1
|
Li D, Zheng P, Huang S. SLC12A9 is an immunological and prognostic biomarker for glioma. Gene 2025; 937:149136. [PMID: 39622394 DOI: 10.1016/j.gene.2024.149136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Glioma is one of the most common malignant brain tumors. It has a high rate of progression and a poor prognosis, and effective biomarkers still need to be identified. The solute carrier family 12 (SLC12) family has been reported to be involved in various physiological and pathological processes, but their functional roles in glioma remain unclear. METHODS Using public datasets, we studied the mutation and expression level of SLC12 family genes in glioma and identified the significantly differentially expressed member solute carrier family 12 member 9 (SLC12A9). We further predicted the prognostic role of SLC12A9 in glioma by using the Kaplan-Meier method and Cox regression analysis. Then, we performed biological functional enrichment analysis. We focused on the relationships between SLC12A9 expression and immune infiltration in glioma. Meanwhile, we conducted in vitro experiments to evaluate the effect of SLC12A9 expression on glioma cells. RESULTS Among the members of the SLC12 family, SLC12A9 had the highest mutation rate in glioma, with gene amplification as the major mutation type, and its expression was significantly upregulated in glioma. Higher SLC12A9 expression was significantly associated with older age, higher grade, wild-type isocitrate dehydrogenase (IDH), and a worse prognosis. The functional enrichment analysis indicated that SLC12A9 is mainly related to ion channel annotation. Gene set enrichment analysis (GSEA) revealed that SLC12A9 was mainly related to the DNA replication pathway. Furthermore, we found that SLC12A9 correlated with tumor-infiltrating immune cells and immune checkpoints. Thus, SLC12A9 may be involved in regulating the immune response of glioma. Finally, our in vitro experiments revealed that silencing SLC12A9 dramatically inhibited glioma cell growth and migration. CONCLUSIONS We showed that SLC12A9 may be a new predictive biomarker for glioma diagnosis, prognosis, and immunotherapy response, offering helpful guidelines to advance glioma treatment.
Collapse
Affiliation(s)
- Danting Li
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China; School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Peilin Zheng
- Department of General Practice, People's Hospital of Longhua, Shenzhen 518109, Guangdong, China.
| | - Shoujun Huang
- College of Life Sciences, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
2
|
Baldari S, Antonini A, Di Rocco G, Toietta G. Expression pattern and prognostic significance of aldehyde dehydrogenase 2 in lung adenocarcinoma as a potential predictor of immunotherapy efficacy. CANCER INNOVATION 2025; 4:e149. [PMID: 39640071 PMCID: PMC11620833 DOI: 10.1002/cai2.149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/30/2024] [Accepted: 05/23/2024] [Indexed: 12/07/2024]
Abstract
Background The incidence of alcohol-associated cancers is higher within Asian populations having an increased prevalence of an inactivating mutation in aldehyde dehydrogenase 2 (ALDH2), a mitochondrial enzyme required for the clearance of acetaldehyde, a cytotoxic metabolite of ethanol. The role of alcohol consumption in promoting lung cancer is controversial, and little attention has been paid to the association between alcohol drinking and pulmonary ALDH2 expression. Methods We performed a comprehensive bioinformatic analysis of multi-omics data available in public databases to elucidate the role of ALDH2 in lung adenocarcinoma (LUAD). Results Transcriptional and proteomic data indicate a substantial pulmonary expression of ALDH2, which is functional for the metabolism of alcohol diffused from the bronchial circulation. ALDH2 expression is higher in healthy lung tissue than in LUAD and inhibits cell cycle, apoptosis, and epithelial-mesenchymal transition pathways. Moreover, low ALDH2 mRNA levels predict poor prognosis and low overall survival in LUAD patients. Interestingly, ALDH2 expression correlates with immune infiltration in LUAD. Conclusions A better understanding of the role of ALDH2 in lung tumor progression and immune infiltration might support its potential use as a prognostic marker and therapeutic target for improving immunotherapeutic response.
Collapse
Affiliation(s)
- Silvia Baldari
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Annalisa Antonini
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic TargetsIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Gabriele Toietta
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| |
Collapse
|
3
|
Wang M, Wang Z, Zou X, Yang D, Xu K. The regulatory role of KIAA1429 in epithelial-mesenchymal transition in cervical cancer via mediating m6A modification of BTG2. Cytotechnology 2025; 77:34. [PMID: 39760059 PMCID: PMC11699186 DOI: 10.1007/s10616-024-00694-3] [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: 10/28/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025] Open
Abstract
Cervical cancer (CC) represents one of the important cancers affecting global female population worldwide. We sought to elucidate the roles and mechanisms of KIAA1429 in the malignant properties of CC cells and the epithelial-mesenchymal transition (EMT) process. KIAA1429 was predicted to be abnormally expressed in CC and correlate with shortened survival of CC patients by GEPIA2 and GSCA databases. High expression of KIAA1429 in human CC cell lines (SiHa, HT-3) was validated by RT-qPCR and Western blot assays. A series of small interfering (si)RNAs including si-KIAA1429-1, si-KIAA1429-2, si-YTHDF2, si-BTG2, and si-negative control (NC) were utilized to interfere the expression levels of KIAA1429, YTHDF2, and BTG2, respectively. Consequently, KIAA1429 silencing attenuated the proliferation, migratory, and invasive functions of CC cells and repressed EMT while promoting CC cell apoptosis. Mechanistically, KIAA1429 could affect N6-methyladenosine (m6A) modification to attenuate the stability of BTG2 mRNA and down-regulate its expression. Additionally, loss of BTG2 partly counteracted the effects of si-KIAA1429 on repressing the malignant activities of CC cells. The aforementioned results collectively demonstrated that KIAA1429-mediated m6A modification of BTG2 and contributed to malignant progression of CC in vitro. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00694-3.
Collapse
Affiliation(s)
- Mingyang Wang
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003 Guizhou China
| | - Zhiliang Wang
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003 Guizhou China
| | - Xiaofeng Zou
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003 Guizhou China
| | - Danhe Yang
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003 Guizhou China
| | - Ke Xu
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003 Guizhou China
| |
Collapse
|
4
|
Jiang F, Ahmad S, Kanwal S, Hameed Y, Tang Q. Key wound healing genes as diagnostic biomarkers and therapeutic targets in uterine corpus endometrial carcinoma: an integrated in silico and in vitro study. Hereditas 2025; 162:5. [PMID: 39833941 DOI: 10.1186/s41065-025-00369-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Uterine Corpus Endometrial Carcinoma (UCEC) is a prevalent gynecologic malignancy with complex molecular underpinnings. This study identifies key woundhealing genes involved in UCEC and elucidates their roles through a comprehensive analysis. METHODS In silico and in vitro experiments. RESULTS Seventy wound healing-associated genes were extracted from the Gene Ontology (GO) database, and a protein-protein interaction (PPI) network was constructed using the STRING database. CytoHubba analysis in Cytoscape identified six pivotal hub genes: CD44, FGF2, FGF10, KDM6A, FN1, and MMP2. These genes exhibited significantly lower expression in UCEC cell lines compared to normal controls, as confirmed by RT-qPCR. Receiver Operating Characteristic (ROC) analysis demonstrated their potential as diagnostic biomarkers, with Area Under the Curve (AUC) values ranging from 0.94 to 1.00. Validation using TCGA datasets revealed consistent downregulation of these genes in UCEC samples, corroborated by immunohistochemical staining. Promoter methylation analysis showed significantly higher methylation levels in UCEC, correlating with decreased mRNA expression and poor survival outcomes. Genetic alteration analysis indicated frequent mutations in FN1 and KDM6A, although these did not significantly affect survival. Functional analysis using the CancerSEA database highlighted the involvement of these genes in critical cancer-related processes, including angiogenesis, apoptosis, and metastasis. Immune correlation studies revealed significant associations with immune inhibitor genes and distinct expression patterns across immune subtypes. Overexpression studies in UCEC cell lines demonstrated that CD44 and MMP2 reduce proliferative ability while enhancing migration and wound healing. CONCLUSION Collectively, these findings underscore the crucial roles of CD44, FGF2, FGF10, KDM6A, FN1, and MMP2 in UCEC pathogenesis, highlighting their potential as biomarkers and therapeutic targets in this malignancy.
Collapse
Affiliation(s)
- Fuchuan Jiang
- Department of Gynaecology and Obstetrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - Sajjad Ahmad
- Gomal Medical College, D. I. Khan, KPK, Pakistan
| | - Sadia Kanwal
- Al Nafees Medical College and Hospital Islamabad, Islamabad, Pakistan
| | - Yasir Hameed
- Department of Biochemistry, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.
| | - Qian Tang
- Department of Gynaecology and Obstetrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
5
|
Wu L, Liu C, Hu W. Comprehensive investigation of matrix metalloproteinases in skin cutaneous melanoma: diagnostic, prognostic, and therapeutic insights. Sci Rep 2025; 15:2152. [PMID: 39820824 PMCID: PMC11739484 DOI: 10.1038/s41598-025-85887-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
The dysregulation of matrix metalloproteinases (MMPs) in skin cutaneous melanoma (SKCM) represents a critical aspect of tumorigenesis. In this study, we investigated the diagnostic, prognostic, and therapeutic aspects of the MMPs in SKCM. Thirteen SKCM cell lines and seven normal skin cell lines were cultured under standard conditions for experimental analyses. RNA and DNA were extracted, followed by RT-qPCR to assess MMP expression and promoter methylation analysis to determine methylation levels. Functional assays, including cell proliferation, colony formation, and wound healing, were conducted post-MMP7 knockdown using siRNA in A375 cells. Databases like GEPIA2, HPA, MEXPRESS, and miRNet were employed for expression, survival, methylation, and miRNA-mRNA network analyses. We investigated the expression and promoter methylation landscape of MMPs in SKCM cell lines, revealing significant (p-value < 0.05) up-regulation of MMP1, MMP7, MMP9, MMP10, MMP11, MMP12, MMP13, MMP14, and MMP25, alongside down-regulation of MMP2, MMP3, and MMP21. Furthermore, our analysis demonstrated a significant (p-value < 0.05) inverse correlation between MMP expression levels and promoter methylation status, suggesting a potential regulatory role of DNA methylation in MMP dysregulation. Notably, MMP7, MMP11, and MMP14 exhibited significant (p-value < 0.05) associations with the overall survival of SKCM patients, emphasizing their prognostic significance. Additionally, Receiver operating characteristic (ROC) curve analysis highlighted the significant (p-value < 0.05) diagnostic potential of MMP7, MMP11, and MMP14 in distinguishing SKCM from normal individuals. Subsequent validation across multiple cohorts confirmed significant (p-value < 0.05) elevated MMP expression levels in SKCM tissues, particularly in advanced disease stages, further emphasizing their role in tumor progression. Furthermore, we elucidated potential regulatory pathways involving miR-22-3p, which targets MMP7, MMP11, and MMP14 genes in SKCM. Our findings also revealed associations between MMP expression and immune modulation, drug sensitivity, and functional states of SKCM cells. Lastly, MMP7 knockdown in A375 cells significantly significant (p-value < 0.05) impacted several characteristics, including cell proliferation, colony formation, and wound healing. Our findings highlight the diagnostic, prognostic, and therapeutic potential of MMP7, MMP11, and MMP14 in SKCM. These MMPs could serve as biomarkers for early detection and targets for therapy. Future efforts should focus on preclinical and clinical validation to translate these insights into personalized diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Lingxia Wu
- Dermatology, Changzhi Second People's Hospital, Changzhi, 046000, Shanxi, China
| | - Chenxiaoxiao Liu
- The First Clinical Institute, Zunyi Medical University, Zunyi, 520300, Guizhou, China
| | - Weicai Hu
- Dermatology, Changzhi Second People's Hospital, Changzhi, 046000, Shanxi, China.
| |
Collapse
|
6
|
Yi J, Gao W, Wu C, Yang R, Yu C. A multidimensional pan-cancer analysis of NDUFA4L2 and verification of the oncogenic value in colon cancer. FASEB J 2025; 39:e70300. [PMID: 39792315 DOI: 10.1096/fj.202402165rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/01/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 4-like 2 (NDUFA4L2) protein is located in the mitochondria and can regulate cell proliferation. Some studies have shown that the high NDUFA4L2 expression is linked with poor prognosis and cancer progression in various patients with cancers. However, the correlation between NDUFA4L2 and pan-cancer is unknown. In this study, we used the databases and investigated the expression and variation of NDUFA4L2 in many cancers in detail to determine its diagnostic and prognostic values. We also validated the oncogenic capability of NDUFA4L2 in colon cancer by experiments or tissue microarray. In most cancers, NDUFA4L2 expression levels are upregulated, and a high level of NDUFA4L2 expression is associated with poor OS. In addition, the results showed that 10 proteins and 30 genes were correlated to NDUFA4L2. An additional analysis demonstrated that these genes and proteins exhibited either negative or positive correlations with specific pathways. Moreover, we discovered that NDUFA4L2 expression was related to tumor stemness, MSI, TMB, immune cell infiltration and ROS production. Our results show that NDUFA4L2 affects the development and mitochondrial function of colon cancer cell lines. Finally, we identified that NDUFA4L2 was sensitive to 10 anticancer drugs. Our study suggest that NDUFA4L2 could serve as a prognostic and therapeutic biomarker for most cancer, including colon cancer.
Collapse
Affiliation(s)
- Jianing Yi
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu, P. R. China
| | - Wenjie Gao
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu, P. R. China
| | - Cheng Wu
- Department of Geriatrics, Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu, P. R. China
| | - Ranzhiqiang Yang
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu, P. R. China
| | - Chunzhao Yu
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu, P. R. China
| |
Collapse
|
7
|
Zhang JN, Li LW, Cao MQ, Liu X, Yi ZL, Liu SS, Liu H. Functional Analysis and Experimental Validation of the Prognostic and Immune Effects of the Oncogenic Protein CDC45 in Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:11-25. [PMID: 39811603 PMCID: PMC11727330 DOI: 10.2147/bctt.s497975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025]
Abstract
Purpose Cell division cycle protein 45 (CDC45) plays a crucial role in DNA replication. This study investigates its role in breast cancer (BC) and its impact on tumor progression. Methods We utilized the GEO database to screen differentially expressed genes (DEGs) and conducted enrichment analysis on these genes. We established a Nomogram model based on CDC45 and other clinical indicators. Additionally, we performed protein-protein interaction (PPI) network construction, drug sensitivity analysis, and immune correlation analysis of CDC45. The function of CDC45 was further verified through cell and animal experiments. Results CDC45 is highly expressed in most tumors, including BC. The expression level of CDC45 was significantly associated with age, sex, race, cancer stage, and molecular subtypes (all p < 0.05). CDC45 was incorporated into a Nomogram model, which showed moderate accuracy in predicting patient prognosis. We also analyzed the co-expression genes of CDC45, including TOPBP1, GINS2, MCM5, GINS1, GINS4, POLE2, MCM2, MCM6, MCM4, and MCM7. Furthermore, CDC45 expression was closely linked to immune infiltration levels, immune checkpoint inhibitors, and the therapeutic response to small molecule drugs. Finally, both in vitro and in vivo experiments confirmed the cancer-promoting effect of CDC45 in BC. Conclusion The expression level of CDC45 is linked to the prognosis, immune infiltration, and drug sensitivity of BC. In vitro and in vivo experiments have confirmed that CDC45 acts as a cancer-promoting protein in breast cancer.
Collapse
Affiliation(s)
- Jia-Ning Zhang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, People’s Republic of China
- Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, People’s Republic of China
| | - Lin-Wei Li
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, People’s Republic of China
- Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, People’s Republic of China
| | - Man-Qing Cao
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, People’s Republic of China
- Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, People’s Republic of China
| | - Xin Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, People’s Republic of China
- Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, People’s Republic of China
| | - Zi-Lu Yi
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, People’s Republic of China
- Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, People’s Republic of China
| | - Sha-Sha Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, People’s Republic of China
- Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, People’s Republic of China
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, People’s Republic of China
- Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, People’s Republic of China
| |
Collapse
|
8
|
Ahmadi M, Motallebinezhad M, Mousavi P, Miladipour AH, Fooladgar S, Ghafouri-Fard S, Fazeli SA. Bioinformatics analysis of mitochondrial metabolism-related genes demonstrates their importance in renal cell carcinoma. Discov Oncol 2025; 16:28. [PMID: 39789365 PMCID: PMC11717778 DOI: 10.1007/s12672-025-01780-4] [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: 10/07/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
PURPOSE Clear cell renal cell carcinoma (ccRCC) is resistant to radiotherapy and chemotherapy. Thus, it is necessary to find new diagnostic markers and therapeutic targets to increase the overall outcomes of ccRCC. Recent studies have shown that therapeutic methods that interfere with the energy transfer system can also positively affect the treatment process. METHODS The present study is focused on finding markers associated with mitochondrial metabolic pathways that affect the outcome of ccRCC. For this purpose, we investigated various aspects of the relationship between mitochondrial metabolism and ccRCC based on analysis of gene network connections and differentially expressed genes, through assessment of protein-protein interaction, mutations, and promoter methylation on the related genes. We also investigated gene interaction with miRNAs and immune infiltration analysis. RESULTS Through these steps, we provided a list of possible diagnostic markers and therapeutic targets for ccRCC. CONCLUSION The current study further proved the importance of mitochondrial metabolic pathways in the pathogenesis of ccRCC and provided a list of possible diagnostic markers and therapeutic targets from these pathways that can be used in ccRCC.
Collapse
Affiliation(s)
- Mohsen Ahmadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Motallebinezhad
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pegah Mousavi
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Amir Hossein Miladipour
- Chronic Kidney Disease Research Center, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shamim Fooladgar
- Department of Biology, School of Converging Sciences and Technologies, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyed Amirhossein Fazeli
- Clinical Research and Development Center, Division of Nephrology, Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Division of Nephrology, Department of Internal Medicine, Taleghani General Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Westermann L, Diergaarde B, Heidegger S, Poeck H, Szczepański MJ, Reichert TE, Spoerl S, Whiteside TL, Spoerl S, Ludwig N. Expression of vesiculation-related genes is associated with a tumor-promoting microenvironment: a pan-cancer analysis. Clin Transl Oncol 2025:10.1007/s12094-024-03796-8. [PMID: 39776398 DOI: 10.1007/s12094-024-03796-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/11/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Small extracellular vesicles (sEV) released by tumor cells (tumor-derived sEV; TEX) mediate intercellular communication between tumor and non-malignant cells and were shown to impact disease progression. This study investigates the relationship between the expression levels of the vesiculation-related genes linked to sEV production and the tumor microenvironment (TME). METHODS Two independent gene sets were analyzed, both previously linked to sEV production in various non-malignant or malignant cells. Expression profiles were compared among 28 tumor types listed in the Cancer Genome Atlas (TCGA). Gene expression and survival analysis (GEPIA2), immunogenomic analysis (TISIDB), and genomic analysis (GSCA) were performed. RESULTS Vesiculation-related genes were overexpressed in tissues of most tumor types compared to healthy tissues, and high expression levels were associated with worse overall survival in cervical squamous cell carcinoma, kidney chromophobe, lower grade glioma, hepatocellular carcinoma, lung squamous cell carcinoma, and pancreatic adenocarcinoma but with improved overall survival in kidney renal clear cell carcinoma. Expression of these signatures correlated with an increased abundance of infiltrating CD4( +) T cells and dendritic cells, a decreased abundance of B cells and eosinophils, and activation of tumor cell apoptosis and epithelial-mesenchymal transition pathways in all tumor types. 17-AAG was identified as a potential drug candidate to target tumors with elevated expression of vesiculation-related genes. CONCLUSIONS Vesiculation-related genes were associated with distinct immunological and genomic landscapes further emphasizing the important role of TEX in cancer progression.
Collapse
Affiliation(s)
- Luisa Westermann
- Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Brenda Diergaarde
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simon Heidegger
- Department of Medicine III, School of Medicine and Health, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Early Clinical Development, Oncology, Genentech, South San Francisco, CA, USA
| | - Hendrik Poeck
- Clinic and Polyclinic for Internal Medicine III, University Hospital Regensburg and Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
| | - Mirosław J Szczepański
- Chair and Department of Biochemistry, Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Torsten E Reichert
- Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Silvia Spoerl
- Department of Internal Medicine 5, Hematology/Oncology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Theresa L Whiteside
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pathology, UPMC Hillman Cancer Center, University of Pittsburgh, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA
- Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steffen Spoerl
- Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Nils Ludwig
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pathology, UPMC Hillman Cancer Center, University of Pittsburgh, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
10
|
Li Q, Li H. Integrating bioinformatics and machine learning to identify AhR-related gene signatures for prognosis and tumor microenvironment modulation in melanoma. Front Immunol 2025; 15:1519345. [PMID: 39835132 PMCID: PMC11743449 DOI: 10.3389/fimmu.2024.1519345] [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: 10/29/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
Background The Aryl Hydrocarbon Receptor (AhR) pathway significantly influences immune cell regulation, impacting the effectiveness of immunotherapy and patient outcomes in melanoma. However, the specific downstream targets and mechanisms by which AhR influences melanoma remain insufficiently understood. Methods Melanoma samples from The Cancer Genome Atlas (TCGA) and normal skin tissues from the Genotype-Tissue Expression (GTEx) database were analyzed to identify differentially expressed genes, which were intersected with a curated list of AhR-related pathway genes. Prognostic models were subsequently developed, and feature genes were identified. Advanced methodologies, including Gene Set Enrichment Analysis (GSEA) and immune cell infiltration analysis, were employed to explore the biological significance of these genes. The stability of the machine learning models and the relationship between gene expression and immune infiltrating cells were validated using three independent melanoma datasets. A mouse melanoma model was used to validate the dynamic changes of the feature genes during tumor progression. The relationship between the selected genes and drug sensitivity, as well as non-coding RNA interactions, was thoroughly investigated. Results Our analysis identified a robust prognostic model, with four AhR-related genes (MAP2K1, PRKACB, KLF5, and PIK3R2) emerging as key contributors to melanoma progression. GSEA revealed that these genes are involved in primary immunodeficiency. Immune cell infiltration analysis demonstrated enrichment of CD4+ naïve and memory T cells, macrophages (M0 and M2), and CD8+ T cells in melanoma, all of which were associated with the expression of the four feature genes. Importantly, the diagnostic power of the prognostic model and the relevance of the feature genes were validated in three additional independent melanoma datasets. In the mouse melanoma model, Map2k1 and Prkacb mRNA levels exhibited a progressive increase with tumor progression, supporting their role in melanoma advancement. Conclusions This study presents a comprehensive analysis of AhR-related genes in melanoma, highlighting MAP2K1, PRKACB, KLF5, and PIK3R2 as key prognostic markers and potential therapeutic targets. The integration of bioinformatics and machine learning provides a robust framework for enhancing prognostic evaluation in melanoma patients and offers new avenues for the development of treatments, particularly for those resistant to current immunotherapies.
Collapse
Affiliation(s)
- Qianru Li
- Department of Dermatology, Wuhan No.1 Hospital, Wuhan, Hubei, China
- Hubei Province & Key Laboratory of Skin Infection and Immunity, Wuhan, Hubei, China
| | - Heli Li
- Division of Child Healthcare, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Li J, Liu L, Zong G, Yang Z, Zhang D, Zhao B. Knockdown of CENPF induces cell cycle arrest and inhibits epithelial‑mesenchymal transition progression in glioma. Oncol Lett 2025; 29:61. [PMID: 39611064 PMCID: PMC11602827 DOI: 10.3892/ol.2024.14807] [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: 04/24/2024] [Accepted: 10/02/2024] [Indexed: 11/30/2024] Open
Abstract
Gliomas are among the most common malignant tumors of the central nervous system. Despite surgical resection followed by postoperative radiotherapy and chemotherapy, their prognosis remains unfavorable. The present study aimed to assess new mechanisms and explore promising prognostic biomarkers for patients with glioma using comprehensive bioinformatics analysis and in vitro and in vivo assays. Overlapping differentially expressed genes were screened from The Cancer Genome Atlas, GSE111260 and GSE16011 samples for protein-protein interaction networks, a risk score model, gene mutation analysis and a nomogram to identify the prognostic hub genes. Subsequently, an immunoassay was performed to determine key genes. Functional and animal assays were then performed to assess the tumorigenesis of the key genes in glioma. Using bioinformatics analysis, centromere protein F (CENPF), kinesin superfamily member 20A, kinesin superfamily protein 4A and marker of proliferation Ki-67 were identified as potential prognostic biomarkers for patients with glioma. Furthermore, CENPF knockdown was demonstrated to suppress the proliferation and metastasis of glioma cells, and induce G2 arrest in the cell cycle. Moreover, CENPF knockdown was revealed to decrease Vimentin and increase E-cadherin levels in glioma cells, and significantly reduce the size and mass of tumors in vivo. Overall, the present study identified new clinical biomarkers and revealed that CENPF may promote glioma progression by regulating the epithelial-mesenchymal transition pathway. By elucidating the complexities of glioma and identifying prognostic biomarkers, the present research enables further improvement of patient outcomes and the advancement of precision medicine for this disease.
Collapse
Affiliation(s)
- Jia Li
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
- Department of Emergency Surgery, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui 236112, P.R. China
| | - Lei Liu
- Department of Emergency Surgery, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui 236112, P.R. China
| | - Gang Zong
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Zhihao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Deran Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
12
|
Zhu C, Liu H, Li Z, Shi Y, Zhao J, Bai Y, Chen Q, Li W. Prognostic Significance and Therapeutic Potential of SERPINE1 in Head and Neck Squamous Cell Carcinoma. Cancer Med 2025; 14:e70605. [PMID: 39817507 PMCID: PMC11736624 DOI: 10.1002/cam4.70605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/24/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND This study aims to elucidate the expression pattern of SERPINE1, assess its prognostic significance, and explore potential therapeutic drugs targeting this molecule. METHODS AND RESULTS In this study, we delved into the variations in gene mutation, methylation patterns, and expression levels of SERPINE1 in head and neck squamous cell carcinoma (HNSCC) and normal tissues, leveraging comprehensive analyses of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. The connection between the biological function of the gene and prognosis was scrutinized through immune infiltration and enrichment analyses. Concurrently, we assessed the potential therapeutic value of SERPINE1 through drug sensitivity analysis. It was observed that, particularly in human papillomavirus (HPV) negative HNSCC, SERPINE1 exhibited elevated expression levels, correlating with poorer prognosis. The infiltration levels of eight cell types, such as eosinophils, Tgd, and macrophages, showed a positive correlation with SERPINE1 expression, whereas infiltration levels of four cell types, including cytotoxic cells, B cells, and pDCs, displayed a negative correlation. Furthermore, copy number variations of SERPINE1 were primarily characterized by homologous amplification, positively correlating with its expression, while methylation showed an inverse correlation. The outcomes of drug sensitivity analysis underscored the potential of SERPINE1 as a therapeutic target. CONCLUSION Elevated expression of SERPINE1 in HNSCC is intricately linked with adverse prognostic outcomes and has the potential to influence the immune microenvironment. Subsequent investigations are imperative to fully elucidate the prognostic implications of SERPINE1 as a biomarker and to unlock its therapeutic promise as a target for intervention.
Collapse
Affiliation(s)
- Changyu Zhu
- Cancer CenterBeijing Tongren Hospital, Capital Medical UniversityBeijingChina
| | - Heshu Liu
- Cancer CenterBeijing Tongren Hospital, Capital Medical UniversityBeijingChina
| | - Zhixin Li
- Cancer CenterBeijing Tongren Hospital, Capital Medical UniversityBeijingChina
| | - Yijun Shi
- Cancer CenterBeijing Tongren Hospital, Capital Medical UniversityBeijingChina
| | - Jingyang Zhao
- Cancer CenterBeijing Tongren Hospital, Capital Medical UniversityBeijingChina
| | - Yuping Bai
- Department of PathologyBeijing Tongren Hospital, Capital Medical University, Beijing Key Laboratory of Head and Neck Molecular Diagnostic PathologyBeijingChina
| | | | - Wei Li
- Cancer CenterBeijing Tongren Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
13
|
Khan J, Ghosh P, Bajpai U, Dwivedi K, Saluja D. Integrated analysis of cell cycle and p53 signaling pathways related genes in breast, colorectal, lung, and pancreatic cancers: implications for prognosis and drug sensitivity for therapeutic potential. Discov Oncol 2024; 15:832. [PMID: 39715832 PMCID: PMC11666898 DOI: 10.1007/s12672-024-01712-8] [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: 08/21/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024] Open
Abstract
Cancer, a leading cause of death worldwide, is projected to increase by 76.6% in new cases and 89.7% in mortality by 2050 (WHO 2022). Among various types, lung cancer is the most prevalent with high morbidity, while breast, colorectal, and pancreatic cancers also show high mortality rates. Cancer progression often involves disruption in cell cycle regulation and signaling pathways, with mutations in genes like TP53, EGFR, and K-RAS playing significant roles. In this study, we analyzed gene expression datasets to identify common molecular signatures across breast, colorectal, lung and pancreatic cancers. Our focus was on genes related to cell cycle regulation and p53 signaling pathway, intending to discover potential biomarkers for improved diagnosis and treatment strategies. The study analyzed GEO datasets; GSE45827, GSE9348, GSE30219, and GSE62165 for breast, colorectal, lung, and pancreatic cancers respectively. Differentially expressed genes (DEGs) were identified using GEO2R, and functional annotation and pathway analysis were performed using WebGestalt. Common cell cycle and p53 signaling genes were acquired from MSigDB using GSEA. A protein-protein interaction network was constructed using STRING and Cytoscape, identifying top hub genes. Validation of hub genes at mRNA and protein levels was done via GEPIA2 and Human Protein Atlas. Survival analysis was conducted using TCGA data by GEPIA2 and LASSO, and drug sensitivity was analyzed with the GSCA drug bank database, highlighting potential therapeutic targets. The study identified 411 common DEGs among these four cancers. Pathway and functional enrichment revealed key biological processes and pathways like p53 signaling, and cell cycle. The intersection of these DEGs with genes involved in cell cycle and p53 signaling, identified 23 common genes that were used for constructing a PPI network. The top 10 hub genes were validated both for mRNA and protein expression, revealing they are significantly overexpressed in all studied cancers. Prognostic relevance showed that MCM4, MCM6, CCNA2, CDC20, and CHEK1 are associated with survival. Additionally, drug sensitivity analysis highlighted key gene-drug interactions, suggesting potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jiyauddin Khan
- Dr B R Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Priyanjana Ghosh
- Dr B R Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Urmi Bajpai
- Department of Biomedical Sciences, Acharya Narendra Dev College University of Delhi, University of Delhi, New Delhi, 110019, India
| | - Kountay Dwivedi
- Center for Clinical Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Daman Saluja
- Dr B R Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India.
- Department of Allied and Basic Sciences, Shri Guru Gobind Singh Tricentenary University, Gurugram, 122505, Haryana, India.
| |
Collapse
|
14
|
Guo S, Yu Y, Bu Y, Ren J, Zhang L, Ma X, Li J, Li A, Liu X. Pan-cancer analysis reveals the prognostic and immunomodulatory potential of super-enhancer-induced ANGPT2 and experimental validation in colorectal cancer. Clin Transl Oncol 2024:10.1007/s12094-024-03818-5. [PMID: 39695006 DOI: 10.1007/s12094-024-03818-5] [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: 08/01/2024] [Accepted: 11/23/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND ANGPT2 plays important roles in cancer development. However, there is still no systematic analysis of ANGPT2 in pan-cancer. METHODS In this paper, we conducted a pan-cancer analysis to investigate the characteristics of ANGPT2. Furthermore, we investigated the impact of genetic and epigenetic factors on ANGPT2 expression by bioinformatics and assays. RESULTS By several TCGA and GEO databases, we identified elevated expression of ANGPT2 in various tumor types. Besides, high expression of ANGPT2 induced poor prognosis of patients in multiple tumors. Through enrichment analysis, we found that ANGPT2 participated in various biological processes, including angiogenesis and immunity. Various immune analyses indicated that high expression of ANGPT2 might suggest a propensity towards a hot tumor microenvironment, but its impact on immunotherapy was negative. Bioinformatics analysis and experiments confirmed that genetic and epigenetic factors explained part of the mechanism behind ANGPT2 abnormal expression. Finally, we screened candidate drugs targeting the ANGPT2 protein by molecular docking and molecular dynamics simulation. CONCLUSION ANGPT2 has diagnostic and prognostic values in multiple tumor types. Though with a hot tumor microenvironment, ANGPT2 high expression patients are not suitable for immunotherapy because of its proangiogenic function, contributing to selecting the exact patients for immunotherapy. Both genetic and epigenetic factors influenced ANGPT2 expression, with the influence of super-enhancer being more pronounced. This paper for the first time did the systematic analysis of ANGPT2 and showed its characteristic in pan-cancer. We summarized the biomarker role of ANGPT2 on tumor diagnosis and prognosis, as well as its target role on tumor immunotherapy.
Collapse
Affiliation(s)
- Songyue Guo
- Department of Oncology, School of Clinical Medicine, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Yanan Yu
- Department of Oncology, School of Clinical Medicine, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Yulan Bu
- Department of Radiotherapy, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Jiao Ren
- Department of Oncology, School of Clinical Medicine, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Lu Zhang
- Department of Oncology, School of Clinical Medicine, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Xiaolin Ma
- Department of Oncology, School of Clinical Medicine, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China
| | - Jiaqiu Li
- Department of Oncology, School of Clinical Medicine, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China.
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China.
| | - Ao Li
- Department of Oncology, School of Clinical Medicine, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China.
| | - Xinling Liu
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, 261053, Shandong, China.
| |
Collapse
|
15
|
Chen X, Zhao J, Shu J, Ying X, Khan S, Sarfaraz S, Mirzaeiebrahimabadi R, Alhomrani M, Alamri AS, ALSuhaymi N. Exploring potential key genes and pathways associatedwith hepatocellular carcinoma prognosis through bioinformatics analysis, followed by experimental validation. Am J Transl Res 2024; 16:7286-7302. [PMID: 39822558 PMCID: PMC11733333 DOI: 10.62347/wier4743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/10/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND Liver Hepatocellular Carcinoma (LIHC) is a prevalent and aggressive liver cancer with limited therapeutic options. Identifying key genes involved in LIHC can enhance our understanding of its molecular mechanisms and aid in the development of targeted therapies. This study aims to identify differentially expressed genes (DEGs) and key hub genes in LIHC using bioinformatics approaches and experimental validation. METHOD We analyzed two LIHC-related datasets, GSE84598 and GSE19665, from the Gene Expression Omnibus (GEO) database to identify DEGs. Differential expression analysis was performed using the limma package in R to identify DEGs between cancerous and non-cancerous liver tissues. A Protein-Protein Interaction (PPI) network was constructed using STRING to determine key hub genes. Further validation of these hub genes was conducted through UALCAN, OncoDB, and the Human Protein Atlas (HPA) databases for mRNA and protein expression levels. Promoter methylation and mutational analyses were performed using cBioPortal. Kaplan-Meier survival analysis assessed the impact of hub gene expression on patient survival. Correlations with immune cell abundance and drug sensitivity were explored using GSCA. Finally, AURKA was knocked down in HepG2 cells, and cell proliferation, colony formation, and wound healing assays were performed. RESULTS Analysis identified 180 DEGs, with four key hub genes, including AURKA, BUB1B, CCNA2, and PTTG1 showing significant overexpression and hypomethylation in LIHC tissues. AURKA knockdown in HepG2 cells led to decreased cell proliferation, reduced colony formation, and impaired wound healing, confirming its role in LIHC progression. These hub genes were also hypomethylated and their elevated expression correlated with poor overall survival. CONCLUSION AURKA, BUB1B, CCNA2, and PTTG1 are crucial for LIHC pathogenesis and may serve as potential biomarkers or therapeutic targets. Our findings provide new insights into LIHC mechanisms and suggest promising avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Xi Chen
- Department of Oncology, Jingdezhen First People’s HospitalJindezhen 333000, Jiangxi, China
| | - Jianhua Zhao
- Department of Oncology, Jingdezhen First People’s HospitalJindezhen 333000, Jiangxi, China
| | - Jiaming Shu
- Department of Oncology, Jingdezhen First People’s HospitalJindezhen 333000, Jiangxi, China
| | - Xueming Ying
- Department of Oncology, Jingdezhen First People’s HospitalJindezhen 333000, Jiangxi, China
| | | | - Sara Sarfaraz
- Department of Bioinformatics, Faculty of Biomedical and Life Sciences, Kohsar University MurreePakistan
| | - Reza Mirzaeiebrahimabadi
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhou, Henan, China
| | - Majid Alhomrani
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif UniversityTaif, Saudi Arabia
- Research Centre for Health Sciences, Taif UniversityTaif, Saudi Arabia
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif UniversityTaif, Saudi Arabia
- Research Centre for Health Sciences, Taif UniversityTaif, Saudi Arabia
| | - Naif ALSuhaymi
- Department of Emergency Medical Services, Faculty of Health Sciences AlQunfudah, Umm Al-Qura UniversityMekkah, Saudi Arabia
| |
Collapse
|
16
|
Wang Y, Xu S, Liu J, Qi P. A Novel Peroxisome-Related Gene Signature Predicts Breast Cancer Prognosis and Correlates with T Cell Suppression. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:887-911. [PMID: 39678026 PMCID: PMC11639899 DOI: 10.2147/bctt.s490154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024]
Abstract
Background Peroxisomes are increasingly linked to cancer development, yet the prognostic role of peroxisome-related genes (PRGs) in breast cancer remains unclear. Objective This study aimed to construct a prognostic model based on PRG expression in breast cancer to clarify their prognostic value and clinical implications. Methods Transcriptomic data from TCGA and GEO were used for training and validation cohorts. TME characteristics were analyzed with ESTIMATE, MCP-counter, and CIBERSORT algorithms. qPCR validated mRNA expression levels of risk genes, and data analysis was conducted in R. Results Univariate and multivariate Cox regression identified a 7-gene PRG risk signature (ACBD5, ACSL5, DAO, NOS2, PEX3, PEX10, and SLC27A2) predicting breast cancer prognosis in training (n=1069), internal validation (n=327), and external validation (merged from four GEO datasets, n=640) datasets. While basal and Her2 subtypes had higher risk scores than luminal subtypes, a significant prognostic impact of the PRG risk signature was seen only in luminal subtypes. The high-risk subgroup exhibited a higher frequency of focal synonymous copy number alterations (SCNAs), arm-level amplifications and deletions, and single nucleotide variations. These increased genomic aberrations were associated with greater immune suppression and reduced CD8+ T cell infiltration. Bulk RNA sequencing and single-cell analyses revealed distinct expression patterns of peroxisome-related genes (PRGs) in the breast cancer TME: PEX3 was primarily expressed in malignant and stromal cells, while ACSL5 showed high expression in T cells. Additionally, the PRG risk signature demonstrated efficacy comparable to that of well-known biomarkers for predicting immunotherapy responses. Drug sensitivity analysis revealed that the PRG high-risk subgroup was sensitive to inhibitors of BCL-2 family proteins (BCL-2, BCL-XL, and MCL1) and other kinases (PLK1, PLK1, BTK, CHDK1, and EGFR). Conclusion The PRG risk signature serves as a promising biomarker for evaluating peroxisomal activity, prognosis, and responsiveness to immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Yunxiang Wang
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| | - Sheng Xu
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| | - Junfeng Liu
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| | - Pan Qi
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| |
Collapse
|
17
|
Lu J, Yang L, Yang X, Chen B, Liu Z. Investigating the clinical significance of OAS family genes in breast cancer: an in vitro and in silico study. Hereditas 2024; 161:50. [PMID: 39633486 PMCID: PMC11619215 DOI: 10.1186/s41065-024-00353-9] [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: 09/17/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Breast cancer is the most common malignancy among women worldwide, characterized by complex molecular and cellular heterogeneity. Despite advances in diagnosis and treatment, there is an urgent need to identify reliable biomarkers and therapeutic targets to improve early detection and personalized therapy. The OAS (2'-5'-oligoadenylate synthetase) family genes, known for their roles in antiviral immunity, have emerged as potential regulators in cancer biology. This study aimed to explore the diagnostic and functional relevance of OAS family genes in breast cancer. METHODOLOGY Breast cancer cell lines and controls were cultured under specific conditions, and DNA and RNA were extracted for downstream analyses. RT-qPCR, bisulfite sequencing, and Western blotting were employed to assess gene expression, promoter methylation, and knockdown efficiency of OAS family genes. Functional assays, including CCK-8, colony formation, and wound healing, evaluated cellular behaviors, while bioinformatics tools (UALCAN, GEPIA, HPA, OncoDB, cBioPortal, and others) validated findings and explored correlations with clinical data. RESULTS The OAS family genes (OAS1, OAS2, OAS3, and OASL) were found to be significantly upregulated in breast cancer cell lines and tissues compared to normal controls. This overexpression was strongly associated with reduced promoter methylation. Receiver operating characteristic (ROC) analysis demonstrated high diagnostic accuracy, with area under the curve (AUC) values exceeding 0.93 for all four genes. Increased OAS expression correlated with advanced cancer stages and poor overall survival in breast cancer patients. Functional analysis revealed their involvement in critical biological processes, including immune modulation and oncogenic pathways. Silencing OAS genes in breast cancer cells significantly inhibited cell proliferation and colony formation, while unexpectedly enhancing migratory capacity. Additionally, correlations with immune cell infiltration, molecular subtypes, and drug sensitivity highlighted their potential roles in the tumor microenvironment and therapeutic response. CONCLUSION The findings of this study established OAS family genes as potential biomarkers and key players in breast cancer progression, offering promise as diagnostic biomarkers and therapeutic targets to address unmet clinical needs.
Collapse
Affiliation(s)
- Jinjun Lu
- Department of General Surgery, Nantong Haimen People's Hospital, NanTong, JiangSu, 226100, China
| | - Lu Yang
- Department of Clinical Laboratory, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Xinghai Yang
- Department of General Surgery, Nantong Haimen People's Hospital, NanTong, JiangSu, 226100, China
| | - Bin Chen
- Department of General Surgery, Nantong Haimen People's Hospital, NanTong, JiangSu, 226100, China
| | - Zheqi Liu
- Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310000, China.
| |
Collapse
|
18
|
Xu C, Lu Z, Hou G, Zhu M. Exploring the function and prognostic value of RPLP0, RPLP1 and RPLP2 expression in lung adenocarcinoma. J Mol Histol 2024; 55:1079-1091. [PMID: 39207634 PMCID: PMC11567986 DOI: 10.1007/s10735-024-10251-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Lung adenocarcinoma (LUAD) is the most common subtype of non-small cell lung cancer (NSCLC) and is characterized by its heterogeneity and poor prognosis. The role of ribosomal proteins RPLP0, RPLP1 and RPLP2 in multiple cancers has been implicated. However, their function in LUAD and their correlation with the poor prognosis of LUAD remains elusive. In this study, we performed a comprehensive bioinformatic analysis of the impact of these ribosomal proteins on LUAD. Our findings reveal that RPLP0, RPLP1 and RPLP2 are overexpressed in LUAD, which are likely attributed to abnormal copy number variations and decreased methylation levels of their promoters. LUAD patients with high expression of RPLP0, RPLP1 or RPLP2 have worse clinical outcomes in terms of overall survival (OS), first progression (FP) and post-progression survival (PPS), indicating poor prognosis. Moreover, the expression of RPLP0, RPLP1 and RPLP2 affects immune cell infiltration in LUAD tissues. Finally, we identified multiple existing drugs that may inhibit the expression of RPLP1 and RPLP2. Collectively, our data implicate the oncogenic role of RPLP0, RPLP1 and RPLP2 in LUAD and underscore their prognostic value in LUAD patients.
Collapse
Affiliation(s)
- Chunyan Xu
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, China
| | - Zhimin Lu
- Department of Outpatient, The Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, China
| | - Guoxin Hou
- Department of Oncology, The Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, China.
| | - Moran Zhu
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, China.
| |
Collapse
|
19
|
Xia T, Zhang Y, Peng H, Jia X, Yang D, Wei L, Li T, Yao W. EVA1B facilitates esophageal squamous carcinoma progression and recruitment of immunosuppressive myeloid-derived suppressor cells in the tumor microenvironment. Pharmacol Res 2024; 210:107521. [PMID: 39603573 DOI: 10.1016/j.phrs.2024.107521] [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: 09/07/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
Eva-1 Homolog B (EVA1B) has been preliminarily found to be associated with prognostic outcomes and immune microenvironment in several human cancer types, but the implications of EVA1B in ESCC remain unclear. Human ESCC and paracancerous tissues were gathered in this study, and EVA1B expression was measured via immunoblotting. EC109 and KYSE-180 ESCC cells were stably infected by sh-EVA1B lentivirus, and functional experiments were subsequently implemented. Syngeneic mouse models were built, and the expansion and recruitment of myeloid-derived suppressor cells (MDSCs) were then evaluated. The results showed that EVA1B presented the notable up-regulation in clinical ESCC tissues versus controls, and was connected to more advanced stages and the abundance of MDSCs. Silencing EVA1B notably attenuated proliferation of ESCC cells and tumor growth in syngeneic mouse models. Moreover, EVA1B suppression resulted in apoptosis and cell cycle arrest, and impaired ESCC cell aggressiveness. Among ESCC patients, EVA1B was strongly correlated to EMT pathway activity. Targeted suppression of EVA1B mitigated the expression of Wnt3a, β-catenin and LRP6 in ESCC cells and tumor xenografts. Additionally, inhibition of EVA1B attenuated the expansion and recruitment of MDSCs within the immune microenvironment based upon the reduction in the percentage of CD11b+Gr-1+ immunosuppressive MDSCs as well as the expression of MDSC expansion stimulators (S100A8, S100A9, Arg-1, and VEGF). Collectively, our findings unveiled the contribution of high expression of EVA1B to ESCC progression and MDSCs expansion and recruitment, indicating that targeted suppression of EVA1B may be a potential treatment choice for ESCC patients.
Collapse
Affiliation(s)
- Tian Xia
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China.
| | - Yongkang Zhang
- Department of Thoracic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Haodong Peng
- Department of Thoracic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Xiangbo Jia
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China.
| | - Dong Yang
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China.
| | - Li Wei
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China.
| | - Tian Li
- Tianjin Medical University, Tianjin 300102, China.
| | - Wenjian Yao
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China.
| |
Collapse
|
20
|
Wang Z, Chen H, Sun L, Wang X, Xu Y, Tian S, Liu X. Uncovering the potential of APOD as a biomarker in gastric cancer: A retrospective and multi-center study. Comput Struct Biotechnol J 2024; 23:1051-1064. [PMID: 38455068 PMCID: PMC10918487 DOI: 10.1016/j.csbj.2024.02.015] [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: 10/21/2023] [Revised: 02/16/2024] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
Gastric cancer (GC) poses a significant health challenge worldwide, necessitating the identification of predictive biomarkers to improve prognosis. Dysregulated lipid metabolism is a well-recognized hallmark of tumorigenesis, prompting investigation into apolipoproteins (APOs). In this study, we focused on apolipoprotein D (APOD) following comprehensive analyses of APOs in pan-cancer. Utilizing data from the TCGA-STAD and GSE62254 cohorts, we elucidated associations between APOD expression and multiple facets of GC, including prognosis, tumor microenvironment (TME), cancer biomarkers, mutations, and immunotherapy response, and identified potential anti-GC drugs. Single-cell analyses and immunohistochemical staining confirmed APOD expression in fibroblasts within the GC microenvironment. Additionally, we independently validated the prognostic significance of APOD in the ZN-GC cohort. Our comprehensive analyses revealed that high APOD expression in GC patients was notably associated with unfavorable clinical outcomes, reduced microsatellite instability and tumor mutation burden, alterations in the TME, and diminished response to immunotherapy. These findings provide valuable insights into the potential prognostic and therapeutic implications of APOD in GC.
Collapse
Affiliation(s)
- Zisong Wang
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Hongshan Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Le Sun
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xuanyu Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Yihang Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Sufang Tian
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xiaoping Liu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
21
|
Zheng S, Liu Q, Wang C, Zhang R, Peng X, Fan J, Xu H, Pan X, Chen N, Liu M, Zhao K. Multi-omics analysis of pyroptosis-related genes for prognosis and immune landscape in head and neck cancer. Clin Transl Med 2024; 14:e70144. [PMID: 39690139 DOI: 10.1002/ctm2.70144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024] Open
Affiliation(s)
- Shikang Zheng
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | - Qinghua Liu
- Medical School of Chinese PLA, Haidian, Beijing, China
| | - Cheng Wang
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | | | - Xin Peng
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | - Junda Fan
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | - Haiming Xu
- Medical School of Chinese PLA, Haidian, Beijing, China
| | - Xiazhi Pan
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Nanxiang Chen
- Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, Beijing, China
| | - Mingbo Liu
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
- Medical School of Chinese PLA, Haidian, Beijing, China
- Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, Beijing, China
| | - Kai Zhao
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| |
Collapse
|
22
|
Meng Y, Meng Y, Zheng H, Huo J, Li P, Shan Y, He J. METTL2B m3C RNA transferase: oncogenic role in ovarian cancer progression via regulation of the mTOR/AKT pathway and its link to the tumor immune microenvironment. BMC Cancer 2024; 24:1455. [PMID: 39592997 PMCID: PMC11600782 DOI: 10.1186/s12885-024-13225-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Aberrant expression of N3-methylcytidine methyltransferase 2B (METTL2B) has been observed in various human malignancies, including those of the prostate, liver, breasts, and bladder. However, its role in ovarian cancer (OC) remains largely unexplored. This research preliminarily investigated METTL2B expression in OC and elucidated the associated molecular mechanisms. METHODS We utilized three publicly available cancer-related databases (Genotype-Tissue Expression, Gene Expression Omnibus, and The Cancer Genome Atlas) to identify gene signatures in patients with OC and normal individuals with a specific focus on METTL2B. The role of METTL2B in OC was evaluated using patient survival data, and its impact on oncogenic behaviors in both cell and animal models, including growth potential, migration, invasion, and the tumor microenvironment, was examined. This assessment was conducted using bioinformatics tools such as Gene Set Cancer Analysis, GeneMANIA, and Tumor Immune Single-cell Hub 2. Additionally, the association between drug sensitivity and METTL2B expression was analyzed using CellMiner. RESULTS METTL2B expression was significantly elevated in OC, highlighting its potential clinical value in the diagnosis and prognosis of OC. Patients with lower METTL2B expression exhibited favorable survival. Furthermore, METTL2B knockdown significantly disrupted oncogenic behaviors in OC cell lines by suppressing the mTOR/AKT signaling pathway. Additionally, bioinformatics-based Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses suggested a close correlation between METTL2B and immune responses. CONCLUSIONS Our research confirmed the upregulation of METTL2B in OC, suggesting its oncogenic function. However, METTL2B expression was negatively correlated with the infiltration scores of multiple immune cells, including cytotoxic cells and T cells, indicating its complex role in the tumor immune microenvironment. These findings highlight the significant clinical value of METTL2B in the diagnosis and prognosis of OC.
Collapse
Affiliation(s)
- Yizi Meng
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, No. 1 Xinmin Road, Changchun, Jilin, 130000, China
| | - Yimei Meng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, No 246, Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Hui Zheng
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, No. 1 Xinmin Road, Changchun, Jilin, 130000, China
| | - Jinru Huo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, No 246, Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Peiling Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, No 246, Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Yanhong Shan
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, No. 1 Xinmin Road, Changchun, Jilin, 130000, China.
| | - Jin He
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, No. 1 Xinmin Road, Changchun, Jilin, 130000, China.
| |
Collapse
|
23
|
Wang F, Huang Z, Li J, Gao X. Clinical relevance and function of HMGB1 gene polymorphism and expression in colorectal cancer. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024:1-11. [PMID: 39589167 DOI: 10.1080/15257770.2024.2432991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/10/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024]
Abstract
High HMGB1 levels contribute to the development and metastasis of tumors such as colorectal cancer (CRC). The current investigation sought to evaluate the association of a functional InDel polymorphism (rs34000982) on the HMGB1 gene with CRC susceptibility and tumor stage and the clinical relevance of HMGB1 gene expression. A total of 600 CRC patients and 600 healthy control individuals were genotyped by a polymerase chain reaction-polyacrylamide gel electrophoresis assay. The findings demonstrated that the rs34000982 Ins allele or Ins/Ins genotype was associated not only with reduced susceptibility to CRC, especially stage III-IV CRC (Ins vs. Del: OR = 0.65, 95%CI = 0.51-0.82, p < 0.001; Ins/Ins vs. Del/Del: OR = 0.29, 95%CI = 0.14- 0.60, p < 0.001), but also with tumor stage. CRC patients carrying the Ins allele or Ins/Ins genotype had a significantly lower risk of stage III-IV tumors (Ins vs. Del: OR = 0.69, 95%CI = 0.53- 0.91; Ins/Ins vs. Del/Del: OR = 0.41, 95%CI = 0.18-0.94). Functional research revealed that the rs34000982 Ins allele enabled hsa-miR-944 to interact with the 3' untranslated region of HMGB1. In addition, HMGB1 gene expression levels were associated not only with multiple immune cell infiltration, but also with multiple anti-CRC drug sensitivities. The current findings suggest that the HMGB1 rs34000982 polymorphism may serve as a marker of CRC susceptibility and progression in the Chinese population, and HMGB1 levels may serve as an anti-CRC drug sensitivity marker.
Collapse
Affiliation(s)
- Fang Wang
- School of Pharmacy, Yancheng Teachers' University, Jiangsu, China
| | - Zhijun Huang
- Department of Surgery, Yancheng First People's Hospital, Jiangsu, China
| | - Jianping Li
- Department of Oncology, Yancheng First People's Hospital, Jiangsu, China
| | - Xueren Gao
- School of Pharmacy, Yancheng Teachers' University, Jiangsu, China
| |
Collapse
|
24
|
Ferrari I, De Grossi F, Lai G, Oliveto S, Deroma G, Biffo S, Manfrini N. CancerHubs: a systematic data mining and elaboration approach for identifying novel cancer-related protein interaction hubs. Brief Bioinform 2024; 26:bbae635. [PMID: 39657701 PMCID: PMC11631132 DOI: 10.1093/bib/bbae635] [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: 08/14/2024] [Revised: 10/31/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
Conventional approaches to predict protein involvement in cancer often rely on defining either aberrant mutations at the single-gene level or correlating/anti-correlating transcript levels with patient survival. These approaches are typically conducted independently and focus on one protein at a time, overlooking nucleotide substitutions outside of coding regions or mutational co-occurrences in genes within the same interaction network. Here, we present CancerHubs, a method that integrates unbiased mutational data, clinical outcome predictions and interactomics to define novel cancer-related protein hubs. Through this approach, we identified TGOLN2 as a putative novel broad cancer tumour suppressor and EFTUD2 as a putative novel multiple myeloma oncogene.
Collapse
Affiliation(s)
- Ivan Ferrari
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Federica De Grossi
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giancarlo Lai
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Stefania Oliveto
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giorgia Deroma
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Stefano Biffo
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Nicola Manfrini
- INGM, Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| |
Collapse
|
25
|
Li X, Xu J, Li J, Gu J, Shang X. Towards simplified graph neural networks for identifying cancer driver genes in heterophilic networks. Brief Bioinform 2024; 26:bbae691. [PMID: 39751645 PMCID: PMC11697181 DOI: 10.1093/bib/bbae691] [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: 11/09/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
The identification of cancer driver genes is crucial for understanding the complex processes involved in cancer development, progression, and therapeutic strategies. Multi-omics data and biological networks provided by numerous databases enable the application of graph deep learning techniques that incorporate network structures into the deep learning framework. However, most existing methods do not account for the heterophily in the biological networks, which hinders the improvement of model performance. Meanwhile, feature confusion often arises in models based on graph neural networks in such graphs. To address this, we propose a Simplified Graph neural network for identifying Cancer Driver genes in heterophilic networks (SGCD), which comprises primarily two components: a graph convolutional neural network with representation separation and a bimodal feature extractor. The results demonstrate that SGCD not only performs exceptionally well but also exhibits robust discriminative capabilities compared to state-of-the-art methods across all benchmark datasets. Moreover, subsequent interpretability experiments on both the model and biological aspects provide compelling evidence supporting the reliability of SGCD. Additionally, the model can dissect gene modules, revealing clearer connections between driver genes in cancers. We are confident that SGCD holds potential in the field of precision oncology and may be applied to prognosticate biomarkers for a wide range of complex diseases.
Collapse
Affiliation(s)
- Xingyi Li
- School of Computer Science, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518063 Guangdong, China
- Faculty of Data Science, City University of Macau, Macau, 999078 Macau, China
| | - Jialuo Xu
- School of Computer Science, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi, China
| | - Junming Li
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518063 Guangdong, China
- School of Software, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi, China
| | - Jia Gu
- School of Software, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi, China
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi, China
| |
Collapse
|
26
|
Gao S, Zhang L, Sun G. GLN2 as a key biomarker and therapeutic target: evidence from a comprehensive pan-cancer study using molecular, functional, and bioinformatic analyses. Discov Oncol 2024; 15:681. [PMID: 39562405 DOI: 10.1007/s12672-024-01516-w] [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: 07/25/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND GNL2, a nuclear protein, is involved in ribosome production and cell cycle regulation. However, its expression and function in different types of tumors are not well understood. Comprehensive studies across multiple cancer types are needed to assess the potential of GNL2 as a diagnostic, prognostic, and immunological marker. METHODS mRNA expression data, copy number alteration threshold data, masked copy number segmentation data, and DNA methylation 450 K data from The Cancer Genome Atlas (TCGA) pan-cancer cohort were obtained from the Firehose database. Additional data, including miRNA, The Cancer Proteome Atlas (TCPA), mutation data, and clinical information, were sourced from the University of California Santa Cruz (UCSC) Xena database. The cBioPortal database facilitates the examination of GNL2 mutation frequency, location, and 3D structure in the TCGA database. Gene Expression Omnibus (GEO) data verified the transcriptome level expression in the TCGA cohort. Protein expression levels were analyzed via the Human Protein Atlas (HPA) database and the Clinical Proteomic Tumor Analysis Consortium (CPTAC) database. Gene set enrichment analysis (GSEA) was employed to investigate the biological role of GNL2 across cancers. Multiple immune infiltration algorithms from the TIMER2.0 database were utilized to examine the correlation between GNL2 expression and the tumor immune microenvironment. The transcriptome-wide immune infiltration results were validated using 72 single-cell datasets from the Tumor Immune Single-cell Hub (TISCH) database. Pan-cancer survival maps were constructed, and GNL2 expression in different molecular subtypes across cancers was examined. The relationship between GNL2 and drug resistance was investigated using data from CellMiner, GDSC, and CTRP. The Comparative Toxicogenomics Database (CTD) was used to identify chemicals affecting GNL2 expression. RESULTS GNL2 is located primarily in the nucleus, and its expression is regulated mainly through somatic copy number alteration (SCNA) and aberrant DNA methylation, according to TCGA data. Database analysis and immunohistochemical results from clinical samples revealed high GNL2 expression in most tumors, which was correlated with diagnostic significance. High GNL2 expression often indicates a poor prognosis with pan-cancer prognostic value. Gene set enrichment analysis (GSEA) suggested that GNL2 is involved in tumor development through cell proliferation-related pathways. GNL2 expression is correlated with the expression of immune-related genes and the infiltration levels of multiple immune cells. The relationships between GNL2 and various drugs and chemicals were examined, revealing its influence on drug sensitivity and identifying five chemicals countering GNL2-mediated pro-cancer effects. CONCLUSION Comprehensive bioinformatics analysis of GNL2 in pan-cancer tissues, combined with experimental validation, elucidated the pan-cancer expression pattern of GNL2, determined its diagnostic and prognostic value, and explored the biological functions of GNL2. GNL2 may be involved in the regulation of cell cycle progression and remodeling of the tumor microenvironment and is associated with poor prognosis as a risk factor in most tumors. The potential of GNL2-based cancer therapies is emphasized, assisting in predicting the response to chemotherapy.
Collapse
Affiliation(s)
- Shuang Gao
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230001, Anhui, China
- Ward 4 of the Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Lei Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Bengbu Medical University, Bengbu, 233080, Anhui, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230001, Anhui, China.
| |
Collapse
|
27
|
Shakoori A, Hosseinzadeh A, Nafisi N, Omranipour R, Sahebi L, Nazanin Hosseinkhan, Ahmadi M, Ghafouri-Fard S, Abtin M. Importance of LINC00852/miR-145-5p in breast cancer: a bioinformatics and experimental study. Discov Oncol 2024; 15:672. [PMID: 39557729 PMCID: PMC11574217 DOI: 10.1007/s12672-024-01553-5] [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: 09/07/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
PURPOSE We aimed to examine the importance of an lncRNA, namely LINC00852, in the pathogenesis of breast cancer. MATERIALS AND METHODS In the current study, we used several online tools to examine the importance of LINC00852 in breast cancer. Then, we examined these findings in 50 pairs of breast cancer tissues and adjacent non-cancerous ones. We also re-evaluated the data of miR-145-5p signature from our recent study. RESULTS While in silico tools revealed down-regulation of LINC00852 in breast cancer samples, expression assays showed significant up-regulation of this lncRNAs in breast cancer samples compared with matching control samples from Iranian patients. miR-145-5p was under-expressed in breast cancer samples compared with non-cancerous samples. LINC00852 could separate breast cancer tissues from adjacent non-malignant tissues with an AUC value of 0.7218 (P value < 0.001). CONCLUSION The current study potentiates LINC00852/miR-145-5p axis as a possible contributor to the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Abbas Shakoori
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, Cancer Institute of Iran, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Dr. Qarib St., Keshavarz Blvd, Tehran, Iran
| | | | - Nahid Nafisi
- Surgery Department, Rasoul Akram Hospital, Clinical Research Development Center (RCRDC), Iran University of Medical Sciences, Tehran, Iran
| | - Ramesh Omranipour
- Breast Disease Research Center (BDRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Surgical Oncology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leyla Sahebi
- Maternal, Fetal and Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin Hosseinkhan
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mohsen Ahmadi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maryam Abtin
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Su Q, Sun H, Mei L, Yan Y, Ji H, Chang L, Wang L. Ribosomal proteins in hepatocellular carcinoma: mysterious but promising. Cell Biosci 2024; 14:133. [PMID: 39487553 PMCID: PMC11529329 DOI: 10.1186/s13578-024-01316-3] [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: 07/09/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
Ribosomal proteins (RPs) are essential components of ribosomes, playing a role not only in ribosome biosynthesis, but also in various extra-ribosomal functions, some of which are implicated in the development of different types of tumors. As universally acknowledged, hepatocellular carcinoma (HCC) has been garnering global attention due to its complex pathogenesis and challenging treatments. In this review, we analyze the biological characteristics of RPs and emphasize their essential roles in HCC. In addition to regulating related signaling pathways such as the p53 pathway, RPs also act in proliferation and metastasis by influencing cell cycle, apoptosis, angiogenesis, and epithelial-to-mesenchymal transition in HCC. RPs are expected to unfold new possibilities for precise diagnosis and individualized treatment of HCC.
Collapse
Affiliation(s)
- Qian Su
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Huizhen Sun
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Ling Mei
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Ying Yan
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Huimin Ji
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China
| | - Le Chang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China.
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China.
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China.
| | - Lunan Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/ National Center of Gerontology, Beijing, P.R. China.
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, P.R. China.
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China.
| |
Collapse
|
29
|
Su Z, He Y, You L, Chen J, Zhang G, Liu Z. SPP1+ macrophages and FAP+ fibroblasts promote the progression of pMMR gastric cancer. Sci Rep 2024; 14:26221. [PMID: 39482333 PMCID: PMC11528032 DOI: 10.1038/s41598-024-76298-w] [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: 02/12/2024] [Accepted: 10/11/2024] [Indexed: 11/03/2024] Open
Abstract
Immunotherapy has become a primary and secondary treatment for gastric cancer (GC) patients with mismatch repair deficiency (dMMR), and is used in both perioperative and advanced stages. The tumor immune microenvironment (TiME) is crucial for immunotherapy efficacy, yet the impact of MMR status on TiME remains understudied. We employed single-cell RNA sequencing (scRNA-seq) to analyze 33 fresh tissue samples from 25 patients, which included 10 normal tissues, 6 dMMR tumor tissues, and 17 pMMR tumor tissues, aiming to characterize the cellular and molecular components of the TiME. The proficient mismatch repair (pMMR) group displayed a significantly higher prevalence of a specific GC cell type, termed GC2, characterized by increased hypoxia, epithelial-mesenchymal transition (EMT), and angiogenic activities compared to the dMMR group. GC2 cells overexpressed BEX3 and GPC3, and they significantly correlated with poorer survival. The pMMR group also showed increased infiltration of SPP1 + macrophages and FAP + fibroblasts, exhibiting strong hypoxic and pro-angiogenic features. Furthermore, a higher proportion of E2 endothelial cells, involved in extracellular matrix (ECM) remodeling and showing heightened VEGF pathway, HIF pathway, and angiogenesis activity, were identified in pMMR patients. Intercellular communication analyses revealed that GC2 cells, SPP1 + macrophages, FAP + fibroblasts, and E2 endothelial cells interact through VEGF, SPP1, and MIF signals, forming a TiME characterized by hypoxia, pro-angiogenesis, and ECM remodeling. This study uncovered TiME heterogeneity among GC patients with different MMR states, highlighting that the pMMR TiME is distinguished by hypoxia, pro-angiogenesis, and ECM remodeling, driven by the presence of GC2 cells, SPP1 + macrophages, FAP + fibroblasts, and E2 endothelial cells. These findings are pivotal for developing targeted immunotherapies for GC patients with pMMR.
Collapse
Affiliation(s)
- Zhixiong Su
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China
| | - Yufang He
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China
| | - Lijie You
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China
| | - Jingbo Chen
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China.
| | - Guifeng Zhang
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China.
| | - Zhenhua Liu
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
30
|
Yu J, Gong Y, Huang X, Bao Y. Prognostic and therapeutic potential of gene profiles related to tertiary lymphoid structures in colorectal cancer. PeerJ 2024; 12:e18401. [PMID: 39494300 PMCID: PMC11531753 DOI: 10.7717/peerj.18401] [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: 06/25/2024] [Accepted: 10/04/2024] [Indexed: 11/05/2024] Open
Abstract
The role of tertiary lymphoid structures (TLS) in oncology is gaining interest, particularly in colorectal carcinoma, yet a thorough analysis remains elusive. This study pioneered a novel TLS quantification system for prognostic and therapeutic response prediction in colorectal carcinoma, alongside a comprehensive depiction of the TLS landscape. Utilizing single-cell sequencing, we established a TLS score within the Tumor Immune Microenvironment (TIME). Analysis of tertiary lymphoid structure-related genes (TLSRGs) in 1,184 patients with colon adenocarcinoma/rectum adenocarcinoma (COADREAD) from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases led to the identification of two distinct molecular subtypes. Differentially expressed genes (DEGs) further segregated these patients into gene subtypes. A TLS score was formulated using gene set variation analysis (GSVA) and its efficacy in predicting immunotherapy outcomes was validated in two independent cohorts. High-scoring patients exhibited a 'hot' immune phenotype, correlating with enhanced immunotherapy efficacy. Key genes in our model, including C5AR1, APOE, CYR1P1, and SPP1, were implicated in COADREAD cell proliferation, invasion, and PD-L1 expression. These insights offer a novel approach to colorectal carcinoma treatment, emphasizing TLS targeting as a potential anti-tumor strategy.
Collapse
Affiliation(s)
- Jinglu Yu
- PuDong Traditional Chinese Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, Pudong New Area, China
| | - Yabin Gong
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaowei Huang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yufang Bao
- PuDong Traditional Chinese Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, Pudong New Area, China
| |
Collapse
|
31
|
Guo Y, Shen B, Lou C, Wang L, Li Y. IGSF1: a biomarker for predicting prognosis, immunotherapy response, and drug candidates in COVID-19 combined hepatocellular carcinoma. Discov Oncol 2024; 15:599. [PMID: 39470901 PMCID: PMC11522225 DOI: 10.1007/s12672-024-01483-2] [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: 07/08/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous malignancy with poor prognosis and a common cause of cancer-related death worldwide, and despite ongoing therapeutic breakthroughs, patient survival benefits are limited. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of coronavirus disease 2019 (COVID-19) and poses a major threat to humanity worldwide. As the epidemic continues to develop, more and more people are infected with SARS-CoV-2, including patients with HCC. However, the relationship between COVID-19 and HCC has not yet been fully elucidated. Our study aimed to identify the shared genetic characteristics and molecular mechanisms between COVID-19 and HCC. The data involved in this study come from Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression(GTEx), and Cancer Cell Line Encyclopedia(CCLE) databases. We used differentially expressed genes to perform enrichment analysis to reveal the biological landscape of COVID-19 combined with HCC. In addition, weighted gene co-expression network analysis (WGCNA) was used to study the co-expression network related to COVID-19 and HCC. We then combined the validation datasets to screen out immunoglobulin superfamily member 1 (IGSF1) as the most important core gene. Finally, we extensively studied the functional expression of IGSF1 in tumor samples, normal tissues, and cancer cell lines. The molecular mechanisms related to COVID-19 and HCC are rarely studied. Our study identifies IGSF1 as a potential therapeutic target and immune-related biomarker for patients with COVID-19 and HCC.
Collapse
Affiliation(s)
- Yuanhui Guo
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Baixuan Shen
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Chaoxuan Lou
- Department of Pharmacy, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Li Wang
- Department of Pharmacy, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Ying Li
- Department of Pharmacy, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
32
|
Zhang QA, Luo WS, Li J, Zhang QW, Guo Q, Chen J, Liang ZQ. Integrative Analysis of Acupuncture Targets and Immune Genes in Diabetes, Diabetic Peripheral Neuropathy, and Adjunct Therapy of Cancer. J Multidiscip Healthc 2024; 17:4939-4962. [PMID: 39492981 PMCID: PMC11529286 DOI: 10.2147/jmdh.s483940] [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: 08/13/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Introduction Acupuncture may help treat diabetes mellitus (DM), diabetic peripheral neuropathy (DPN), and adjunct therapy for cancer, but the biological mechanisms and immune-related genes involved are unclear; this study aims to clarify these aspects. Methods Comprehensive gene expression analysis revealed differentially expressed genes (DEGs) among DM, DPN, and control samples. Key genes from WGCNA were intersected with DEGs and acupuncture targets. Inflammatory responses, immune processes, signaling pathways, immune cell infiltration, and microRNA-gene interactions were studied. Hub immune-related genes' dysregulation was analyzed for copy number variation and gene methylation. A pan-cancer nomogram model was created to predict survival based on various factors, linking hub genes to cancer properties. Results Our analysis found 3,217 and 2,191 DEGs in DM/control and DPN/DM comparisons, respectively, and identified 1,830 potential acupuncture targets. We pinpointed 21 key genes in DM and 43 in DPN, involved in inflammatory responses, immune processes, CAMKK2, and cAMP signaling pathways. Distinct immune cell infiltration patterns, including M0 and M2 macrophages, neutrophils, and follicular helper T cells, were noted. Further analysis revealed microRNAs and TF genes interacting with immune hub genes in both conditions. Dysregulation of eight hub immune-related genes was linked to copy number variation and gene methylation, correlating with cancer prognosis. Co-occurrence of single nucleotide variations and oncogenic mutations was observed in these genes. The pan-cancer nomogram model showed strong prognostic capabilities, and a significant association was found between the eight genes and cancer properties like angiogenesis, EMT, and cell cycle progression. Discussion Our findings underscore the pivotal roles of MAPK3, IL1RN, SOD2, CTSD, ESR1, SLC1A1, NPY, and CCR2 in the immune response mediated by acupuncture in the context of DM, DPN, and adjunct therapy for cancer.
Collapse
Affiliation(s)
- Quan-Ai Zhang
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Wang-Sheng Luo
- Department of Cardiology, the First Affiliated Hospital of University of South China, Hengyang, People’s Republic of China
| | - Ji Li
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Qi-Wen Zhang
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Qin Guo
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Jian Chen
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Anhui Province Rural Revitalization Collaborative Technical Service Center, Huangshan University, Huangshan, People’s Republic of China
- Department of Public Health, International College, Krirk University, Bangkok, Thailand
| | - Zhi-Qiang Liang
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
33
|
Cao Z, Zhu J, Wang Z, Peng Y, Zeng L. Comprehensive pan-cancer analysis reveals ENC1 as a promising prognostic biomarker for tumor microenvironment and therapeutic responses. Sci Rep 2024; 14:25331. [PMID: 39455818 PMCID: PMC11512054 DOI: 10.1038/s41598-024-76798-9] [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: 06/24/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Accumulating research showed that ENC1 plays a critical role in maintaining the physiological functions. However, little is known about its role in predicting prognosis and immunotherapy response across cancers. In our results, compared to normal tissues, most cancer tissues exhibit increased ENC1 expression. We found that the most common type of genetic variation was gene mutation. In addition, a positive correlation was found between CNV and ENC1 expression. Moreover, the overexpression of ENC1 was positively correlated with poor clinical outcomes. The GSEA results showed that ENC1 is closely correlated with tumor-promoting biological functions in most cancers. ENC1 is also closely negatively associated with the infiltration levels of T cells, activated NK cells, and B cells. Most immunomodulators are positively associated with ENC1. Further, we verified that inhibition of ENC1 expression suppressed the proliferation and migration of breast cancer, pancreatic cancer and glioma cells. In conclusion, our study demonstrated that ENC1 plays a protumorigenic role in most cancers. Additionally, ENC1 is closely correlated with tumor microenvironment features and immune checkpoint inhibitors expression. Overall, ENC1 could serve as a promising potential prognostic biomarker in various tumors.
Collapse
Affiliation(s)
- Zhenyu Cao
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Jinfeng Zhu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan Province, China
| | - Zicheng Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yuhuai Peng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Liyun Zeng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China.
| |
Collapse
|
34
|
Wei H, Ma Y, Chen S, Zou C, Wang L. Multi-omics analysis identifies PTTG1 as a prognostic biomarker associated with immunotherapy and chemotherapy resistance. BMC Cancer 2024; 24:1315. [PMID: 39455949 PMCID: PMC11520140 DOI: 10.1186/s12885-024-13060-5] [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: 08/16/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Pituitary tumor-transforming gene 1 (PTTG1) is an important gene in tumour development. However, the relevance of PTTG1 in tumour prognosis, immunotherapy response, and medication sensitivity in human pan-cancer has to be determined. METHODS TIMER, GEPIA, the human protein atlas, GEPIA, TISCH2, and cBioportal examined the gene expression, protein expression, prognostic value, and genetic modification landscape of PTTG1 in 33 malignancies based on the TCGA cohort. The association between PTTG1 and tumour immunity, tumour microenvironment, immunotherapy response, and anticancer drug sensitivity was investigated using GSCA, TIDE, and CellMiner CDB. Molecular docking was used to validate the possible chemotherapeutic medicines for PTTG1. Additionally, siRNA-mediated knockdown was employed to confirm the probable role of PTTG1 in paclitaxel-resistant cells. RESULTS PTTG1 is overexpressed and associated with poor survival in most tumors. Functional enrichment study revealed that PTTG1 is involved in the cell cycle and DNA replication. A substantial connection between PTTG1 expression and immune cell infiltration points to PTTG1's possible role in the tumour microenvironment. High PTTG1 expression is associated with tumour immunotherapy resistance. The process could be connected to PTTG1, which mediates T cell exhaustion and promotes cytotoxic T lymphocyte malfunction. Furthermore, PTTG1 was found to be substantially linked with sensitivity to several anticancer medications. Suppressing PTTG1 with siRNA reduced clone formation and migration, implying that PTTG1 may play a role in paclitaxel resistance. CONCLUSION PTTG1 shows potential as a cancer diagnostic, prognostic, and chemosensitivity marker. Increased PTTG1 expression is linked to resistance to cancer treatment. The mechanism could be linked to PTTG1's role in promoting cytotoxic T lymphocyte dysfunction and mediating T cell exhaustion. It is feasible to consider PTTG1, which is expressed on Treg and Tprolif cells, as a new therapeutic target for overcoming immunotherapy resistance.
Collapse
Affiliation(s)
- Handong Wei
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Yaxin Ma
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Shuxing Chen
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Chunlin Zou
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| | - Lihui Wang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
35
|
Zheng Y, Yang Y, Xiong Q, Ma Y, Zhu Q. Establishment and Verification of a Novel Gene Signature Connecting Hypoxia and Lactylation for Predicting Prognosis and Immunotherapy of Pancreatic Ductal Adenocarcinoma Patients by Integrating Multi-Machine Learning and Single-Cell Analysis. Int J Mol Sci 2024; 25:11143. [PMID: 39456925 PMCID: PMC11508839 DOI: 10.3390/ijms252011143] [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: 08/25/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has earned a notorious reputation as one of the most formidable and deadliest malignant tumors. Within the tumor microenvironment, cancer cells have acquired the capability to maintain incessant expansion and increased proliferation in response to hypoxia via metabolic reconfiguration, leading to elevated levels of lactate within the tumor surroundings. However, there have been limited studies specifically investigating the association between hypoxia and lactic acid metabolism-related lactylation in PDAC. In this study, multiple machine learning approaches, including LASSO regression analysis, XGBoost, and Random Forest, were employed to identify hub genes and construct a prognostic risk signature. The implementation of the CERES score and single-cell analysis was used to discern a prospective therapeutic target for the management of PDAC. CCK8 assay, colony formation assays, transwell, and wound-healing assays were used to explore both the proliferation and migration of PDAC cells affected by CENPA. In conclusion, we discovered two distinct subtypes characterized by their unique hypoxia and lactylation profiles and developed a risk score to evaluate prognosis, as well as response to immunotherapy and chemotherapy, in PDAC patients. Furthermore, we indicated that CENPA may serve as a promising therapeutic target for PDAC.
Collapse
Affiliation(s)
| | | | | | | | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; (Y.Z.)
| |
Collapse
|
36
|
Zarei M, Sadri F, Mohajeri Khorasani A, Mirinezhad M, Mousavi P. The pan-cancer landscape presented ITGA7 as a prognostic determinant, tumor suppressor, and oncogene in multiple tumor types. FASEB J 2024; 38:e70098. [PMID: 39373985 DOI: 10.1096/fj.202400917r] [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: 04/22/2024] [Revised: 08/09/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
Integrin α7 (ITGA7) is an extracellular matrix-binding protein. Integrins are the main type of cell adhesive molecules in mammals, playing a role in many biological pathways. Although various studies have shown correlations between ITGA7 and various types of cancer, a comprehensive study at a pan-cancer level has not yet been conducted. In this study, we investigated the function of ITGA7 in distinct tumor types using the multi-omics relevant information, then two CeRNA regulatory network was drawn to identify the ITGA7 hub regulatory RNAs. The results indicated that the expression of ITGA7 varies in different tumors. Overexpression of ITGA7 was correlated with a worse OS in BLCA, LGG, and UVM, and the downregulation of ITGA7 was related to a worse OS in PAAD. In addition, BLCA, and UVM showed poor PFS in association with ITGA7 overexpression, and PAAD, SARC, and THCA indicated poor PFS in correlation with ITGA7 under expression. Further analyses of ITGA7 gene alteration data showed that ITGA7 amplifications may have an impact on Kidney Chromophobe prognosis. In 20 types of tumors, ITGA7 expression was linked to cancer-associated fibroblast infiltration. ITGA7 expression was linked to cancer-associated fibroblast infiltration. ITGA7-Related Gene Enrichment Analysis indicated that ITGA7 expression-correlated and functional binding genes were enriched in homotypic cell-cell adhesion, focal adhesion, and ECM-receptor interaction. This pan-cancer study found that abnormal expression of ITGA7 was correlated with poor prognosis and metastasis in different types of tumors. Thus, the ITGA7 gene may prove to be a promising biomarker for the prognosis and complication prevention of different cancers.
Collapse
Affiliation(s)
- Mahboobeh Zarei
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Sadri
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | - Amirhossein Mohajeri Khorasani
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - MohammadReza Mirinezhad
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pegah Mousavi
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
37
|
Bespalov D, Pino D, Vidal-Guirao S, Franquesa J, Lopez-Ramajo D, Filgaira I, Wan L, O'Sullivan PA, Ley SC, Forcales SV, Rojas JJ, Izquierdo-Serra M, Soler C, Manils J. Bioinformatic analysis of molecular characteristics and oncogenic features of CARD14 in human cancer. Sci Rep 2024; 14:22972. [PMID: 39362963 PMCID: PMC11452207 DOI: 10.1038/s41598-024-74565-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024] Open
Abstract
Aberrant caspase recruitment domain family member 14 (CARD14) signaling has been strongly associated with inflammatory skin conditions. CARD14 acts as a scaffold protein, ultimately activating the transcription factor NF-KB. Although primarily studied in the context of inflammation, recent research has suggested its potential implications in tumorigenesis. In this study, we gathered The Cancer Genome Atlas (TCGA) tumor data to gauge the involvement of CARD14 in cancer, including genetic alterations, expression patterns, survival correlations, immune cell infiltration and functional interactions across diverse cancer types. We found heightened CARD14 expression in most tumors and there was a significant correlation between CARD14 expression and the prognosis of patients for certain tumors. For instance, patients with higher CARD14 expression had a better prognosis in sarcoma, lung, cervix and head and neck cancers. Moreover, CARD14 expression positively correlated with neutrophil infiltration in most of the cancer types analyzed. Finally, enrichment analysis showed that epithelial development and differentiation pathways were involved in the functional mechanism of CARD14. Our results show that CARD14 may have the potential to become a prognostic biomarker in several cancers, hence, further prospective studies will be required for its validation.
Collapse
Affiliation(s)
- Daniil Bespalov
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, 08007, Spain
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 08907, Spain
| | - Dayana Pino
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, 08007, Spain
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 08907, Spain
| | - Sònia Vidal-Guirao
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, 08007, Spain
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 08907, Spain
| | - Júlia Franquesa
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, 08007, Spain
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 08907, Spain
| | - Daniel Lopez-Ramajo
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, 08007, Spain
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 08907, Spain
| | - Ingrid Filgaira
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, 08007, Spain
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 08907, Spain
| | - Li Wan
- Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences, 190 Kuaiyuan Avenue, Guangzhou, 510530, China
| | - Paul A O'Sullivan
- MRC Centre for Molecular Bacteriology & Infection, Imperial College London, London, SW7 2AZ, UK
| | - Steven C Ley
- Institute of Immunity & Transplantation, Royal Free Hospital, University College London, London, NW3 2PP, UK
| | - Sonia Vanina Forcales
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, 08007, Spain
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 08907, Spain
| | - Juan José Rojas
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, 08007, Spain
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 08907, Spain
| | - Mercè Izquierdo-Serra
- Neurohysiology Group, Department of Biomedicine, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, 08036, Spain
| | - Concepció Soler
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, 08007, Spain
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 08907, Spain
| | - Joan Manils
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Barcelona, 08007, Spain.
- Immunity, Inflammation and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, 08907, Spain.
- Serra Húnter Programme, Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, Universitat de Barcelona, Feixa Llarga s/n, L'Hospitalet de Llobregat, 08907, Spain.
| |
Collapse
|
38
|
Wang L, Fan J, Wu S, Cheng S, Zhao J, Fan F, Gao C, Qiao R, Sheng Q, Hu Y, Zhang Y, Liu P, Jiao Z, Wei T, Lei J, Chen Y, Qin H. LTBR acts as a novel immune checkpoint of tumor-associated macrophages for cancer immunotherapy. IMETA 2024; 3:e233. [PMID: 39429877 PMCID: PMC11487550 DOI: 10.1002/imt2.233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 10/22/2024]
Abstract
Tumor-associated macrophages (TAMs) greatly contribute to immune checkpoint inhibitor (ICI) resistance of cancer. However, its underlying mechanisms and whether TAMs can be promising targets to overcome ICI resistance remain to be unveiled. Through integrative analysis of immune multiomics data and single-cell RNA-seq data (iMOS) in lung adenocarcinoma (LUAD), lymphotoxin β receptor (LTBR) is identified as a potential immune checkpoint of TAMs, whose high expression, duplication, and low methylation are correlated with unfavorable prognosis. Immunofluorescence staining shows that the infiltration of LTBR+ TAMs is associated with LUAD stages, immunotherapy failure, and poor prognosis. Mechanistically, LTΒR maintains immunosuppressive activity and M2 phenotype of TAMs by noncanonical nuclear factor kappa B and Wnt/β-catenin signaling pathways. Macrophage-specific knockout of LTBR hinders tumor growth and prolongs survival time via blocking TAM immunosuppressive activity and M2 phenotype. Moreover, TAM-targeted delivery of LTΒR small interfering RNA improves the therapeutic effect of ICI via reversing TAM-mediated immunosuppression, such as boosting cytotoxic CD8+ T cells and inhibiting granulocytic myeloid-derived suppressor cells infiltration. Taken together, we bring forth an immune checkpoint discovery pipeline iMOS, identify LTBR as a novel immune checkpoint of TAMs, and propose a new immunotherapy strategy by targeting LTBR+ TAMs.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Jieyi Fan
- Department of Aerospace MedicineFourth Military Medical UniversityXi'anChina
| | - Sifan Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Shilin Cheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Junlong Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Fan Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Chunchen Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Rong Qiao
- Department of Clinical Oncology, Xijing HospitalFourth Military Medical UniversityXi'anChina
| | - Qiqi Sheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Yiyang Hu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Yong Zhang
- Department of Pulmonary Medicine, Xijing HospitalFourth Military Medical UniversityXi'anChina
| | - Pengjun Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Zhe Jiao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Tiaoxia Wei
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Jie Lei
- Department of Thoracic Surgery, Tangdu HospitalFourth Military Medical UniversityXi'anChina
| | - Yan Chen
- Department of Clinical Oncology, Xijing HospitalFourth Military Medical UniversityXi'anChina
| | - Hongyan Qin
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| |
Collapse
|
39
|
Wang J, Guo T, Mi Y, Meng X, Xu S, Dai F, Sun C, Huang Y, Wang J, Zhu L, Hou J, Wu S. A tumour-associated macrophage-based signature for deciphering prognosis and immunotherapy response in prostate cancer. IET Syst Biol 2024; 18:155-171. [PMID: 39138838 PMCID: PMC11490193 DOI: 10.1049/syb2.12097] [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: 02/23/2024] [Revised: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 08/15/2024] Open
Abstract
For the multistage progression of prostate cancer (PCa) and resistance to immunotherapy, tumour-associated macrophage is an essential contributor. Although immunotherapy is an important and promising treatment modality for cancer, most patients with PCa are not responsive towards it. In addition to exploring new therapeutic targets, it is imperative to identify highly immunotherapy-sensitive individuals. This research aimed to establish a signature risk model, which derived from the macrophage, to assess immunotherapeutic responses and predict prognosis. Data from the UCSC-XENA, GEO and TISCH databases were extracted for analysis. Based on both single-cell datasets and bulk transcriptome profiles, a macrophage-related score (MRS) consisting of the 10-gene panel was constructed using the gene set variation analysis. MRS was highly correlated with hypoxia, angiogenesis, and epithelial-mesenchymal transition, suggesting its potential as a risk indicator. Moreover, poor immunotherapy responses and worse prognostic performance were observed in the high-MRS group of various immunotherapy cohorts. Additionally, APOE, one of the constituent genes of the MRS, affected the polarisation of macrophages. In particular, the reduced level of M2 macrophage and tumour progression suppression were observed in PCa xenografts which implanted in Apolipoprotein E-knockout mice. The constructed MRS has the potential as a robust prognostic prediction tool, and can aid in the treatment selection of PCa, especially immunotherapy options.
Collapse
Affiliation(s)
- Jian Wang
- Department of UrologyAffiliated Hospital of Jiangnan UniversityWuxiChina
- Department of UrologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Tao Guo
- Department of UrologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yuanyuan Mi
- Department of UrologyAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Xiangyu Meng
- Department of UrologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Shuang Xu
- Department of UrologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Feng Dai
- Department of UrologyAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Chengwen Sun
- Department of UrologyAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Yi Huang
- Department of UrologyAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Jun Wang
- Department of UrologyAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Lijie Zhu
- Department of UrologyAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Jianquan Hou
- Department of UrologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Sheng Wu
- Department of UrologyAffiliated Hospital of Jiangnan UniversityWuxiChina
| |
Collapse
|
40
|
Mao Q, Qiao Z, Wang Q, Zhao W, Ju H. Construction and validation of a machine learning-based immune-related prognostic model for glioma. J Cancer Res Clin Oncol 2024; 150:439. [PMID: 39352539 PMCID: PMC11445300 DOI: 10.1007/s00432-024-05970-5] [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: 08/13/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Glioma stands as the most prevalent primary brain tumor found within the central nervous system, characterized by high invasiveness and treatment resistance. Although immunotherapy has shown potential in various tumors, it still faces challenges in gliomas. This study seeks to develop and validate a prognostic model for glioma based on immune-related genes, to provide new tools for precision medicine. METHODS Glioma samples were obtained from a database that includes the ImmPort database. Additionally, we incorporated ten machine learning algorithms to assess the model's performance using evaluation metrics like the Harrell concordance index (C-index). The model genes were further studied using GSCA, TISCH2, and HPA databases to understand their role in glioma pathology at the genomic, molecular, and single-cell levels, and validate the biological function of IKBKE in vitro experiments. RESULTS In this study, a total of 199 genes associated with prognosis were identified using univariate Cox analysis. Subsequently, a consensus prognostic model was developed through the application of machine learning algorithms. In which the Lasso + plsRcox algorithm demonstrated the best predictive performance. The model showed a good ability to distinguish two groups in both the training and test sets. Additionally, the model genes were closely related to immunity (oligodendrocytes and macrophages), and mutation burden. The results of in vitro experiments showed that the expression level of the IKBKE gene had a significant effect on the apoptosis and migration of GL261 glioma cells. Western blot analysis showed that down-regulation of IKBKE resulted in increased expression of pro-apoptotic protein Bax and decreased expression of anti-apoptotic protein Bcl-2, which was consistent with increased apoptosis rate. On the contrary, IKBKE overexpression caused a decrease in Bax expression an increase in Bcl-2 expression, and a decrease in apoptosis rate. Tunel results further confirmed that down-regulation of IKBKE promoted apoptosis, while overexpression of IKBKE reduced apoptosis. In addition, cells with down-regulated IKBKE had reduced migration in scratch experiments, while cells with overexpression of IKBKE had increased migration. CONCLUSION This study successfully constructed a glioma prognosis model based on immune-related genes. These findings provide new perspectives for glioma prognosis assessment and immunotherapy.
Collapse
Affiliation(s)
- Qi Mao
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Zhi Qiao
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Qiang Wang
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Wei Zhao
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Haitao Ju
- Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China.
| |
Collapse
|
41
|
Li S, Peng Y, Chen M, Zhao Y, Xiong Y, Li J, Luo P, Wang H, Zhao F, Zhao Q, Cui Y, Chen S, Zhou JG, Wang S. Facilitating integrative and personalized oncology omics analysis with UCSCXenaShiny. Commun Biol 2024; 7:1200. [PMID: 39341906 PMCID: PMC11439031 DOI: 10.1038/s42003-024-06891-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
The continuous generation of multi-omics and phenotype data is propelling advancements in precision oncology. UCSCXenaShiny was developed as an interactive tool for exploring thousands of cancer datasets available on UCSC Xena. However, its capacity for comprehensive and personalized pan-cancer data analysis is being challenged by the growing demands. Here, we introduce UCSCXenaShiny v2, a milestone update through a variety of improvements. Firstly, by integrating multidimensional data and implementing adaptable sample settings, we create a suite of robust TPC (TCGA, PCAWG, CCLE) analysis pipelines. These pipelines empower users to conduct in-depth analyses of correlation, comparison, and survival in three modes: Individual, Pan-cancer and Batch screen. Additionally, the tool includes download interfaces that enable users to access diverse data and outcomes, several features also facilitate the joint analysis of drug sensitivity and multi-omics of cancer cell lines. UCSCXenaShiny v2 is an open-source R package and a web application, freely accessible at https://github.com/openbiox/UCSCXenaShiny .
Collapse
Affiliation(s)
- Shensuo Li
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, PR China
| | - Yuzhong Peng
- School of Pharmacy, Macau University of Science and Technology, Macau, SAR, PR China
| | - Minjun Chen
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, PR China
| | - Yankun Zhao
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, PR China
| | - Yi Xiong
- Xiangya School of Medicine, Central South University, Changsha, 410013, PR China
| | - Jianfeng Li
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, PR China
| | - Haitao Wang
- Center for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau, SAR, PR China
- Thoracic Surgery Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, 20892, USA
| | - Fei Zhao
- University of the Chinese Academy of Sciences, Beijing, China
| | - Qi Zhao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yanru Cui
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Sujun Chen
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, PR China.
| | - Jian-Guo Zhou
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, PR China.
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Shixiang Wang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
- Department of Biomedical Informatics, School of Life Sciences, Central South University, Changsha, PR China.
| |
Collapse
|
42
|
Xu J, Hao J, Liao X, Shang X, Li X. SSCI: Self-Supervised Deep Learning Improves Network Structure for Cancer Driver Gene Identification. Int J Mol Sci 2024; 25:10351. [PMID: 39408682 PMCID: PMC11476395 DOI: 10.3390/ijms251910351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
The pathogenesis of cancer is complex, involving abnormalities in some genes in organisms. Accurately identifying cancer genes is crucial for the early detection of cancer and personalized treatment, among other applications. Recent studies have used graph deep learning methods to identify cancer driver genes based on biological networks. However, incompleteness and the noise of the networks will weaken the performance of models. To address this, we propose a cancer driver gene identification method based on self-supervision for graph convolutional networks, which can efficiently enhance the structure of the network and further improve predictive accuracy. The reliability of SSCI is verified by the area under the receiver operating characteristic curves (AUROC), the area under the precision-recall curves (AUPRC), and the F1 score, with respective values of 0.966, 0.964, and 0.913. The results show that our method can identify cancer driver genes with strong discriminative power and biological interpretability.
Collapse
Affiliation(s)
- Jialuo Xu
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China; (J.X.); (J.H.); (X.L.); (X.S.)
| | - Jun Hao
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China; (J.X.); (J.H.); (X.L.); (X.S.)
| | - Xingyu Liao
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China; (J.X.); (J.H.); (X.L.); (X.S.)
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China; (J.X.); (J.H.); (X.L.); (X.S.)
| | - Xingyi Li
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China; (J.X.); (J.H.); (X.L.); (X.S.)
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518063, China
| |
Collapse
|
43
|
Yang J, Chen Y, Zhang S, Gao X. Clinical significance of RETN gene expression and rs3219175 G > a polymorphism in cancer. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024:1-11. [PMID: 39324380 DOI: 10.1080/15257770.2024.2408735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/21/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
The inflammatory cytokine resistin, which is encoded by the RETN gene, plays a variety of roles in cancer. This study aimed to assess the relationship between RETN gene expression and cancer stage, survival prognosis, immune infiltration, and drug sensitivity, and whether the rs3219175 G > A polymorphism affected the expression of the RETN gene and cancer risk. The clinical significance of RETN gene expression and the rs3219175 polymorphism in cancer was analyzed by the GSCA platform, GTEx database and STATA software. The results showed that RETN gene expression was associated with the stage of thyroid carcinoma, survival prognosis and immune infiltration of certain cancers, and sensitivity to multiple drugs. The rs3219175 polymorphism could influence the expression of the RETN gene in a wide range of tissues. Furthermore, RETN gene rs3219175 polymorphism was significantly associated with cancer risk [GA vs. GG: OR = 2.27, 95%CI = 1.26-4.09; (GA + AA) vs. GG: OR = 2.23, 95%CI = 1.28-3.88; A vs. G: OR = 1.72, 95%CI = 1.15-2.58]. In conclusion, the current study suggested that resistin might serve as a prognostic marker and therapeutic target for certain cancers, and the rs3219175 polymorphism might be used as a marker for predicting cancer risk.
Collapse
Affiliation(s)
- Jiaojiao Yang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yuqing Chen
- School of Pharmacy, Yancheng Teachers' University, Yancheng, China
| | - Shulong Zhang
- Department of General Surgery, Xuhui District Central Hospital of Shanghai, Shanghai, China
| | - Xueren Gao
- School of Pharmacy, Yancheng Teachers' University, Yancheng, China
| |
Collapse
|
44
|
Liu J, Bing Z, Wang J. Comprehensive pan-cancer analysis and experiments revealed R3HDM1 as a novel predictive biomarker for prognosis and immune therapy response. Front Genet 2024; 15:1404348. [PMID: 39376739 PMCID: PMC11456529 DOI: 10.3389/fgene.2024.1404348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/10/2024] [Indexed: 10/09/2024] Open
Abstract
Background R3HDM1, an RNA binding protein with one R3H domain, remains uncharacterized in terms of its association with tumor progression, malignant cell regulation, and the tumor immune microenvironment. This paper aims to fill this gap by analyzing the potential of R3HDM1 in diagnosis, prognosis, chemotherapy, and immune function across various cancers. Methods Data was collected from the Firehost database (http://gdac.broadinstitute.org) to obtain the TCGA pan-cancer queue containing tumor and normal samples. Additional data on miRNA, TCPA, mutations, and clinical information were gathered from the UCSC Xena database (https://xenabrowser.net/datapages/). The mutation frequency and locus of R3HDM1 in the TCGA database were examined using the cBioPortal. External validation through GEO data was conducted to assess the differential expression of R3HDM1 in different cancers. Protein expression levels were evaluated using the Clinical Proteomics Tumor Analysis Alliance (CPTAC). The differential expression of R3HDM1 was verified in lung adenocarcinoma cell lines and normal lung glandular epithelial cells via RT-qPCR. Cell migration and proliferation experiments were conducted by knocking down the expression of R3HDM1 in two lung adenocarcinoma cell lines using small interfering RNA. The biological role of R3HDM1 in pan-cancer was explored using the GSEA method. Multiple immune infiltration algorithms from the TIMER2.0 database was employed to investigate the correlation between R3HDM1 expression and the tumor immune microenvironment. Validation of transcriptome immune infiltration was based on 140 single-cell datasets from the TISCH database. The study also characterized a pan-cancer survival profile and analyzed the differential expression of R3HDM1 in different molecular subtypes. The relationship between R3HDM1 and drug resistance was investigated using four chemotherapy data sources: CellMiner, GDSC, CTRP and PRISM. The impact of chemicals on the expression of R3HDM1 was explored through the CTD database. Result The study revealed differential expression of R3HDM1 in various tumors, indicating its potential as an early diagnostic marker. Changes in somatic copy number (SCNA) and DNA methylation were identified as factors contributing to abnormal expression levels. Additionally, the study found that R3HDM1 expression is associated with clinical features, metabolic pathways, and important pathways related to metastasis and the immune system. High expression of R3HDM1 was linked to poor prognosis across different tumors and altered drug sensitivity. Furthermore, the expression of R3HDM1 showed significant correlations with immune modulatory molecules and biomarkers of lymphocyte subpopulation infiltration. Finally, the study highlighted four chemicals that could influence the expression of R3HDM1. Conclusion Overall, this study proposes that R3HDM1 expression is a promising biomarker for predicting the prognosis of cancer, especially lung adenocarcinoma, and the efficacy of immunotherapy, demonstrating the rationale for further exploration in the development of anti-tumor therapies.
Collapse
Affiliation(s)
- Jiawei Liu
- School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Zhitong Bing
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China
- Gansu Laboratory of Isotope, Gansu Provincial Laboratory, Lanzhou, Gansu, China
| | - Junling Wang
- School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
45
|
Li Y, Yu Z. Pan-cancer analysis reveals copper transporters as promising potential targets. Heliyon 2024; 10:e37007. [PMID: 39281483 PMCID: PMC11402228 DOI: 10.1016/j.heliyon.2024.e37007] [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: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 09/18/2024] Open
Abstract
Background Copper transport proteins (SLC31A1, ATP7A, ATP7B) regulate copper levels in the body and may be involved in tumor development. However, their comprehensive expression and function across various cancers remain unclear. Methods The expressions of copper transporters in 33 tumors and normal tissues were analyzed using TCGA, GTEx, CCLE, ULCAN, and HPA databases. Cox regression assessed their impact on patient survival. Gene alterations were explored using cBioPortal. Spearman correlation tests were performed to investigate the associations between copper transporters and tumor mutation burden (TMB), microsatellite instability (MSI), and infiltration of immune cells. Gene functions were analyzed using STRING and GeneMANIA databases. Drug sensitivity was assessed using GSCALite database. ATP7B expression in lung squamous cell carcinoma (LUSC) was validated by immunohistochemical staining. Results Copper transporters exhibited variable expression patterns across various cancer types, indicating their potential dual role as either oncogenes or tumor suppressor genes, depending on the cancer type. Significant associations were found between these transporters and tumor stage, as well as prognosis in most tumors studied. Pathway analysis identified links between copper transporters and tumor-related pathways like apoptosis and RAS/MAPK. Copy number variation (CNV) analysis revealed varying degrees of gene amplification and deletion of copper transporters in most tumors. Copper transporters exhibited strong correlations with immune features, including TMB, MSI, and immune-infiltrating cells, suggesting their potential role in guiding immunotherapy. They were also associated with sensitivity to various chemotherapeutic and immunotherapeutic drugs. Immunohistochemical tests validated the correlation between elevated ATP7B level and worse progression-free survival (PFS) in LUSC. Conclusion Copper transporters may serve as potential tumor markers and therapeutic targets.
Collapse
Affiliation(s)
- Yueqin Li
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, China
- NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhen Yu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
46
|
Huang L, Xie Y, Jiang S, Gong B, Feng Y, Shan H. Identification of the shared gene MXD3 signatures and biological mechanism in patients with hip pain and prostate cancer. Medicine (Baltimore) 2024; 103:e39592. [PMID: 39287260 PMCID: PMC11404923 DOI: 10.1097/md.0000000000039592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Prostate cancer (PRAD) is recognized as having a significant effect on systemic illnesses. This study examined possible immune cells, metabolic pathways, and genes that may explain the interaction between PRAD and hip pain. We used information retrieved from the Cancer Genome Atlas and the Gene Expression Omnibus databases. To find common genes, we utilized differential expression analysis and weighted gene co-expression network analysis. The genes that were shared were subjected to pathway enrichment studies using Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes. Additionally, hub genes were analyzed using LASSO regression, and a receiver operating characteristic curve was generated based on the screening outcomes. The genes for the nodes were chosen in a protein-protein interaction network that was built. Single-sample gene-set enrichment analysis was performed to identify the differentially expressed genes. Immunohistochemistry staining confirmed hub gene expression, and single-sample gene-set enrichment analysis assessed immune cell infiltration. We concluded by comparing MAX dimerization protein 3 (MXD3) and MAX interactor 1 (MXI1) expression in tumor tissues using Uniform Manifold Approximation and Projection and violin plots in the Tumor lmmune Single-cell Hub database. After analyzing the intersection of the differentially expressed genes and weighted gene co-expression network analysis-significant module genes, we determined that MXD3 was the best shared diagnostic biomarker for PRAD and hip pain. One potential predictor of PRAD development was the MXI1 node gene, which was found in the protein-protein interaction network. The analyses revealed that MXD3 had a relatively positive correlation with neutrophil and T-helper cell infiltration levels, whereas MXI1 had a negative correlation with mast and Tgd cell levels. Tumors had lower levels of MXI1 expression and higher levels of MXD3 expression compared to normal tissues. Endothelial cells, induced pluripotent stem cells, and smooth muscle cells were all found to express MXI1. This is the first study to investigate the close genetic link between hip pain and PRAD using bioinformatics technologies. The 2 most significant genes involved in crosstalk between PRAD and hip pain were MXD3 and MXI1. The immunological responses triggered by T cells, mast cells, and neutrophils may be crucial in the relationship between PRAD and hip pain.
Collapse
Affiliation(s)
- Liang Huang
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Yu Xie
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Shusuan Jiang
- Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Binbin Gong
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yao Feng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Shan
- Department of Emergency Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| |
Collapse
|
47
|
Zhang Z, Xiao Y, Zhao S, Liu J, Zeng J, Xiao F, Liao B, Shan X, Zhu H, Guo H. FAM109B plays a tumorigenic role in low-grade gliomas and is associated with tumor-associated macrophages (TAMs). J Transl Med 2024; 22:833. [PMID: 39256832 PMCID: PMC11389277 DOI: 10.1186/s12967-024-05641-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/29/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Family with sequence similarity 109, member B (FAM109B) is involved in endocytic transport and affects genetic variation in brain methylation. It is one of the important genes related to immune cell-associated diseases. In the tumor immune system, methylation can regulate tumor immunity and influence the maturation and functional response of immune cells. Whether FAM109B is involved in tumor progression and its correlation with the tumor immune microenvironment has not yet been disclosed. METHODS A comprehensive pan-cancer analysis of FAM109B expression, prognosis, immunity, and TMB was conducted. The expression, clinical features, and prognostic value of FAM109B in low-grade gliomas (LGG) were evaluated using TCGA, CGGA, and Gravendeel databases. The expression of FAM109B was validated by qRT-PCR, immunohistochemistry (IHC), and Western blotting (WB). The relationship between FAM109B and methylation, Copy Number Variation (CNV), prognosis, immune checkpoints (ICs), and common chemotherapy drug sensitivity in LGG was explored through Cox regression, Kaplan-Meier curves, and Spearman correlation analysis. FAM109B levels and their distribution were studied using the TIMER database and single-cell analysis. The potential role of FAM109B in gliomas was further investigated through in vitro and in vivo experiments. RESULTS FAM109B was significantly elevated in various tumor types and was associated with poor prognosis. Its expression was related to aggressive progression and poor prognosis in low-grade glioma patients, serving as an independent prognostic marker for LGG. Glioma grade was negatively correlated with FAM109B DNA promoter methylation. Immune infiltration and single-cell analysis showed significant expression of FAM109B in tumor-associated macrophages (TAMs). The expression of FAM109B was closely related to gene mutations, immune checkpoints (ICs), and chemotherapy drugs in LGG. In vitro studies showed increased FAM109B expression in LGG, closely related to cell proliferation. In vivo studies showed that mice in the sh-FAM109B group had slower tumor growth, slower weight loss, and longer survival times. CONCLUSIONS FAM109B, as a novel prognostic biomarker for low-grade gliomas, exhibits specific overexpression in TAMs and may be a potential therapeutic target for LGG patients.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Yao Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Siyi Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Jun Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Jie Zeng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Feng Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Bin Liao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
- Institute of Neuroscience, Nanchang University, Jiangxi, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China
| | - Xuesong Shan
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China
| | - Hong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China.
- Institute of Neuroscience, Nanchang University, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China.
| | - Hua Guo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, China.
- Institute of Neuroscience, Nanchang University, Jiangxi, China.
- Jiangxi Province Key Laboratory of Neurological Diseases, Jiangxi, China.
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, China.
| |
Collapse
|
48
|
Huang J, Mao L, Lei Q, Guo AY. Bioinformatics tools and resources for cancer and application. Chin Med J (Engl) 2024; 137:2052-2064. [PMID: 39075637 PMCID: PMC11374212 DOI: 10.1097/cm9.0000000000003254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Indexed: 07/31/2024] Open
Abstract
ABSTRACT Tumor bioinformatics plays an important role in cancer research and precision medicine. The primary focus of traditional cancer research has been molecular and clinical studies of a number of fundamental pathways and genes. In recent years, driven by breakthroughs in high-throughput technologies, large-scale cancer omics data have accumulated rapidly. How to effectively utilize and share these data is particularly important. To address this crucial task, many computational tools and databases have been developed over the past few years. To help researchers quickly learn and understand the functions of these tools, in this review, we summarize publicly available bioinformatics tools and resources for pan-cancer multi-omics analysis, regulatory analysis of tumorigenesis, tumor treatment and prognosis, immune infiltration analysis, immune repertoire analysis, cancer driver gene and driver mutation analysis, and cancer single-cell analysis, which may further help researchers find more suitable tools for their research.
Collapse
Affiliation(s)
- Jin Huang
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lingzi Mao
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Qian Lei
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - An-Yuan Guo
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
49
|
Yang L, Wei Q, Chen X, Yang Y, Huang Q, Wang B, Ma X. Identification of HDAC10 as a candidate oncogene in clear cell renal carcinoma that facilitates tumor proliferation and metastasis. Diagn Pathol 2024; 19:120. [PMID: 39237939 PMCID: PMC11378624 DOI: 10.1186/s13000-024-01493-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/06/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) remains one of the most lethal urological malignancies even though a great number of improvements in diagnosis and management have achieved over the past few decades. Accumulated evidence revealed that histone deacetylases (HDACs) play vital role in cell proliferation, differentiation and apoptosis. Nevertheless, the biological functions of histone deacetylation modification related genes in ccRCC remains poorly understood. METHOD Bulk transcriptomic data and clinical information of ccRCC patients were obtained from the TCGA database and collected from the Chinese PLA General Hospital. A total of 36 histone deacetylation genes were selected and studied in our research. Univariate cox regression analysis, least absolute shrinkage and selection operator (LASSO) regression, random forest (RF) analysis, and protein-protein interaction (PPI) network analysis were applied to identify key genes affecting the prognosis of ccRCC. The 'oncoPredict' algorithm was utilized for drug-sensitive analysis. Gene Set Enrichment Analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis was used to explore the potential biological function. The ssGSEA algorithm was used for tumor immune microenvironment analysis. The expression levels of HDAC10 were validated by RT-PCR and immunohistochemistry (IHC). 5-ethynyl-2'-deoxyuridine (EdU assay), CCK-8 assay, cell transwell migration and invasion assay and colony formation assay were performed to detect the proliferation and invasion ability of ccRCC cells. A nomogram incorporating HDAC10 and clinicopathological characteristics was established to predict the prognosis of ccRCC patients. RESULT Two machine learning algorithms and PPI analysis identified four histone deacetylation genes that have a significant association with the prognosis of ccRCC, with HDAC10 being the key gene among them. HDAC10 is highly expressed in ccRCC and its high expression is associated with poor prognosis for ccRCC patients. Pathway enrichment and the experiments of EdU staining, CCK-8 assay, cell transwell migration and invasion assay and colony formation assay demonstrated that HDAC10 mediated the proliferation and metastasis of ccRCC cells and involved in reshaping the tumor microenvironment (TME) of ccRCC. A clinically reliable prognostic predictive model was established by incorporating HDAC10 and other clinicopathological characteristics ( https://nomogramhdac10.shinyapps.io/HDAC10_Nomogram/ ). CONCLUSION Our study found the increased expression of HDAC10 was closely associated with poor prognosis of ccRCC patients. HDAC10 showed a pro-tumorigenic effect on ccRCC and promote the proliferation and metastasis of ccRCC, which may provide new light on targeted therapy for ccRCC.
Collapse
Affiliation(s)
- Luojia Yang
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qin Wei
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200125, China
| | - Xinran Chen
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yang Yang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qingbo Huang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Baojun Wang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Xin Ma
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
50
|
Cheng G, Xu J, Wang H, Chen J, Huang L, Qian ZR, Fan Y. mtPCDI: a machine learning-based prognostic model for prostate cancer recurrence. Front Genet 2024; 15:1430565. [PMID: 39296545 PMCID: PMC11408181 DOI: 10.3389/fgene.2024.1430565] [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: 05/10/2024] [Accepted: 08/22/2024] [Indexed: 09/21/2024] Open
Abstract
Background This research seeks to formulate a prognostic model for forecasting prostate cancer recurrence by examining the interaction between mitochondrial function and programmed cell death (PCD). Methods The research involved analyzing four gene expression datasets from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) using univariate Cox regression. These analyses identified genes linked with mitochondrial function and PCD that correlate with recurrence prognosis. Various machine learning algorithms were then employed to construct an optimal predictive model. Results A key outcome was the creation of a mitochondrial-related programmed cell death index (mtPCDI), which effectively predicts the prognosis of prostate cancer patients. It was observed that individuals with lower mtPCDI exhibited higher immune activity, correlating with better recurrence outcomes. Conclusion The study demonstrates that mtPCDI can be used for personalized risk assessment and therapeutic decision-making, highlighting its clinical significance and providing insights into the biological processes affecting prostate cancer recurrence.
Collapse
Affiliation(s)
- Guoliang Cheng
- Department of Urology Surgery, The Fourth People's Hospital of Jinan, Jinan, Shandong, China
| | - Junrong Xu
- Department of Urology Surgery, The Fourth People's Hospital of Jinan, Jinan, Shandong, China
| | - Honghua Wang
- Department of Urology Surgery, The Fourth People's Hospital of Jinan, Jinan, Shandong, China
| | - Jingzhao Chen
- Beidou Precision Medicine Institute, Guangzhou, China
| | - Liwei Huang
- Beidou Precision Medicine Institute, Guangzhou, China
| | - Zhi Rong Qian
- Beidou Precision Medicine Institute, Guangzhou, China
| | - Yong Fan
- Department of Urology Surgery, The Fourth People's Hospital of Jinan, Jinan, Shandong, China
| |
Collapse
|