilaria Cicalini
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Odontoiatry, "G. d'Annunzio" University, Chieti, Italy
| | - Antonio Maria Chiarelli
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Piero Chiacchiaretta
- Department of Innovative Technologies in Medicine and Odontoiatry, "G. d'Annunzio" University, Chieti, Italy.
| | - David Perpetuini
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Consuelo Rosa
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
| | | | - Martina d'Annibale
- Department of Radiology, SS. Annunziata Hospital, "G. d'Annunzio" University, Via dei Vestini, 66100, ChietiChieti, Italy
| | - Stefano Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Giulio Cocco
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
- Unit of Ultrasound in Internal Medicine, Department of Medicine and Science of Aging, "G. D'Annunzio" University, Chieti, Italy
| | - Marco Narciso
- Department of Radiology, SS. Annunziata Hospital, "G. d'Annunzio" University, Via dei Vestini, 66100, ChietiChieti, Italy
| | - Antonio Corvino
- Medical, Movement and Wellbeing Sciences Department, Via Medina 40, 80133, Naples, Italy
| | - Sebastiano Cinalli
- Division of Pathology, ASST of Valtellina and Alto Lario, Sondrio, Italy
| | - Domenico Genovesi
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, "G. D'Annunzio" University of Chieti, Via dei Vestini, 66100, Chieti, Italy
| | - Paola Lanuti
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Science, "G. D'Annunzio" University of Chieti, Via dei Vestini, 66100, Chieti, Italy
| | - Silvia Valentinuzzi
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Pharmacy, "G. D'Annunzio" University of Chieti, Via dei Vestini, 66100, Chieti, Italy
| | - Damiana Pieragostino
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Odontoiatry, "G. d'Annunzio" University, Chieti, Italy
| | - Davide Brocco
- Clinical Oncology Unit, SS. Annunziata Hospital, Via dei Vestini, 66100, Chieti, Italy
| | - Regina G H Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Nicola Tinari
- Department of Medicine and Aging Science, "G. D'Annunzio" University of Chieti, Via dei Vestini, 66100, Chieti, Italy
| | - Stefano L Sensi
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Psychological, Health and Territory Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Piero Del Boccio
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Pharmacy, "G. D'Annunzio" University of Chieti, Via dei Vestini, 66100, Chieti, Italy
| | - Massimo Caulo
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
- Department of Radiology, SS. Annunziata Hospital, "G. d'Annunzio" University, Via dei Vestini, 66100, ChietiChieti, Italy
| | - Andrea Delli Pizzi
- Department of Innovative Technologies in Medicine and Odontoiatry, "G. d'Annunzio" University, Chieti, Italy
- Department of Radiology, SS. Annunziata Hospital, "G. d'Annunzio" University, Via dei Vestini, 66100, ChietiChieti, Italy
| |
Collapse
|
26
|
Sun M, Shen W, Guo X, Liao Y, Huang Y, Hu M, Ye P, Liu R. A critical review of advances in tumor metabolism abnormalities induced by nitrosamine disinfection by-products in drinking water. Toxicol Sci 2024; 199:12-28. [PMID: 38291902 DOI: 10.1093/toxsci/kfae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024] Open
Abstract
Intensified sanitation practices amid the recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak might result in the increased release of chloramine disinfectants into surface water, significantly promoting the formation of nitrosamine disinfection by-products (DBPs) in drinking water. Unfortunately, these nitrosamine DBPs exhibit significant genotoxic, carcinogenic, and mutagenic properties, whereas chlorinating disinfectants remain in global practice. The current review provides valuable insights into the occurrence, identification, contamination status, exposure limits, and toxicity of the new unregulated disinfection by-products (nitrosamine DBPs) in drinking water. As a result, concentrations of nitrosamine DBPs far exceed allowable limits in drinking water, and prolonged exposure has the potential to cause metabolic disorders, a critical step in tumor initiation and progression. Importantly, based on recent research, we have concluded the role of nitrosamines DBPs in different metabolic pathways. Remarkably, nitrosamine DBPs can induce chronic inflammation and initiate tumors by activating sphingolipid and polyunsaturated fatty acid metabolism. Regarding amino acid and nucleotide metabolism, nitrosamine DBPs can inhibit tryptophan metabolism and de novo nucleotide synthesis. Moreover, inhibition of de novo nucleotide synthesis fails to repair DNA damage induced by nitrosamines. Additionally, the accumulation of lactate induced by nitrosamine DBPs may act as a pivotal signaling molecule in communication within the tumor microenvironment. However, with the advancement of tumor metabolomics, understanding the role of nitrosamine DBPs in causing cancer by inducing metabolic abnormalities significantly lags behind, and specific mechanisms of toxic effects are not clearly defined. Urgently, further studies exploring this promising area are needed.
Collapse
Affiliation(s)
- Mingjun Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, P. R. China
| | - Weitao Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, P. R. China
| | - Xinxin Guo
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, P. R. China
| | - Yinghao Liao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, P. R. China
| | - Yang Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, P. R. China
| | - Mohan Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, P. R. China
| | - Ping Ye
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, P. R. China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, P. R. China
| |
Collapse
|
27
|
Chen M, Zhang C, Li H, Zheng S, Li Y, Yuan M, Chen Y, Wu J, Sun Q. PLA2G4A and ACHE modulate lipid profiles via glycerophospholipid metabolism in platinum-resistant gastric cancer. J Transl Med 2024; 22:249. [PMID: 38454407 PMCID: PMC10921739 DOI: 10.1186/s12967-024-05055-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/01/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Bioactive lipids involved in the progression of various diseases. Nevertheless, there is still a lack of biomarkers and relative regulatory targets. The lipidomic analysis of the samples from platinum-resistant in gastric cancer patients is expected to help us further improve our understanding of it. METHODS We employed LC-MS based untargeted lipidomic analysis to search for potential candidate biomarkers for platinum resistance in GC patients. Partial least squares discriminant analysis (PLS-DA) and variable importance in projection (VIP) analysis were used to identify differential lipids. The possible molecular mechanisms and targets were obtained by metabolite set enrichment analysis and potential gene network screened. Finally, verified them by immunohistochemical of a tissue microarray. RESULTS There were 71 differential lipid metabolites identified in GC samples between the chemotherapy-sensitivity group and the chemotherapy resistance group. According to Foldchange (FC) value, VIP value, P values (FC > 2, VIP > 1.5, p < 0.05), a total of 15 potential biomarkers were obtained, including MGDG(43:11)-H, Cer(d18:1/24:0) + HCOO, PI(18:0/18:1)-H, PE(16:1/18:1)-H, PE(36:2) + H, PE(34:2p)-H, Cer(d18:1 + hO/24:0) + HCOO, Cer(d18:1/23:0) + HCOO, PC(34:2e) + H, SM(d34:0) + H, LPC(18:2) + HCOO, PI(18:1/22:5)-H, PG(18:1/18:1)-H, Cer(d18:1/24:0) + H and PC(35:2) + H. Furthermore, we obtained five potential key targets (PLA2G4A, PLA2G3, DGKA, ACHE, and CHKA), and a metabolite-reaction-enzyme-gene interaction network was built to reveal the biological process of how they could disorder the endogenous lipid profile of platinum resistance in GC patients through the glycerophospholipid metabolism pathway. Finally, we further identified PLA2G4A and ACHE as core targets of the process by correlation analysis and tissue microarray immunohistochemical verification. CONCLUSION PLA2G4A and ACHE regulated endogenous lipid profile in the platinum resistance in GC patients through the glycerophospholipid metabolism pathway. The screening of lipid biomarkers will facilitate earlier precision medicine interventions for chemotherapy-resistant gastric cancer. The development of therapies targeting PLA2G4A and ACHE could enhance platinum chemotherapy effectiveness.
Collapse
Affiliation(s)
- Menglin Chen
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Cancan Zhang
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Huaizhi Li
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Shanshan Zheng
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Yaqi Li
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Mengyun Yuan
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Yuxuan Chen
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jian Wu
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, Jiangsu, China.
| | - Qingmin Sun
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
28
|
Yang J, Xu S, Cheng J, Yin X, Yan D, Li X. CXCL10 and its receptor in patients with chronic hepatitis B and their ability to predict HBeAg seroconversion during antiviral treatment with TDF. J Med Virol 2024; 96:e29516. [PMID: 38469895 DOI: 10.1002/jmv.29516] [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: 08/19/2023] [Revised: 12/19/2023] [Accepted: 02/27/2024] [Indexed: 03/13/2024]
Abstract
The serum chemokine C-X-C motif ligand-10 (CXCL10) and its unique receptor (CXCR3) may predict the prognosis of patients with chronic hepatitis B (CHB) treated with tenofovir disoproxil fumarate (TDF). Nevertheless, there are few reports on the profile of CXCL10 and CXCR3 and their clinical application in HBeAg (+) CHB patients during TDF antiviral therapy. CXCL10 and CXCR3 were determined in 118 CHB patients naively treated with TDF for at least 96 weeks at baseline and at treatment weeks 12 and 24. In addition, gene set enrichment analysis was used to examine the associated dataset from Gene Expression Omnibus and explore the gene sets associated with HBeAg seroconversion (SC). The change of CXCL10 (ΔCXCL10, baseline to 48-week TDF treatment) and CXCR3 (ΔCXCR3) is closely related to the possibility of HBeAg SC of CHB patients under TDF treatment. Immunohistochemical analysis of CXCL10/CXCR3 protein in liver tissue shows that there is a significant difference between paired liver biopsy samples taken before and after 96 weeks of successful TDF treatment of CHB patients (11 pairs) but no significance for unsuccessful TDF treatment (14 pairs). Multivariate Cox analysis suggests that the ΔCXCL10 is an independent predictive indicator of HBeAg SC, and the area under the receiver operating characteristic curve of the ΔCXCL10 in CHB patients is 0.8867 (p < 0.0001). Our results suggest that a lower descending CXCL10 level is associated with an increased probability of HBeAg SC of CHB patients during TDF therapy. Moreover, liver tissue CXCL10 might be involved in the immunological process of HBeAg SC.
Collapse
Affiliation(s)
- Jiezuan Yang
- The First Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Shaoyan Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jinlin Cheng
- The First Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Xuying Yin
- The First Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Dong Yan
- The First Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Hangzhou, China
| | - Xuefen Li
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
29
|
Bläsius K, Ludwig L, Knapp S, Flaßhove C, Sonnabend F, Keller D, Tacken N, Gao X, Kahveci-Türköz S, Grannemann C, Babendreyer A, Adrain C, Huth S, Baron JM, Ludwig A, Düsterhöft S. Pathological mutations reveal the key role of the cytosolic iRhom2 N-terminus for phosphorylation-independent 14-3-3 interaction and ADAM17 binding, stability, and activity. Cell Mol Life Sci 2024; 81:102. [PMID: 38409522 PMCID: PMC10896983 DOI: 10.1007/s00018-024-05132-3] [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: 11/29/2023] [Accepted: 01/15/2024] [Indexed: 02/28/2024]
Abstract
The protease ADAM17 plays an important role in inflammation and cancer and is regulated by iRhom2. Mutations in the cytosolic N-terminus of human iRhom2 cause tylosis with oesophageal cancer (TOC). In mice, partial deletion of the N-terminus results in a curly hair phenotype (cub). These pathological consequences are consistent with our findings that iRhom2 is highly expressed in keratinocytes and in oesophageal cancer. Cub and TOC are associated with hyperactivation of ADAM17-dependent EGFR signalling. However, the underlying molecular mechanisms are not understood. We have identified a non-canonical, phosphorylation-independent 14-3-3 interaction site that encompasses all known TOC mutations. Disruption of this site dysregulates ADAM17 activity. The larger cub deletion also includes the TOC site and thus also dysregulated ADAM17 activity. The cub deletion, but not the TOC mutation, also causes severe reductions in stimulated shedding, binding, and stability of ADAM17, demonstrating the presence of additional regulatory sites in the N-terminus of iRhom2. Overall, this study contrasts the TOC and cub mutations, illustrates their different molecular consequences, and reveals important key functions of the iRhom2 N-terminus in regulating ADAM17.
Collapse
Affiliation(s)
- Katharina Bläsius
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Lena Ludwig
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Sarah Knapp
- Institute of Biochemistry and Molecular Biology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Charlotte Flaßhove
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Friederike Sonnabend
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Diandra Keller
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Nikola Tacken
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Xintong Gao
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Selcan Kahveci-Türköz
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Caroline Grannemann
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Colin Adrain
- Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, Northern Ireland
| | - Sebastian Huth
- Department of Dermatology and Allergology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Jens Malte Baron
- Department of Dermatology and Allergology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
30
|
Lin W, Si X, Zhao Z, Chen F, Xu J, Huang W, Lin J, Chen Z, Huang Z. Applying Untargeted Lipidomics to Evaluate the Efficacy of Combined Neoadjuvant Chemotherapy and Immunotherapy for Esophageal Squamous Carcinoma Treatment. J Proteome Res 2024; 23:663-672. [PMID: 38175711 DOI: 10.1021/acs.jproteome.3c00527] [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] [Indexed: 01/05/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) is an aggressive malignant tumor with a poor prognosis due to insidious symptoms that make early diagnosis difficult. Despite the combination of multiple treatment modalities, the recurrence and mortality rates of ESCC remain high. Neoadjuvant chemotherapy combined with immunotherapy is an emerging treatment modality that improves the prognosis of patients with ESCC. However, owing to the presence of hyperprogression and pseudoprogression, the currently used methods cannot accurately evaluate the efficacy of this therapy in patients, thus creating an evaluation bias and depriving these patients of the opportunity to benefit. We used untargeted lipidomics to identify the differences in lipid composition between cancer specimens and normal tissue specimens in the neoadjuvant chemotherapy combined with the immunotherapy group and the surgery-alone group of esophageal cancer patients and constructed a prediction model based on sphingomyelin 12:1;2O/30:0 and triglyceride (TG) 60:3 | TG 18:0_24:1_18 using a machine learning approach, which helps to better evaluate the neoadjuvant efficacy of combination therapy and better guide the treatment of ESCC.
Collapse
Affiliation(s)
- Weijie Lin
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Xianzhe Si
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Zhihuang Zhao
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Feng Chen
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Jie Xu
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Wenbo Huang
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Jianqing Lin
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Zhiyao Chen
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Zhijun Huang
- Department of Gastrointestinal and Esophageal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| |
Collapse
|
31
|
Zhu M, Zhu Z, Zhang N, Ma J, Huang N, He S, Lu X. Identification of miRNA, lncRNA and circRNA associated with gastric cancer metabolism through sequencing and bioinformatics analysis. Pathol Res Pract 2024; 254:155151. [PMID: 38290402 DOI: 10.1016/j.prp.2024.155151] [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: 10/20/2023] [Revised: 12/09/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
Gastric cancer remains a highly prevalent malignancy worldwide with its molecular features poorly understood. To gain full insight into its genetic landscape, we performed whole-transcriptome sequencing on human tumors and adjacent non-tumors to predict the function of microRNA, long coding RNA, circular RNA, and mRNA, as well as estimate their correlation with gastric cancer characteristics through construction of ceRNA, WGCNA and PPI network. Functional enrichment analysis annotated nucleic acid binding, enzyme activity and binding related to differentially expressed miRNAs (dif-miRNAs); energy binding and enzyme binding related to dif-lncRNAs; protein binding and enzyme activity related to dif-circRNAs; protein digestion and absorption related to dif-mRNAs. The expression of key miR-135a-5p, lncRNAs-MSTRG.48856.1, ENST00000569981, MSTRG.22826.1, ENST00000564492, circRNAs-CCSER2, FNDC3B, CORO1C, FAM214A were validated by real-time PCR. The ceRNA network filtered 14 miRNAs, 30 lncRNAs, and 6 mRNA in the lncRNA-ceRNA axis and 8 miRNAs, 9 circRNAs, and 3 mRNA in the circRNA-ceRNA axis. Genes involved in ceRNA were annotated to be closely related to tumor material synthesis and metabolism. The WGCNA network filtered gene clusters related to TNM traits and extracted the hub genes CLDN10, CD177, newGene_35523, newGene_51201, CEACAM7, and newGene_46634. These genes were associated with cell proliferation, metabolism, and enzyme activity regulation. The PPI network analyzed the stable interaction relationships of the hub genes. Our research provides a valuable resource for understanding the molecular mechanisms of gastric cancer from the perspective of tumor metabolism.
Collapse
Affiliation(s)
- Meng Zhu
- College of Basic Medicine, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Zenghui Zhu
- College of Basic Medicine, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Ning Zhang
- Department of Pathology, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia 750004, China.
| | - Jingwei Ma
- The Second Department of Surgical Oncology, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Ningbo Huang
- College of Basic Medicine, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Shuixiang He
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi 710061, China
| | - Xinlan Lu
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
32
|
Jia XM, Hao H, Zhang Q, Yang MX, Wang N, Sun SL, Yang ZN, Jin YR, Wang J, Du YF. The bioavailability enhancement and insight into the action mechanism of poorly soluble natural compounds from co-crystals preparation: Oridonin as an example. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155179. [PMID: 37925890 DOI: 10.1016/j.phymed.2023.155179] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 10/04/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Natural bioactive molecules are important sources for the development of new drugs. However, most of them were limited in clinical applications due to their low aqueous solubility and bioavailability. Oridonin (ORI) is a powerful anticancer compound with above characteristics. OBJECTIVE This study aimed to find an effective method to improve the bioavailability of poorly soluble natural compounds, and explore the action mechanisms of them to promote their application. RESULTS In this study, ORI-nicotinamide (NCT) cocrystal was successfully prepared for the first time to overcome the defects of ORI. The solubility and oral bioavailability of cocrystal (COC) increased 1.34 and 1.18 times compared with ORI. Moreover, MTT assay was applied to compare the cytotoxicity of positive control drug sorafenib with ORI and COC. The IC50 values of sorafenib, ORI and COC on HepG2 cells were 7.61, 8.79 and 7.36 nmol·mL-1, which indicated that the cytotoxicity of ORI could be enhanced by cocrystal preparation. The cellular metabolomics was innovatively introduced to gain insight into the difference of cytotoxicity mechanism between ORI and COC. The results showed that there were 78 metabolites with significant differences in content between the two groups, while these differential metabolites were related to 11 metabolic pathways. Among these, glycerophospholipid metabolism and cysteine and methionine metabolism were the significant differential pathways, and the downregulation of PC(14:0/16:1(9z)) and upregulation of homocysteine were the likely main reasons for higher cytotoxicity of COC. CONCLUSIONS This study has presented novel approaches for enhancing the bioavailability and drug efficacy of natural compounds, while also offering fresh insights into the underlying action mechanisms of pharmaceutical cocrystals.
Collapse
Affiliation(s)
- Xin-Ming Jia
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei 050017, PR China
| | - Han Hao
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei 050017, PR China
| | - Qian Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei 050017, PR China
| | - Meng-Xin Yang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei 050017, PR China
| | - Nan Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei 050017, PR China
| | - Shi-Lin Sun
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei 050017, PR China
| | - Ze-Nan Yang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei 050017, PR China
| | - Yi-Ran Jin
- Department of Clinical Pharmacy, The Second Hospital of Hebei Medical University, 215 West Heping Road, Shijiazhuang, Hebei 050000, PR China.
| | - Jing Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei 050017, PR China.
| | - Ying-Feng Du
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei 050017, PR China.
| |
Collapse
|
33
|
Jiao R, Jiang W, Xu K, Luo Q, Wang L, Zhao C. Lipid metabolism analysis in esophageal cancer and associated drug discovery. J Pharm Anal 2024; 14:1-15. [PMID: 38352954 PMCID: PMC10859535 DOI: 10.1016/j.jpha.2023.08.019] [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: 04/03/2023] [Revised: 07/27/2023] [Accepted: 08/29/2023] [Indexed: 02/16/2024] Open
Abstract
Esophageal cancer is an upper gastrointestinal malignancy with a bleak prognosis. It is still being explored in depth due to its complex molecular mechanisms of occurrence and development. Lipids play a crucial role in cells by participating in energy supply, biofilm formation, and signal transduction processes, and lipid metabolic reprogramming also constitutes a significant characteristic of malignant tumors. More and more studies have found esophageal cancer has obvious lipid metabolism abnormalities throughout its beginning, progress, and treatment resistance. The inhibition of tumor growth and the enhancement of antitumor therapy efficacy can be achieved through the regulation of lipid metabolism. Therefore, we reviewed and analyzed the research results and latest findings for lipid metabolism and associated analysis techniques in esophageal cancer, and comprehensively proved the value of lipid metabolic reprogramming in the evolution and treatment resistance of esophageal cancer, as well as its significance in exploring potential therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Ruidi Jiao
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518000, China
| | - Wei Jiang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
| | - Kunpeng Xu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
| | - Qian Luo
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Luhua Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518000, China
| | - Chao Zhao
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
| |
Collapse
|
34
|
Zhou W, Guo S, Zhang J, Yan Y, Wu J, Liu X. An emerging biomarker for the diagnosis and treatment of esophageal squamous cell carcinoma - Aurora A. Comput Biol Med 2024; 168:107759. [PMID: 38043467 DOI: 10.1016/j.compbiomed.2023.107759] [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: 09/16/2023] [Revised: 10/27/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a prominent form of esophageal cancer. Aurora A (AURKA), an enzyme that phosphorylates serine and threonine, has a vital function in controlling the process of separating chromosomes during cell division. The contribution of this entity has been documented in the advancement of malignant proliferations, including tumors occurring in the breast, stomach, and ovaries. METHODS The potential molecular mechanism of AURKA is comprehensively examined through the analysis of bulk RNA-seq and single-cell RNA-seq data obtained from publicly available databases. This analysis encompasses various aspects such as expression levels, prognosis, and functional pathways, among others. RESULTS The upregulation of AURKA in ESCC has been found to be correlated with the overall survival of patients. The functional annotation and pathway enrichment analysis conducted in this study lead to the conclusion that AURKA participates in the regulation of a number of malignant processes connected to cell proliferation, such as cell cycle control, apoptosis, and the p53 signaling pathway. Additionally, AURKA has been found to be associated with drug sensitivity and has an impact on the infiltration of tumor-infiltrating immune cells in ESCC. CONCLUSIONS AURKA exhibits potential as a prognostic and therapeutic biomarker linked to the regulation of cell cycle and cell proliferation.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Siyu Guo
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jingyuan Zhang
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu Yan
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jiarui Wu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xiao Liu
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
35
|
Wu Z, Yu X, Zhang S, He Y, Guo W. Novel roles of PIWI proteins and PIWI-interacting RNAs in human health and diseases. Cell Commun Signal 2023; 21:343. [PMID: 38031146 PMCID: PMC10685540 DOI: 10.1186/s12964-023-01368-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Non-coding RNA has aroused great research interest recently, they play a wide range of biological functions, such as regulating cell cycle, cell proliferation, and intracellular substance metabolism. Piwi-interacting RNAs (piRNAs) are emerging small non-coding RNAs that are 24-31 nucleotides in length. Previous studies on piRNAs were mainly limited to evaluating the binding to the PIWI protein family to play the biological role. However, recent studies have shed more lights on piRNA functions; aberrant piRNAs play unique roles in many human diseases, including diverse lethal cancers. Therefore, understanding the mechanism of piRNAs expression and the specific functional roles of piRNAs in human diseases is crucial for developing its clinical applications. Presently, research on piRNAs mainly focuses on their cancer-specific functions but lacks investigation of their expressions and epigenetic modifications. This review discusses piRNA's biogenesis and functional roles and the recent progress of functions of piRNA/PIWI protein complexes in human diseases. Video Abstract.
Collapse
Affiliation(s)
- Zeyu Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Xiao Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Yuting He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China.
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China.
| |
Collapse
|
36
|
He L, Zhao C, Xu J, Li W, Lu Y, Gong Y, Gu D, Wang X, Guo F. A potential novel biomarker: comprehensive analysis of prognostic value and immune implication of CES3 in colonic adenocarcinoma. J Cancer Res Clin Oncol 2023; 149:13239-13255. [PMID: 37480527 DOI: 10.1007/s00432-023-05156-5] [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: 05/30/2023] [Accepted: 07/09/2023] [Indexed: 07/24/2023]
Abstract
PURPOSE Colon cancer is the most common malignant tumor in the intestine. Abnormal Carboxylesterases 3 (CES3) expression had been reported to be correlated to multiple tumor progression. However, the association among CES3 expression and prognostic value and immune effects in colonic adenocarcinoma (COAD) were unclear. PATIENTS AND METHODS The transcription and expression data of CES3 and corresponding clinical information was downloaded from The Cancer Genome Atlas (TCGA). The CES3 protein expression and the prognostic value were verified based on tissue microarray data. The Cancer immune group Atlas (TCIA), Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and the GSE78220 immunotherapy cohort were used to forecast immunotherapy efficacy. Finally, a prognostic immune signature was constructed and verified. RESULTS Compared with normal colon tissues, the expression of mRNA and protein levels of CES3 were downregulated in tumor tissues. CES3 expression was associated with TIICs. Hihg-CES3 COAD patients had better efficacy of concurrent immunotherapy. CES3-related immune genes (CRIs) were identified and were then used to construct prognostic immune signature and had been successfully verified in GES39582. CONCLUSION CES3 might be a potential immune-related gene and promising prognostic biomarker in COAD.
Collapse
Affiliation(s)
- Lulu He
- Department of Oncology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| | - Chenyi Zhao
- Department of Oncology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jingjing Xu
- Central Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenjing Li
- Department of Clinical Laboratory, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yujie Lu
- Department of Oncology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yang Gong
- Department of Oncology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Dingyi Gu
- Department of Oncology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| | - Xiaoyan Wang
- Department of Oncology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| | - Feng Guo
- Department of Oncology, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China.
| |
Collapse
|
37
|
Lei Y, Zhou B, Meng X, Liang M, Song W, Liang Y, Gao Y, Wang M. A risk score model based on lipid metabolism-related genes could predict response to immunotherapy and prognosis of lung adenocarcinoma: a multi-dataset study and cytological validation. Discov Oncol 2023; 14:188. [PMID: 37874388 PMCID: PMC10597940 DOI: 10.1007/s12672-023-00802-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/08/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Lipid metabolism is a key factor in tumorigenesis and drug resistance, and models related to lipid metabolism have shown potential to predict survival and curative effects of adjuvant therapy in various cancers. However, the relationship between lipid metabolism and prognosis and treatment response of lung adenocarcinoma (LUAD) are still unclear. METHODS We enrolled seven bulk RNA-sequence datasets (GSE37745, GSE19188, GSE30219, GSE31547, GSE41271, GSE42127, and GSE72094) from the GEO database and one single-cell RNA-sequencing dataset (GSE117570) from the TISCH2 database. Non-negative matrix factorization (NMF) was utilized to construct the risk score model based on lipid score calculated by GSVA algorithm. Phs000452.v3, PMID: 26359337, PMID: 32472114, PRJEB23709 datasets were used to test the response to immunotherapy. Drug sensitivity analysis was assessed according to the GDSC database, and immunotherapy response was evaluated using the Wilcoxon test. Cellular function assays including clone formation, EDU assays and flow cytometry were implemented to explore the phenotype alteration caused by the knockdown of PTDSS1, which is one of key gene in risk score model. RESULTS We analyzed both bulk and single-cell RNA sequencing data to establish and validate a risk score model based on 18 lipid metabolism-related genes with significant impact on prognosis. After divided the patients into two groups according to risk score, we identified differences in lipid-related metabolic processes and a detailed portrait of the immune landscapes of high- and low-risk groups. Moreover, we investigated the potentials of our risk score in predicting response to immunotherapy and drug sensitivity. In addition, we silenced PTDSS1 in LUAD cell lines, and found that the proliferation of the cells was weakened, and the apoptosis of the cells was increased. CONCLUSION Our study highlights the crucial roles of lipid metabolism in LUAD and provides a reliable risk score model, which can aid in predicting prognosis and response to immunotherapy. Furthermore, we investigated the roles of PTDSS1 in LUAD carcinogenesis, which showed that PTDSS1 regulated proliferation and apoptosis of LUAD cells.
Collapse
Affiliation(s)
- Yangyang Lei
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Boxuan Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangzhi Meng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weijian Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Minghui Wang
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
38
|
Gao X, Liu X, Wang Y, Wang T, Fang D, Hu K. Effects of Clostridium butyricum on Intestinal Microflora and Metabolism of Eriocheir sinensis. Int J Mol Sci 2023; 24:13784. [PMID: 37762084 PMCID: PMC10531170 DOI: 10.3390/ijms241813784] [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: 08/01/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Clostridium butyricum, a new probiotic in recent years, can produce butyric acid and short-chain fatty acids. It has the characteristics of strong acid and alkali resistance, high temperature resistance, and strong resistance to most antibiotics, and has more advantages than other probiotics. However, the action mechanism of C. butyricum on Eriocheir sinensis is still unclear and needs further study. In this study, when C. butyricum was added to the basic diet, the number of living bacteria was 0, 1 × 106 and 1 × 108 CFU/g, respectively. The E. sinensis were randomly divided into three groups: (blank control group, experimental group 1 (1 × 106 CFU/g) and experimental group 2 (1 × 108 CFU/g)). They were fed an experimental diet for 28 days. The effects of C. butyricum on E. sinensis were studied by detecting the differences in non-specific immune indexes, intestinal microflora, and metabolites between serum and hepatopancreas. The results showed that C. butyricum could improve the antioxidant ability of E. sinensis serum and hepatopancreas, protect intestinal tissues, and promote the absorption of nutrients. At the same time, it can enhance the microbial diversity and richness of the E. sinensis gut flora. LC-MS metabolomics was used to detect the metabolism of intestinal flora. It was found that C. butyricum could up-regulate lysophosphatidylcholine in the intestine. Through the KEGG enrichment pathway, it was found that significantly different metabolites were mainly concentrated in six metabolic pathways. The purine metabolism and alanine, aspartate, and glutamate metabolism pathways showed a downward trend, indicating that the addition of C. butyricum to feed could reduce purine metabolism, promote the water-salt balance of the organism's cells, and reduce inflammation. In this study, it was found that the addition of certain concentrations of C. butyricum to feed could improve the antioxidant ability of E. sinensis, improve the intestinal flora environment, and promote the growth of beneficial bacteria in the gut. This can promote the body's metabolism, which is more conducive to its growth.
Collapse
Affiliation(s)
- Xiaoning Gao
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| | - Xueting Liu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| | - Yali Wang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| | - Tianwei Wang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| | - Di Fang
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| | - Kun Hu
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
39
|
Chen N, Xu X, Fan Y. Immune checkpoint inhibitors in the treatment of oesophageal squamous cell carcinoma: where are we and where are we going? Ther Adv Med Oncol 2023; 15:17588359231189420. [PMID: 37547447 PMCID: PMC10399266 DOI: 10.1177/17588359231189420] [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: 02/14/2023] [Accepted: 07/05/2023] [Indexed: 08/08/2023] Open
Abstract
Oesophageal squamous cell carcinoma (ESCC) is a kind of malignant tumour with high invasiveness and a poor prognosis. Immunotherapy, especially immune checkpoint inhibitors (ICIs), is a rapidly growing therapeutic method that activates and enhances anti-tumour immunity to treat patients with malignancy. Several clinical trials have confirmed the efficacy of ICIs in the treatment of ESCC. ICIs have been approved for the treatment of patients with ESCC. However, only a subset of patients can obtain excellent benefits from ICI therapy. In recent years, there has been a growing interest in exploring predictive biomarkers of immunotherapy response. In this review, we highlighted the predictive biomarkers for the prognosis of ESCC patients treated with ICIs and pointed out the existing problems and the direction of future research in this field.
Collapse
Affiliation(s)
- Ning Chen
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiaoling Xu
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| | - Yun Fan
- Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
40
|
Perazzoli G, García-Valdeavero OM, Peña M, Prados J, Melguizo C, Jiménez-Luna C. Evaluating Metabolite-Based Biomarkers for Early Diagnosis of Pancreatic Cancer: A Systematic Review. Metabolites 2023; 13:872. [PMID: 37512579 PMCID: PMC10384620 DOI: 10.3390/metabo13070872] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/13/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers, with five-year survival rates around 10%. The only curative option remains complete surgical resection, but due to the delay in diagnosis, less than 20% of patients are eligible for surgery. Therefore, discovering diagnostic biomarkers for early detection is crucial for improving clinical outcomes. Metabolomics has become a powerful technology for biomarker discovery, and several metabolomic-based panels have been proposed for PDAC diagnosis, but these advances have not yet been translated into the clinic. Therefore, this review focused on summarizing metabolites identified for the early diagnosis of PDAC in the last five years. Bibliographic searches were performed in the PubMed, Scopus and WOS databases, using the terms "Biomarkers, Tumor", "Pancreatic Neoplasms", "Early Diagnosis", "Metabolomics" and "Lipidome" (January 2018-March 2023), and resulted in the selection of fourteen original studies that compared PDAC patients with subjects with other pancreatic diseases. These investigations showed amino acid and lipid metabolic pathways as the most commonly altered, reflecting their potential for biomarker research. Furthermore, other relevant metabolites such as glucose and lactate were detected in the pancreas tissue and body fluids from PDAC patients. Our results suggest that the use of metabolomics remains a robust approach to improve the early diagnosis of PDAC. However, these studies showed heterogeneity with respect to the metabolomics techniques used and further studies will be needed to validate the clinical utility of these biomarkers.
Collapse
Affiliation(s)
- Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Olga M García-Valdeavero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Mercedes Peña
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Cristina Jiménez-Luna
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), 18014 Granada, Spain
| |
Collapse
|
41
|
Demir Karaman E, Işık Z. Multi-Omics Data Analysis Identifies Prognostic Biomarkers across Cancers. Med Sci (Basel) 2023; 11:44. [PMID: 37489460 PMCID: PMC10366886 DOI: 10.3390/medsci11030044] [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: 05/19/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 07/26/2023] Open
Abstract
Combining omics data from different layers using integrative methods provides a better understanding of the biology of a complex disease such as cancer. The discovery of biomarkers related to cancer development or prognosis helps to find more effective treatment options. This study integrates multi-omics data of different cancer types with a network-based approach to explore common gene modules among different tumors by running community detection methods on the integrated network. The common modules were evaluated by several biological metrics adapted to cancer. Then, a new prognostic scoring method was developed by weighting mRNA expression, methylation, and mutation status of genes. The survival analysis pointed out statistically significant results for GNG11, CBX2, CDKN3, ARHGEF10, CLN8, SEC61G and PTDSS1 genes. The literature search reveals that the identified biomarkers are associated with the same or different types of cancers. Our method does not only identify known cancer-specific biomarker genes, but also proposes new potential biomarkers. Thus, this study provides a rationale for identifying new gene targets and expanding treatment options across cancer types.
Collapse
Affiliation(s)
- Ezgi Demir Karaman
- Department of Computer Engineering, Institute of Natural and Applied Sciences, Dokuz Eylul University, Izmir 35390, Turkey
| | - Zerrin Işık
- Department of Computer Engineering, Faculty of Engineering, Dokuz Eylul University, Izmir 35390, Turkey
| |
Collapse
|
42
|
Zhang J, Liu S, Song J, Zhou J, Zeng Q, Lin Z, Yu K, Zhang S, Qiu M, Chen Y, Hu Z. Improvement of postoperative quality of life in patients with esophageal squamous cell carcinoma: does tea consumption have a role? BMC Public Health 2022; 22:2165. [DOI: 10.1186/s12889-022-14646-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022] Open
Abstract
Abstract
Background
To investigate the effect of tea consumption on the improvement of postoperative quality of life in male patients with esophageal squamous cell carcinoma (ESCC).
Methods
The quality of life information of 290 male patients with ESCC was collected. The time to deterioration and the number of events in each area of quality of life was calculated by time-to-deterioration (TTD) model. The association between postoperative tea drinking and postoperative quality of life in male ESCC patients was investigated using the Cox proportional risk model.
Results
Postoperative tea-drinking patients experienced delayed TTD in multiple domains, including general health, physical, role, emotional, and cognitive function, fatigue, nausea and vomiting, dyspnea, loss of appetite, constipation, diarrhea, eating problems, difficulty swallowing, choking while swallowing saliva, dry mouth, taste difficulties, coughing, and speech problems. The multivariate Cox regression analysis showed that drinking tea after surgery improved quality of life, including physical function (HR = 0.722, 95% CI: 0.559-0.933), role function (HR = 0.740, 95% CI: 0.557-0.983), eating problems (HR = 0.718, 95% CI: 0.537-0.960), odynophagia (HR = 0.682, 95% CI: 0.492-0.945), trouble swallowing saliva (HR = 0.624, 95% CI: 0.444-0.877), coughing (HR = 0.627, 95% CI: 0.442-0.889) and speech problems (HR = 0.631, 95% CI: 0.441-0.903). Furthermore, the improvement was more significant in patients who drank tea before surgery and continued to drink tea after surgery.
Conclusions
Postoperative tea drinking had a positive effect on delay in clinical deterioration and improvements in multiple functions and symptoms associated with ESCC in men.
Collapse
|
43
|
Lee YH, Lee NR, Lee CH. Comprehensive Metabolite Profiling of Four Different Beans Fermented by Aspergillus oryzae. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227917. [PMID: 36432017 PMCID: PMC9695057 DOI: 10.3390/molecules27227917] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
Fermented bean products are used worldwide; most of the products are made using only a few kinds of beans. However, the metabolite changes and contents in the beans generally used during fermentation are unrevealed. Therefore, we selected four different beans (soybean, Glycine max, GM; wild soybean, Glycine soja, GS; common bean, Phaseolus vulgaris, PV; and hyacinth bean, Lablab purpureus, LP) that are the most widely consumed and fermented with Aspergillus oryzae. Then, metabolome and multivariate statistical analysis were performed to figure out metabolite changes during fermentation. In the four beans, carbohydrates were decreased, but amino acids and fatty acids were increased in the four beans as they fermented. The relative amounts of amino acids were relatively abundant in fermented PV and LP as compared to other beans. In contrast, isoflavone aglycones (e.g., daidzein, glycitein, and genistein) and DDMP-conjugated soyasaponins (e.g., soyasaponins βa and γg) were increased in GM and GS during fermentation. Notably, these metabolite changes were more significant in GS than GM. In addition, the increase of antioxidant activity in fermented GS was significant compared to other beans. We expect our research provides a basis to extend choice for bean fermentation for consumers and food producers.
Collapse
Affiliation(s)
- Yeon Hee Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Na-Rae Lee
- Research Institute for Bioactive-Metabolome Network, Konkuk University, Seoul 05029, Republic of Korea
- Correspondence: (N.-R.L.); (C.H.L.); Tel.: +82-2-2049-6177 (C.H.L.)
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
- Research Institute for Bioactive-Metabolome Network, Konkuk University, Seoul 05029, Republic of Korea
- Correspondence: (N.-R.L.); (C.H.L.); Tel.: +82-2-2049-6177 (C.H.L.)
| |
Collapse
|
44
|
Tokareva A, Chagovets V, Starodubtseva N, Rodionov V, Kometova V, Chingin K, Frankevich V. Lipidomic markers of breast cancer malignant tumor histological types. BIOMEDITSINSKAYA KHIMIYA 2022; 68:375-382. [DOI: 10.18097/pbmc20226805375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The molecular profile of a tumor is associated with its histological type and can be used both to study the mechanisms of tumor progression and to diagnose it. In this work, changes in the lipid profile of a malignant breast tumor and the adjacent tissue were studied. The potential possibility of determining the histological type of the tumor by its lipid profile was evaluated. Lipid profiling was performed by reverse-phase chromato-mass-spectrometric analysis the tissue of lipid extract with identification of lipids by characteristic fragments. Potential lipid markers of the histological type of tumor were determined using the Kruskal-Wallis test. Impact of lipid markers was calculated by MetaboAnalyst. Classification models were built by support vector machines with linear kernel and 1-vs-1 architecture. Models were validated by leave-one out cross-validation. Accuracy of models based on microenvironment tissue, were 99% and 75%, accuracy of models, based on tumor tissue, were 90% and 40% for the positive ion mode and negative ion mode respectively. The lipid profile of marginal (adjacent) tissue can be used for identification histological types of breast cancer. Glycerophospholipid metabolism pathway changes were statistically significant in the adjacent tissue and tumor tissue.
Collapse
Affiliation(s)
- A.O. Tokareva
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - V.V. Chagovets
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - N.L. Starodubtseva
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - V.V. Rodionov
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - V.V. Kometova
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - K.S. Chingin
- Jiangxi Key Laboratory for Mass Spectrometry and Instrumentation, East China University of Technology, Nanchang, China
| | - V.E. Frankevich
- Academician V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia; Siberian State Medical University, Tomsk, 634050 Russia
| |
Collapse
|
45
|
Wei Y, Li Y, Chen Y, Liu P, Huang S, Zhang Y, Sun Y, Wu Z, Hu M, Wu Q, Wu H, Liu F, She T, Ning Z. ALDH1: A potential therapeutic target for cancer stem cells in solid tumors. Front Oncol 2022; 12:1026278. [PMID: 36387165 PMCID: PMC9650078 DOI: 10.3389/fonc.2022.1026278] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/12/2022] [Indexed: 12/02/2022] Open
Abstract
Solid tumors can be divided into benign solid tumors and solid malignant tumors in the academic community, among which malignant solid tumors are called cancers. Cancer is the second leading cause of death in the world, and the global incidence of cancer is increasing yearly New cancer patients in China are always the first. After the concept of stem cells was introduced in the tumor community, the CSC markers represented by ALDH1 have been widely studied due to their strong CSC cell characteristics and potential to be the driving force of tumor metastasis. In the research results in the past five years, it has been found that ALDH1 is highly expressed in various solid cancers such as breast cancer, lung cancer, colorectal cancer, liver cancer, gastric cancer, cervical cancer, esophageal cancer, ovarian cancer, head,and neck cancer. ALDH1 can activate and transform various pathways (such as the USP28/MYC signaling pathway, ALDH1A1/HIF-1α/VEGF axis, wnt/β-catenin signaling pathway), as well as change the intracellular pH value to promote formation and maintenance, resulting in drug resistance in tumors. By targeting and inhibiting ALDH1 in tumor stem cells, it can enhance the sensitivity of drugs and inhibit the proliferation, differentiation, and metastasis of solid tumor stem cells to some extent. This review discusses the relationship and pathway of ALDH1 with various solid tumors. It proposes that ALDH1 may serve as a diagnosis and therapeutic target for CSC, providing new insights and new strategies for reliable tumor treatment.
Collapse
Affiliation(s)
- Yaolu Wei
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yan Li
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yenan Chen
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Pei Liu
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Sheng Huang
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yuping Zhang
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yanling Sun
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhe Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Meichun Hu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qian Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Hongnian Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Fuxing Liu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| | - Tonghui She
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| | - Zhifeng Ning
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| |
Collapse
|
46
|
Yang XL, Wang P, Ye H, Jiang M, Su YB, Peng XX, Li H, Zhang JY. Untargeted serum metabolomics reveals potential biomarkers and metabolic pathways associated with esophageal cancer. Front Oncol 2022; 12:938234. [PMID: 36176418 PMCID: PMC9513043 DOI: 10.3389/fonc.2022.938234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/16/2022] [Indexed: 12/24/2022] Open
Abstract
Metabolomics has been reported as an efficient tool to screen biomarkers that are related to esophageal cancer. However, the metabolic biomarkers identifying malignant degrees and therapeutic efficacy are still largely unknown in the disease. Here, GC-MS-based metabolomics was used to understand metabolic alteration in 137 serum specimens from patients with esophageal cancer, which is approximately two- to fivefold as many plasma specimens as the previous reports. The elevated amino acid metabolism is in sharp contrast to the reduced carbohydrate as a characteristic feature of esophageal cancer. Comparative metabolomics showed that most metabolic differences were determined between the early stage (0–II) and the late stage (III and IV) among the 0–IV stages of esophageal cancer and between patients who received treatment and those who did not receive treatment. Glycine, serine, and threonine metabolism and glycine were identified as the potentially overlapped metabolic pathway and metabolite, respectively, in both disease progress and treatment effect. Glycine, fructose, ornithine, and threonine can be a potential array for the evaluation of disease prognosis and therapy in esophageal cancer. These results highlight the means of identifying previously unknown biomarkers related to esophageal cancer by a metabolomics approach.
Collapse
Affiliation(s)
- Xiao-li Yang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Peng Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology and College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Hua Ye
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology and College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Ming Jiang
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Yu-bin Su
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xuan-xian Peng
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
| | - Hui Li
- State Key Laboratory of Bio-Control, School of Life Sciences, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, University City, Guangzhou, China
- *Correspondence: Jian-ying Zhang, ; Hui Li,
| | - Jian-ying Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Jian-ying Zhang, ; Hui Li,
| |
Collapse
|
47
|
Yan C, Wu D, Gan L, Wang J, Yang W, Xu B. Significant metabolic alterations in non-small cell lung cancer patients by epidermal growth factor receptor-targeted therapy and PD-1/PD-L1 immunotherapy. Front Pharmacol 2022; 13:949745. [PMID: 36034789 PMCID: PMC9403486 DOI: 10.3389/fphar.2022.949745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 07/20/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Cancer-related deaths are primarily attributable to lung cancer, of which non-small cell lung cancer (NSCLC) is the most common type. Molecular targeting therapy and antitumor immunotherapy have both made great strides in the treatment of NSCLC, but their underlying mechanisms remain unclear, especially from a metabolic perspective. Methods: Herein, we used a nontargeted metabolomics approach based on liquid chromatography-mass spectrometry to analyze the metabolic response of NSCLC patients to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) or PD-1/PD-L1 inhibitors. Multiple analyses, including principal component analysis (PCA), orthogonal partial least squares-discriminant analysis (OPLS-DA) and pathway analysis, were used for metabolic data analysis. Additionally, differential metabolites were analysed and identified by publically available and integrated databases. Results: After treatment with EGFR-TKIs or PD-1/PD-L1 inhibitors, glutamate/glutamine, phenylalanine, n-acetyl-l-leucine, n-acetyl-d-tryptophan, D-n-valine, arachidonic acid, and linoleic acid levels were significantly increased in patients with NSCLC, whereas carnitine, stearyl carnitine, palmitoyl carnitine, linoleic carnitine, and palmitic acid levels were markedly decreased. Compared with newly diagnosed, untreated patients, there were three shared metabolic pathways (phenylalanine metabolism, glycerophospholipid metabolism, and D-glutamine and D-glutamate metabolism) in the EGFR-TKIs or PD-1/PD-L1 inhibitor-treated groups, all of which were related to lipid and amino acid metabolism. Moreover, there were significant differences in lipid metabolism (glycerophospholipid metabolism and phosphatidylinositol signaling) and amino acid metabolism (tryptophan metabolism) between the EGFR-TKI and PD-1/PD-L1 inhibitor groups. Conclusion: Our results show that EGFR-TKIs and PD-1/PD-L1 inhibitors induce changes in carnitine, amino acids, fatty acids, and lipids and alter related metabolic pathways in NSCLC patients. Endogenous metabolism changes occur due to drug action and might be indicative of antitumor therapeutic effect. These findings will provide new clues for identifying the antitumor mechanism of these two treatments from the perspective of metabolism.
Collapse
Affiliation(s)
- Chen Yan
- Department of Clinical Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dan Wu
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Lingling Gan
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Jun Wang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenyu Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bei Xu
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- *Correspondence: Bei Xu,
| |
Collapse
|
48
|
Early Diagnosis of Oral Squamous Cell Carcinoma Based on Histopathological Images Using Deep and Hybrid Learning Approaches. Diagnostics (Basel) 2022; 12:diagnostics12081899. [PMID: 36010249 PMCID: PMC9406837 DOI: 10.3390/diagnostics12081899] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common head and neck cancer types, which is ranked the seventh most common cancer. As OSCC is a histological tumor, histopathological images are the gold diagnosis standard. However, such diagnosis takes a long time and high-efficiency human experience due to tumor heterogeneity. Thus, artificial intelligence techniques help doctors and experts to make an accurate diagnosis. This study aimed to achieve satisfactory results for the early diagnosis of OSCC by applying hybrid techniques based on fused features. The first proposed method is based on a hybrid method of CNN models (AlexNet and ResNet-18) and the support vector machine (SVM) algorithm. This method achieved superior results in diagnosing the OSCC data set. The second proposed method is based on the hybrid features extracted by CNN models (AlexNet and ResNet-18) combined with the color, texture, and shape features extracted using the fuzzy color histogram (FCH), discrete wavelet transform (DWT), local binary pattern (LBP), and gray-level co-occurrence matrix (GLCM) algorithms. Because of the high dimensionality of the data set features, the principal component analysis (PCA) algorithm was applied to reduce the dimensionality and send it to the artificial neural network (ANN) algorithm to diagnose it with promising accuracy. All the proposed systems achieved superior results in histological image diagnosis of OSCC, the ANN network based on the hybrid features using AlexNet, DWT, LBP, FCH, and GLCM achieved an accuracy of 99.1%, specificity of 99.61%, sensitivity of 99.5%, precision of 99.71%, and AUC of 99.52%.
Collapse
|
49
|
Screening of four key genes in esophageal carcinoma based on TCGA and GEO data and verification of anti-proliferative effect of LAPTM4B knockdown in esophageal carcinoma cells invitro. Arch Biochem Biophys 2022; 728:109352. [PMID: 35863479 DOI: 10.1016/j.abb.2022.109352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/11/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022]
Abstract
Esophageal carcinoma (ESCA) is one of the most prevalent and aggressive malignancies of the gastrointestinal tract and constitutes sixth primary cause of cancer-related death worldwide. It is urgently needed to identify effective therapeutic targets. Differentially expressed genes (DEGs) involved in ESCA were identified via bioinformatics analysis. Four DEGs were selected for further analysis using Gene Expression Profiling Interactive Analysis, Human Protein Atlas, UALCAN web portal, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. 5-ethynyl-2'-deoxyuridine incorporation and cell counting kit-8 assays were used to evaluate cell proliferation. Western blot analysis was used to detect the protein levels of lysosomal-associated transmembrane protein 4B (LAPTM4B), Notch1, hairy and enhancer of split 1 (Hes1), and hairy and enhancer of split-related with YRPW motif 1 (Hey1). Results showed that LAPTM4B, Bcl-2 homology domain 3 (BH3)-interacting domain death agonist (BID), epithelial cell transforming sequence 2 (ECT2), and aurora kinase A (AURKA) were upregulated in several types of tumors including ESCA and correlated with tumor stage and tumor histology based on bioinformatics analysis. KEGG pathway analysis suggested that LAPTM4B-associated genes were significantly enriched in Notch pathway. Meanwhile, BID-, ECT2-, and AURKA-correlated genes were particularly enriched in p53 signaling pathway. Additionally, we found that LAPTM4B silencing inhibited cell proliferation and Notch pathway in ESCA cells. Notch1 overexpression abrogated LAPTM4B knockdown-induced proliferation reduction in ESCA cells. In conclusion, LAPTM4B silencing inhibited proliferation in ESCA cells by inactivating the Notch pathway.
Collapse
|
50
|
Jiang W, Wang X, Su S, Du S, Song H. Identifying the shared genes and KEGG pathways of Resolvin D1-targeted network and osteoarthritis using bioinformatics. Bioengineered 2022; 13:9839-9854. [PMID: 35436417 PMCID: PMC9161845 DOI: 10.1080/21655979.2022.2061288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Osteoarthritis (OA) is a common chronic degenerative disease characterized by the loss of articular cartilage, which causes loss of joint function and reduce quality of life. Resolvin D1 (RvD1) has shown interesting anti-inflammatory effects; however, the mechanism of action of RvD1 in OA remains unclear. The aim of this study was to investigate the potential mechanism of RvD1 in OA by bioinformatics and partial in vitro mechanisms. Here, 106 shared differentially expressed genes (DEGs) were identified based on the GSE82107, GSE55235, GSE55457 dataset; 700 DEGs were identified based on GSE169077. Enrichment analyses of these genes were then successively conducted. RvD1-targeted genes and KEGG pathways are identified by STITCH. 27 shared KEGG pathways were identified among RvD1-targeted pathways and OA. Furthermore, cell apoptosis assay, western blotting, real-time fluorescent quantitative PCR (qRT-PCR), enzyme linked immunosorbent assay (ELISA) were used to confirm the expression levels of the key genes of shared Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways between RvD1-targeted and OA in IL-1β treated rat knee chondrocytes. The results showed that RvD1-targeted pathways and the expression of nuclear p65, p53, and p-JNK were inhibited in the RvD1 group compared with the IL-1β group. Thus, the findings indicate that RvD1 may inhibit the development of OA through NF/kB, p53, MAPK/JNK, PI3K-AKT signaling pathways, and act as a treatment for OA.
Collapse
Affiliation(s)
- Wenjun Jiang
- Department of Sports Medicine and Rehabilitation, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, Shandong, China
| | - Xiaoying Wang
- Department of Sports Medicine and Rehabilitation, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, Shandong, China
| | - Siwei Su
- Department of Sports Medicine and Rehabilitation, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, Shandong, China
| | - Sen Du
- Department of Sports Medicine and Rehabilitation, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, Shandong, China
| | - Hongqiang Song
- Department of Sports Medicine and Rehabilitation, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, Shandong, China
| |
Collapse
|