1
|
Zuo Z, Wen R, Jing S, Chen X, Liu R, Xue J, Zhang L, Li Q. Ganoderma lucidum (Curtis) P. Karst. Immunomodulatory Protein Has the Potential to Improve the Prognosis of Breast Cancer Through the Regulation of Key Prognosis-Related Genes. Pharmaceuticals (Basel) 2024; 17:1695. [PMID: 39770537 PMCID: PMC11677753 DOI: 10.3390/ph17121695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Breast cancer in women is the most commonly diagnosed and most malignant tumor. Although luminal A breast cancer (LumA) has a relatively better prognosis, it still has a persistent pattern of recurrence. Ganoderma lucidum (Curtis) P. Karst. is a kind of traditional Chinese medicine and has antitumor effects. In this study, we aimed to identify the genes relevant to prognosis, find novel targets, and investigate the function of the bioactive protein from G. lucidum, called FIP-glu, in improving prognosis. Methods: Gene expression data and clinical information of LumA breast cancer patients were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Using bioinformatics methods, a predictive risk model was constructed to predict the prognosis for each patient. The cell counting kit-8 (CCK8) and clone formation assays were used to validate gene function. The ability of FIP-glu to regulate RNA levels of risk genes was validated. Results: Six risk genes (slit-roundabout GTPase-activating protein 2 (SRGAP2), solute carrier family 35 member 2 (SLC35A2), sequence similarity 114 member A1 (FAM114A1), tumor protein P53-inducible protein 11 (TP53I11), transmembrane protein 63C (TMEM63C), and polymeric immunoglobulin receptor (PIGR)) were identified, and a prognostic model was constructed. The prognosis was worse in the high-risk group and better in the low-risk group. The receiver operating characteristic (ROC) curve confirmed the model's accuracy. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses showed that the differentially expressed genes (DEGs) between the high- and low-risk groups were significantly enriched in the immune responses. TMEM63C could promote tumor viability, growth, and proliferation in vitro. FIP-glu significantly regulated these risk genes, and attenuated the promoting effect of TMEM63C in breast cancer cells. Conclusions: SRGAP2, SLC35A2, FAM114A1, TP53I11, TMEM63C, and PIGR were identified as the potential risk genes for predicting the prognosis of patients. TMEM63C could be a potential novel therapeutic target. Moreover, FIP-glu was a promising drug for improving the prognosis of LumA breast cancer.
Collapse
Affiliation(s)
- Zanwen Zuo
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
| | - Ruihua Wen
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
| | - Shuang Jing
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
| | - Xianghui Chen
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Ruisang Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life Science and Health Engineering, Hubei University of Technology, Wuhan 430068, China
| | - Jianping Xue
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
| | - Lei Zhang
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Qizhang Li
- Innovative Drug R&D Center, Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China; (Z.Z.)
| |
Collapse
|
2
|
Zeng X, Yang D, Zhang J, Li K, Wang X, Ma F, Liao X, Wang Z, Zeng X, Zhang P. Integrating machine learning, bioinformatics and experimental verification to identify a novel prognostic marker associated with tumor immune microenvironment in head and neck squamous carcinoma. Front Immunol 2024; 15:1501486. [PMID: 39720726 PMCID: PMC11666523 DOI: 10.3389/fimmu.2024.1501486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Head and neck squamous carcinoma (HNSC), characterized by a high degree of malignancy, develops in close association with the tumor immune microenvironment (TIME). Therefore, identifying effective targets related to HNSC and TIME is of paramount importance. Here, we employed the ESTIMATE algorithm to compute immune and stromal cell scores for HNSC samples from the TCGA database and identified differentially expressed genes (DEGs) based on these scores. Subsequently, we utilized four machine learning algorithms to identify four key genes: ITM2A, FOXP3, WIPF1, and RSPO1 from DEGs. Through a comprehensive pan-cancer analysis, our study identified aberrant expression of ITM2A across various tumor types, with a significant association with the TIME. Specifically, ITM2A expression was markedly reduced and correlated with poor prognosis in HNSC. Functional enrichment analysis revealed that ITM2A is implicated in multiple immune-related pathways, including immune-infiltrating cells, immune checkpoints, and immunotherapeutic responses. ITM2A expression was observed in various immune cell populations through single-cell analysis. Furthermore, we showed that ITM2A overexpression inhibited the growth of HNSC cells. Our results suggest that ITM2A may be a novel prognostic marker associated with TIME.
Collapse
Affiliation(s)
- Xiaoxia Zeng
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Dunhui Yang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Jin Zhang
- Department of Otolaryngology, The Second People’s Hospital of Yibin, Yibin, Sichuan, China
| | - Kang Li
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Xijia Wang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Fang Ma
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Xianqin Liao
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Zhen Wang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Xianhai Zeng
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Peng Zhang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Chen X, Zuo Z, Li X, Li Q, Zhang L. Identification of a Potential PGK1 Inhibitor with the Suppression of Breast Cancer Cells Using Virtual Screening and Molecular Docking. Pharmaceuticals (Basel) 2024; 17:1636. [PMID: 39770478 PMCID: PMC11676932 DOI: 10.3390/ph17121636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Breast cancer is the second most common malignancy worldwide and poses a significant threat to women's health. However, the prognostic biomarkers and therapeutic targets of breast cancer are unclear. A prognostic model can help in identifying biomarkers and targets for breast cancer. In this study, a novel prognostic model was developed to optimize treatment, improve clinical prognosis, and screen potential phosphoglycerate kinase 1 (PGK1) inhibitors for breast cancer treatment. METHODS Using data from the Gene Expression Omnibus (GEO) database, differentially expressed genes (DEGs) were identified in normal individuals and breast cancer patients. The biological functions of the DEGs were examined using bioinformatics analysis. A novel prognostic model was then constructed using the DEGs through LASSO and multivariate Cox regression analyses. The relationship between the prognostic model, survival, and immunity was also evaluated. In addition, virtual screening was conducted based on the risk genes to identify novel small molecule inhibitors of PGK1 from Chemdiv and Targetmol libraries. The effects of the potential inhibitors were confirmed through cell experiments. RESULTS A total of 230 up- and 325 down-regulated DEGs were identified in HER2, LumA, LumB, and TN breast cancer subtypes. A new prognostic model was constructed using ten risk genes. The analysis from The Cancer Genome Atlas (TCGA) indicated that the prognosis was poorer in the high-risk group compared to the low-risk group. The accuracy of the model was confirmed using the ROC curve. Furthermore, functional enrichment analyses indicated that the DEGs between low- and high-risk groups were linked to the immune response. The risk score was also correlated with tumor immune infiltrates. Moreover, four compounds with the highest score and the lowest affinity energy were identified. Notably, D231-0058 showed better inhibitory activity against breast cancer cells. CONCLUSIONS Ten genes (ACSS2, C2CD2, CXCL9, KRT15, MRPL13, NR3C2, PGK1, PIGR, RBP4, and SORBS1) were identified as prognostic signatures for breast cancer. Additionally, results showed that D231-0058 (2-((((4-(2-methyl-1H-indol-3-yl)-1,3-thiazol-2-yl)carbamoyl)methyl)sulfanyl)acetic acid) may be a novel candidate for treating breast cancer.
Collapse
Affiliation(s)
- Xianghui Chen
- School of Medicine, Shanghai University, Shanghai 200444, China
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Zanwen Zuo
- Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China
| | - Xianbin Li
- School of Computer and Big Data Science, Jiujiang University, Jiujiang 332000, China
| | - Qizhang Li
- Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China
| | - Lei Zhang
- School of Medicine, Shanghai University, Shanghai 200444, China
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
- Innovative Drug Research Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| |
Collapse
|
4
|
Mei N, Gong S, Wang L, Wang L, Wang J, Li J, Bao Y, Zhang H, Wang H. Identification of a Prognostic Model Based on NK Cell-Related Genes in Multiple Myeloma Using Single-Cell and Transcriptomic Data Analysis. Blood Lymphat Cancer 2024; 14:31-48. [PMID: 38854627 PMCID: PMC11162243 DOI: 10.2147/blctt.s461529] [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: 03/06/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024]
Abstract
Background Multiple myeloma (MM), an incurable plasma cell malignancy. The significance of the relationship between natural killer (NK) cell-related genes and clinical factors in MM remains unclear. Methods Initially, we extracted NK cell-related genes from peripheral blood mononuclear cells (PBMC) of healthy donors and MM samples by employing single-cell transcriptome data analysis in TISCH2. Subsequently, we screened NK cell-related genes with prognostic significance through univariate Cox regression analysis and protein-protein interaction (PPI) network analysis. Following the initial analyses, we developed potential subtypes and prognostic models for MM using consensus clustering and lasso regression analysis. Additionally, we conducted a correlation analysis to explore the relationship between clinical features and risk scores. Finally, we constructed a weighted gene co-expression network analysis (WGCNA) and identified differentially expressed genes (DEGs) within the MM cohort. Results We discovered that 153 NK cell-related genes were significantly associated with the prognosisof MM patients (P <0.05). Patients in NK cluster A exhibited poorer survival outcomes compared to those in cluster B. Furthermore, our NK cell-related genes risk model revealed that patients with a high risk score had significantly worse prognoses (P <0.05). Patients with a high risk score were more likely to exhibit adverse clinical markers. Additionally, the nomogram based on NK cell-related genes demonstrated strong prognostic performance. The enrichment analysis indicated that immune-related pathways were significantly correlated with both the NK subtypes and the NK cell-related genes risk model. Ultimately, through the combined use of WGCNA and DEGs analysis, and by employing Venn diagrams, we determined that ITM2C is an independent prognostic marker for MM patients. Conclusion In this study, we developed a novel model based on NK cell-related genes to stratify the prognosis of MM patients. Notably, higher expression levels of ITM2C were associated with more favorable survival outcomes in these patients.
Collapse
Affiliation(s)
- Nan Mei
- Department of Hematology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, People’s Republic of China
| | - Sha Gong
- Department of Hematology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, People’s Republic of China
| | - Lizhao Wang
- Department of Breast Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, People’s Republic of China
| | - Lu Wang
- Department of Hematology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, People’s Republic of China
| | - Jincheng Wang
- Department of Hematology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, People’s Republic of China
| | - Jianpeng Li
- Department of Urology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, People’s Republic of China
| | - Yingying Bao
- Institute of Gene and Cell Therapy, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, People’s Republic of China
| | - Huanming Zhang
- Department of Hematology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, People’s Republic of China
| | - Huaiyu Wang
- Department of Hematology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, People’s Republic of China
| |
Collapse
|
5
|
Vanmathi P, Jose D. An ensemble-based serial cascaded attention network and improved variational auto encoder for breast cancer prognosis prediction using data. Comput Methods Biomech Biomed Engin 2024; 27:98-115. [PMID: 38006210 DOI: 10.1080/10255842.2023.2280883] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023]
Abstract
Breast cancer is one of the most common types of cancer in women and it produces a huge amount of death rate in the world. Early recognition is lessening its impact. The early recognition of breast cancer could convince patients to receive surgical therapy, which will significantly improve the chance of restoration. This information is used by the machine learning technique to find links between them and appraise our forecasts of fresh occurrences. Later recognition of breast cancer can lead to death. An accurate prescient framework for breast cancer prediction is urgently needed in the current era. In order to accomplish the objective, an adaptive ensemble model is proposed for breast cancer prognosis prediction using data. At the initial stage, the raw data are fetched from benchmark datasets. It is then followed by data cleaning and preprocessing. Subsequently, the pre-processed data is fed into the Improved Variational Autoencoder (IVAE), where the deep features are extracted. Finally, the resultant features are given as input to the Ensemble-based Serial Cascaded Attention Network (ESCANet), which is built with Deep Temporal Convolution Network (DTCN), Bi-directional Long Short-Term Memory (BiLSTM), and Recurrent Neural Network (RNN). The effectiveness of the model is validated and compared with conventional methodologies. Therefore, the results elucidate that the proposed methodology achieves extensive results; thus, it increases the system's efficiency.
Collapse
Affiliation(s)
- P Vanmathi
- Full time Research Scholar, Department of ECE, KCG College of Technology, Karapakkam, Chennai, Tamil Nadu, India
| | - Deepa Jose
- Professor, Department of ECE, KCG College of Technology, Karapakkam, Chennai, Tamil Nadu, India
| |
Collapse
|
6
|
Suba Z. Rosetta Stone for Cancer Cure: Comparison of the Anticancer Capacity of Endogenous Estrogens, Synthetic Estrogens and Antiestrogens. Oncol Rev 2023; 17:10708. [PMID: 37152665 PMCID: PMC10154579 DOI: 10.3389/or.2023.10708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 03/30/2023] [Indexed: 05/09/2023] Open
Abstract
This work presents the history of the recognition of principal regulatory capacities of estrogen hormones having been mistakenly regarded as breast cancer promoting agents for more than 120 years. Comprehensive analysis of the results of clinical, epidemiological, immunological and molecular studies justified that endogenous estrogens are the principal regulators of embryonic development, survival and reproduction via orchestrating appropriate expression and even edition of all genes in mammalians. Medical use of chemically modified synthetic estrogens caused toxic complications; thromboembolic events and increased cancer risk in female organs as they proved to be endocrine disruptors deregulating estrogen receptors (ERs) rather than their activators. Synthetic estrogen treatment exhibits ambiguous correlations with cancer risk at different sites, which may be attributed to an inhibition of the unliganded activation of estrogen receptors (ERs) coupled with compensatory liganded activation. The principle of estrogen induced breast cancer led to the introduction of antiestrogen therapies against this tumor; inhibition of the liganded activation of estrogen receptors and aromatase enzyme activity. The initial enthusiasm turned into disappointment as the majority of breast cancers proved to be primarily resistant to antiestrogens. In addition, nearly all patients showing earlier good tumor responses to endocrine therapy, later experienced secondary resistance leading to metastatic disease and fatal outcome. Studying the molecular events in tumors responsive and unresponsive to antiestrogen therapy, it was illuminated that a complete inhibition of liganded ER activation stimulates the growth of cancers, while a successful compensatory upregulation of estrogen signal may achieve DNA restoration, tumor regression and patient's survival. Recognition of the principal role of endogenous estrogens in gene expression, gene edition and DNA repair, estrogen treatment and stimulation of ER expression in patients may bring about a great turn in medical practice.
Collapse
|
7
|
Prieto-Fernández L, Menéndez ST, Otero-Rosales M, Montoro-Jiménez I, Hermida-Prado F, García-Pedrero JM, Álvarez-Teijeiro S. Pathobiological functions and clinical implications of annexin dysregulation in human cancers. Front Cell Dev Biol 2022; 10:1009908. [PMID: 36247003 PMCID: PMC9554710 DOI: 10.3389/fcell.2022.1009908] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Annexins are an extensive superfamily of structurally related calcium- and phospholipid-binding proteins, largely conserved and widely distributed among species. Twelve human annexins have been identified, referred to as Annexin A1-13 (A12 remains as of yet unassigned), whose genes are spread throughout the genome on eight different chromosomes. According to their distinct tissue distribution and subcellular localization, annexins have been functionally implicated in a variety of biological processes relevant to both physiological and pathological conditions. Dysregulation of annexin expression patterns and functions has been revealed as a common feature in multiple cancers, thereby emerging as potential biomarkers and molecular targets for clinical application. Nevertheless, translation of this knowledge to the clinic requires in-depth functional and mechanistic characterization of dysregulated annexins for each individual cancer type, since each protein exhibits varying expression levels and phenotypic specificity depending on the tumor types. This review specifically and thoroughly examines the current knowledge on annexin dysfunctions in carcinogenesis. Hence, available data on expression levels, mechanism of action and pathophysiological effects of Annexin A1-13 among different cancers will be dissected, also further discussing future perspectives for potential applications as biomarkers for early diagnosis, prognosis and molecular-targeted therapies. Special attention is devoted to head and neck cancers (HNC), a complex and heterogeneous group of aggressive malignancies, often lately diagnosed, with high mortality, and scarce therapeutic options.
Collapse
Affiliation(s)
- Llara Prieto-Fernández
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Sofía T. Menéndez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - María Otero-Rosales
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Irene Montoro-Jiménez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Hermida-Prado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Juana M. García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Saúl Álvarez-Teijeiro
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria Del Principado de Asturias (ISPA), Instituto Universitario de Oncología Del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Zhang T, Yu S, Zhao S. ANXA9 as a novel prognostic biomarker associated with immune infiltrates in gastric cancer. PeerJ 2021; 9:e12605. [PMID: 35003923 PMCID: PMC8684324 DOI: 10.7717/peerj.12605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 11/16/2021] [Indexed: 11/26/2022] Open
Abstract
Background Gastric cancer (GC) is the most prevalent malignancy among the digestive system tumors. Increasing evidence has revealed that lower mRNA expression of ANXA9 is associated with a poor prognosis in colorectal cancer. However, the role of ANXA9 in GC remains largely unknown. Material and Methods The Gene Expression Profiling Interactive Analysis (GEPIA) and Human Protein Atlas databases were used to investigate the expression of ANXA9 in GC, which was then validated in the four Gene Expression Omnibus (GEO) datasets. The diagnostic value of ANXA9 for GC patients was demonstrated using a receiver operating characteristic (ROC) curve. The correlation between ANXA9 expression and clinicopathological parameters was analyzed in The Cancer Genome Atlas (TCGA) and UALCAN databases. The Kaplan-Meier (K-M) survival curve was used to elucidate the relationship between ANXA9 expression and the survival time of GC patients. We then performed a gene set enrichment analysis (GSEA) to explore the biological functions of ANXA9. The relationship of ANXA9 expression and cancer immune infiltrates was analyzed using the Tumor Immune Estimation Resource (TIMER). In addition, the potential mechanism of ANXA9 in GC was investigated by analyzing its related genes. Results ANXA9 was significantly up-regulated in GC tissues and showed obvious diagnostic value. The expression of ANXA9 was related to the age, gender, grade, TP53 mutation, and histological subtype of GC patients. We also found that ANXA9 expression was associated with immune-related biological function. ANXA9 expression was also correlated with the infiltration level of CD8+ T cells, neutrophils, and dendritic cells in GC. Additionally, copy number variation (VNV) of ANXA9 occurred in GC patients. Function enrichment analyses revealed that ANXA9 plays a role in the GC progression by interacting with its related genes. Conclusions Our results provide strong evidence of ANXA9 expression as a prognostic indicator related to immune responses in GC.
Collapse
|
9
|
Large Rab GTPases: Novel Membrane Trafficking Regulators with a Calcium Sensor and Functional Domains. Int J Mol Sci 2021; 22:ijms22147691. [PMID: 34299309 PMCID: PMC8303950 DOI: 10.3390/ijms22147691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/28/2022] Open
Abstract
Rab GTPases are major coordinators of intracellular membrane trafficking, including vesicle transport, membrane fission, tethering, docking, and fusion events. Rab GTPases are roughly divided into two groups: conventional “small” Rab GTPases and atypical “large” Rab GTPases that have been recently reported. Some members of large Rab GTPases in mammals include Rab44, Rab45/RASEF, and Rab46. The genes of these large Rab GTPases commonly encode an amino-terminal EF-hand domain, coiled-coil domain, and the carboxyl-terminal Rab GTPase domain. A common feature of large Rab GTPases is that they express several isoforms in cells. For instance, Rab44’s two isoforms have similar functions, but exhibit differential localization. The long form of Rab45 (Rab45-L) is abundantly distributed in epithelial cells. The short form of Rab45 (Rab45-S) is predominantly present in the testes. Both Rab46 (CRACR2A-L) and the short isoform lacking the Rab domain (CRACR2A-S) are expressed in T cells, whereas Rab46 is only distributed in endothelial cells. Although evidence regarding the function of large Rab GTPases has been accumulating recently, there are only a limited number of studies. Here, we report the recent findings on the large Rab GTPase family concerning their function in membrane trafficking, cell differentiation, related diseases, and knockout mouse phenotypes.
Collapse
|
10
|
Hsa-miR-105-1 Regulates Cisplatin-Resistance in Ovarian Carcinoma Cells by Targeting ANXA9. ACTA ACUST UNITED AC 2021; 2021:6662486. [PMID: 33680718 PMCID: PMC7929659 DOI: 10.1155/2021/6662486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/02/2021] [Indexed: 01/29/2023]
Abstract
Purpose Cisplatin is one of the most effective drugs for treating ovarian carcinoma (OC), which is among the most lethal types of carcinoma. However, the chemoresistance to cisplatin that develops over time leads to a poor clinical outcome for many OC patients. Therefore, it is necessary to clearly understand the molecular mechanisms of chemoresistance. In this study, we examined how Hsa-miR-105-1 functions in cisplatin-resistant OC cells. Methods The levels of Hsa-miR-105-1 expression in cisplatin-sensitive and resistant OC cell lines were detected by qRT-PCR. The target gene of Hsa-miR-105-1 was predicted by using the TargetScan and Starbase databases and verified by the double luciferase reporter gene assay. The target gene of Hsa-miR-105-1 was identified as ANXA9, and ANXA9 expression was evaluated by qRT-PCR, western blotting, and immunofluorescence. To validate the function of Hsa-miR-105-1 in OC cells, we silenced or overexpressed Hsa-miR-105-1 in cisplatin-sensitive or resistant OC cell lines, respectively. Furthermore, the expression levels of several apoptosis-related proteins, including P53, P21, E2F1, Bcl-2, Bax, and caspase-3, were examined by western blot analysis. Results The levels of Hsa-miR-105-1 expression were abnormally downregulated in cisplatin-resistant OC cells, while ANXA9 expression was significantly upregulated in those cells. Treatment with an Hsa-miR-105-1 inhibitor promoted the expression of ANXA9 mRNA and protein, enhanced the resistance to cisplatin, and attenuated the cell apoptosis induced by cisplatin in cisplatin-sensitive OC cells. Moreover, treatment with Hsa-miR-105-1 mimics inhibited ANXA9 expression, which further increased the levels of P53, P21, and Bax expression and decreased the levels of E2F1 and Bcl-2 expression, finally resulting in an increased sensitivity to cisplatin in cisplatin-resistant OC cells. Conclusion We found that a downregulation of Hsa-miR-105-1 expression enhanced cisplatin-resistance, while an upregulation of Hsa-miR-105-1 restored the sensitivity of OC cells to cisplatin. The Hsa-miR-105-1/ANXA9 axis plays an important role in the cisplatin-resistance of OC cells.
Collapse
|
11
|
Mathias C, Groeneveld CS, Trefflich S, Zambalde EP, Lima RS, Urban CA, Prado KB, Ribeiro EMSF, Castro MAA, Gradia DF, de Oliveira JC. Novel lncRNAs Co-Expression Networks Identifies LINC00504 with Oncogenic Role in Luminal A Breast Cancer Cells. Int J Mol Sci 2021; 22:ijms22052420. [PMID: 33670895 PMCID: PMC7957645 DOI: 10.3390/ijms22052420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/18/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are functional transcripts with more than 200 nucleotides. These molecules exhibit great regulatory capacity and may act at different levels of gene expression regulation. Despite this regulatory versatility, the biology of these molecules is still poorly understood. Computational approaches are being increasingly used to elucidate biological mechanisms in which these lncRNAs may be involved. Co-expression networks can serve as great allies in elucidating the possible regulatory contexts in which these molecules are involved. Herein, we propose the use of the pipeline deposited in the RTN package to build lncRNAs co-expression networks using TCGA breast cancer (BC) cohort data. Worldwide, BC is the most common cancer in women and has great molecular heterogeneity. We identified an enriched co-expression network for the validation of relevant cell processes in the context of BC, including LINC00504. This lncRNA has increased expression in luminal subtype A samples, and is associated with prognosis in basal-like subtype. Silencing this lncRNA in luminal A cell lines resulted in decreased cell viability and colony formation. These results highlight the relevance of the proposed method for the identification of lncRNAs in specific biological contexts.
Collapse
Affiliation(s)
- Carolina Mathias
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-900, PR, Brazil; (C.M.); (E.P.Z.); (K.B.P.); (E.M.S.F.R.); (D.F.G.)
| | - Clarice S. Groeneveld
- Cartes d’Identité des Tumeurs Program, Ligue Nationale Contre le Cancer, 75013 Paris, France;
- Oncologie Moleculaire, Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue Contre le Cancer, 75005 Paris, France
| | - Sheyla Trefflich
- Bioinformatics and Systems Biology Laboratory, Polytechnic Center, Federal University of Parana (UFPR), Curitiba 81520-260, PR, Brazil; (S.T.); (M.A.A.C.)
| | - Erika P. Zambalde
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-900, PR, Brazil; (C.M.); (E.P.Z.); (K.B.P.); (E.M.S.F.R.); (D.F.G.)
| | - Rubens S. Lima
- Breast Disease Center, Hospital Nossa Senhora das Graças, Curitiba 80810040, PR, Brazil; (R.S.L.); (C.A.U.)
| | - Cícero A. Urban
- Breast Disease Center, Hospital Nossa Senhora das Graças, Curitiba 80810040, PR, Brazil; (R.S.L.); (C.A.U.)
| | - Karin B. Prado
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-900, PR, Brazil; (C.M.); (E.P.Z.); (K.B.P.); (E.M.S.F.R.); (D.F.G.)
| | - Enilze M. S. F. Ribeiro
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-900, PR, Brazil; (C.M.); (E.P.Z.); (K.B.P.); (E.M.S.F.R.); (D.F.G.)
| | - Mauro A. A. Castro
- Bioinformatics and Systems Biology Laboratory, Polytechnic Center, Federal University of Parana (UFPR), Curitiba 81520-260, PR, Brazil; (S.T.); (M.A.A.C.)
| | - Daniela F. Gradia
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-900, PR, Brazil; (C.M.); (E.P.Z.); (K.B.P.); (E.M.S.F.R.); (D.F.G.)
| | - Jaqueline C. de Oliveira
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-900, PR, Brazil; (C.M.); (E.P.Z.); (K.B.P.); (E.M.S.F.R.); (D.F.G.)
- Correspondence:
| |
Collapse
|