1
|
Saulle I, Limanaqi F, Garziano M, Murno ML, Artusa V, Strizzi S, Giovarelli M, Schulte C, Aiello J, Clerici M, Vanetti C, Biasin M. Impact of endoplasmic reticulum aminopeptidases 1 (ERAP1) and 2 (ERAP2) on neutrophil cellular functions. Front Cell Dev Biol 2025; 12:1506216. [PMID: 39839670 PMCID: PMC11747162 DOI: 10.3389/fcell.2024.1506216] [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: 10/04/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction Endoplasmic reticulum aminopeptidases 1 (ERAP1) and 2 (ERAP2) modulate a plethora of physiological processes for the maintenance of homeostasis in different cellular subsets at both intra and extracellular level. Materials and methods In this frame, the extracellular supplementation of recombinant human (rh) ERAP1 and ERAP2 (300 ng/ml) was used to mimic the effect of stressor-induced secretion of ERAPs on neutrophils isolated from 5 healthy subjects. In these cells following 3 h or 24 h rhERAP stimulation by Western Blot, RT-qPCR, Elisa, Confocal microscopy, transwell migration assay, Oxygraphy and Flow Cytometry we assessed: i) rhERAP internalization; ii) activation; iii) migration; iv) oxygen consumption rate; v) reactive oxygen species (ROS) accumulation; granule release; vi) phagocytosis; and vii) autophagy. Results We observed that following stimulation rhERAPs: i) were internalized by neutrophils; ii) triggered their activation as witnessed by increased percentage of MAC-1+CD66b+ expressing neutrophils, cytokine expression/release (IL-1β, IL-8, CCL2, TNFα, IFNγ, MIP-1β) and granule enzyme secretion (myeloperoxidase, Elastase); iii) increased neutrophil migration capacity; iv) increased autophagy and phagocytosis activity; v) reduced ROS accumulation and did not influence oxygen consumption rate. Conclusion Our study provides novel insights into the biological role of ERAPs, and indicates that extracellular ERAPs, contribute to shaping neutrophil homeostasis by promoting survival and tolerance in response to stress-related inflammation. This information could contribute to a better understanding of the biological bases governing immune responses, and to designing ERAP-based therapeutic protocols to control neutrophil-associated human diseases.
Collapse
Affiliation(s)
- Irma Saulle
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Milano, Italy
| | - Fiona Limanaqi
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
| | - Micaela Garziano
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Milano, Italy
| | - Maria Luisa Murno
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
| | - Valentina Artusa
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Milano, Italy
| | - Sergio Strizzi
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
| | - Matteo Giovarelli
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
| | - Carsten Schulte
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
| | - Jacopo Aiello
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
| | - Mario Clerici
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Milano, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Don Carlo Gnocchi, Milano, Italy
| | - Claudia Vanetti
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
| | - Mara Biasin
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
2
|
Lin PW, Lin ZR, Wang WW, Guo AS, Chen YX. Identification of immune-inflammation targets for intracranial aneurysms: a multiomics and epigenome-wide study integrating summary-data-based Mendelian randomization, single-cell-type expression analysis, and DNA methylation regulation. Int J Surg 2025; 111:346-359. [PMID: 39051921 PMCID: PMC11745758 DOI: 10.1097/js9.0000000000001990] [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: 05/23/2024] [Accepted: 07/14/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Dysfunction of the immune system and inflammation plays a vital role in developing intracranial aneurysms (IAs). However, the progress of genetic pathophysiology is complicated and not entirely elaborated. This study aimed to explore the genetic associations of immune-related and inflammation-related genes (IIRGs) with IAs and their subtypes using Mendelian randomization, colocalization test, and integrated multiomics functional analysis. METHODS The authors conducted a summary-data-based Mendelian randomization (SMR) analysis using data from several genome-wide association studies of gene expression (31 684 European individuals) and protein quantitative trait loci (35 559 Icelanders), as well as information on IAs and their subtypes from The International Stroke Genetics Consortium (IGSC) for discovery phase and the FinnGen study for replication. This analysis aimed to determine the causal relationship between IIRGs and the risk of IAs and their subtypes. Further functional analyses, including DNA methylation regulation (1980, European individuals), single-cell-type expression analysis, and protein-protein interaction, were conducted to detect the specific cell type with enriched expression and discover potential drug targets. RESULTS After integrating multiomics evidence from expression quantitative trait loci (eQTL) and protein quantitative trait loci (pQTL), the authors found that tier 1: RELT [odds ratio (OR): 0.14, 95% CI: 0.04-0.50], TNFSF12 (OR: 1.24, 95% CI: 1.24-1.43), tier 3: ICAM5 (OR: 0.89, 95% CI: 0.82-0.96), and ERAP2 (OR: 1.07, 95% CI: 1.02-1.12) were associated with the risk of IAs; tier 3: RELT (OR: 0.11, 95% CI: 0.02-0.54), ERAP2 (OR: 1.08, 95% CI: 1.02-1.13), and TNFSF12 (OR: 1.24, 95% CI: 1.05-1.47) were associated with the risk of aneurysmal subarachnoid hemorrhage (aSAH); and tier 1: RELT (OR: 0.04, 95% CI: 0.01-0.30) was associated with the risk of unruptured intracranial aneurysms (uIAs). Further functional analyses showed that RELT was regulated by cg06382664 and cg18850434 and ICAM5 was regulated by cg04295144 in IAs; RELT was regulated by cg06382664, cg08770935, cg16533363, and cg18850434 in aSAH; and RELT was regulated by cg06382664 and cg21810604 in uIAs. In addition, the authors found that H6PD (OR: 1.13, 95% CI: 1.01-1.28), NT5M (OR: 1.91, 95% CI: 1.21-3.01), and NPTXR (OR: 1.13, 95% CI: 1.01-1.26) were associated with IAs; NT5M (OR: 2.13, 95% CI: 1.23-3.66) was associated aSAH; and AP4M1 (OR: 0.06, 95% CI: 0.01-0.42) and STX7 (OR: 3.97, 95% CI: 1.41-11.18) were related to uIAs. STX7 and TNFSF12 were mainly enriched in microglial cells, whereas H6PD, STX7 , and TNFSF12 were mainly enriched in astrocytes. CONCLUSIONS After integrating multiomics evidence, the authors eventually identified IIRGs: RELT, TNFSF12, ICAM5 , and ERAP2 were the novel therapy targets for IAs. These new results confirmed a vital role of immune and inflammation in the etiology of IAs, contributing to enhance our understanding of the immune and inflammatory mechanisms in the pathogenesis of IAs and revealing the complex genetic causality of IAs.
Collapse
Affiliation(s)
- Peng-Wei Lin
- The School of Clinical Medicine, Fujian Medical University, Zhangzhou Affiliated Hospital of Fujian Medical University, Fuzhou
| | - Zhen-Rong Lin
- Department of Neurosurgery, Zhangzhou Municipal Hospital of Fujian Province and Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, People’s Republic of China
| | - Wei-Wei Wang
- Department of Neurosurgery, Zhangzhou Municipal Hospital of Fujian Province and Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, People’s Republic of China
| | - Ai-Shun Guo
- Department of Neurosurgery, Zhangzhou Municipal Hospital of Fujian Province and Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian Province, People’s Republic of China
| | - Yu-Xiang Chen
- The School of Clinical Medicine, Fujian Medical University, Zhangzhou Affiliated Hospital of Fujian Medical University, Fuzhou
| |
Collapse
|
3
|
Li J, Han Y, Zhao N, Lv L, Ma P, Zhang Y, Li M, Sun H, Deng J, Zhang Y. Identification of immune- and oxidative stress-related signature genes as potential targets for mRNA vaccines for pancreatic cancer patients. Medicine (Baltimore) 2024; 103:e38666. [PMID: 38968513 PMCID: PMC11224846 DOI: 10.1097/md.0000000000038666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 07/07/2024] Open
Abstract
Adenocarcinoma of the pancreas (PAAD) is one of the deadliest malignant tumors, and messenger ribonucleic acid vaccines, which constitute the latest generation of vaccine technology, are expected to lead to new ideas for the treatment of pancreatic cancer. The Cancer Genome Atlas-PAAD and Genotype-Tissue Expression data were merged and analyzed. Weighted gene coexpression network analysis was used to identify gene modules associated with tumor mutational burden among the genes related to both immunity and oxidative stress. Differentially expressed immune-related oxidative stress genes were screened via univariate Cox regression analysis, and these genes were analyzed via nonnegative matrix factorization. After immune infiltration analysis, least absolute shrinkage and selection operator regression combined with Cox regression was used to construct the model, and the usefulness of the model was predicted based on the receiver operating characteristic curve and decision curve analysis curves after model construction. Finally, metabolic pathway enrichment was analyzed using gene set enrichment analysis combined with Kyoto Encyclopedia of Genes and Genomes and gene ontology biological process analyses. This model consisting of the ERAP2, mesenchymal-epithelial transition factor (MET), CXCL9, and angiotensinogen (AGT) genes can be used to help predict the prognosis of pancreatic cancer patients more accurately than existing models. ERAP2 is involved in immune activation and is important in cancer immune evasion. MET binds to hepatocyte growth factor, leading to the dimerization and phosphorylation of c-MET. This activates various signaling pathways, including MAPK and PI3K, to regulate the proliferation, invasion, and migration of cancer cells. CXCL9 overexpression is associated with a poor patient prognosis and reduces the number of CD8 + cytotoxic T lymphocytes in the PAAD tumor microenvironment. AGT is cleaved by the renin enzyme to produce angiotensin 1, and AGT-converting enzyme cleaves angiotensin 1 to produce angiotensin 2. Exposure to AGT-converting enzyme inhibitors after pancreatic cancer diagnosis is associated with improved survival. The 4 genes identified in the present study - ERAP2, MET, CXCL9, and AGT - are expected to serve as targets for messenger ribonucleic acid vaccine development and need to be further investigated in depth.
Collapse
Affiliation(s)
- Jiaxu Li
- Institute of Health Service and Transfusion Medicine, Beijing, PR China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, PR China
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, PR China
| | - Yongjiao Han
- Institute of Health Service and Transfusion Medicine, Beijing, PR China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, PR China
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, PR China
| | - Ning Zhao
- Institute of Health Service and Transfusion Medicine, Beijing, PR China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, PR China
| | - Liping Lv
- Institute of Health Service and Transfusion Medicine, Beijing, PR China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, PR China
| | - Ping Ma
- Institute of Health Service and Transfusion Medicine, Beijing, PR China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, PR China
| | - Yangyang Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, PR China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, PR China
| | - Mingyuan Li
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, PR China
| | - Hua Sun
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, PR China
| | - Jiang Deng
- Institute of Health Service and Transfusion Medicine, Beijing, PR China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, PR China
| | - Yanyu Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, PR China
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, PR China
| |
Collapse
|
4
|
Wen S, Zou R, Du X, Pan R, Li R, Xia J, Xu C, Wang R, Jiang F, Zhou G, Feng J, Zhu M, Wang X, Shen B. Identification of macrophage-related genes correlated with prognosis and immunotherapy efficacy in non-small cell lung cancer. Heliyon 2024; 10:e27170. [PMID: 38500993 PMCID: PMC10945138 DOI: 10.1016/j.heliyon.2024.e27170] [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: 08/01/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Background Malignant tumours, particularly non-small cell lung cancer (NSCLC), pose a significant threat to human health due to their prevalence and lethality. Treatment methods for NSCLC vary greatly among individuals, making it crucial to identify predictive markers. Moreover, during tumour initiation and progression, tumour cells can release signaling molecules to induce polarization of macrophages towards a more tumour friendly M2 phenotype, which can promote tumour growth, metastasis, and drug resistance. Methods We employed a comprehensive approach, combining bulk RNA-seq and single-cell sequencing analysis. Results In our study, we used bulk RNA-seq and single-cell sequencing methods to analyze differential cells in NSCLC and adjacent tissues, searching for relevant marker genes that can predict prognosis and drug efficacy. We scrutinized biological phenomena such as macrophage-related gene methylation, copy number variation, and alternative splicing. Additionally, we utilized a co-culture technique of immune and tumour cells to explore the role of these genes in macrophage polarization. Our findings revealed distinct differences in macrophages between cancerous and adjacent tissues. We identified ANP32A, CCL20, ERAP2, MYD88, TMEM126B, TUBB6, and ZNF655 as macrophage-related genes that correlate with NSCLC patient prognosis and immunotherapy efficacy. Notably, ERAP2, TUBB6, CCL20, and TMEM126B can induce macrophage M0 to M2 polarization, promoting tumour proliferation. Conclusion These findings significantly contribute to our understanding of the NSCLC tumour immune microenvironment. They pave the way for further research into the potential of these genes as targets for regulating tumour occurrence and development.
Collapse
Affiliation(s)
| | | | - Xiaoyue Du
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Rongtian Pan
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Rutao Li
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Jingwei Xia
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Cong Xu
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Ruotong Wang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Feng Jiang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Guoren Zhou
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Jifeng Feng
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Miaolin Zhu
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Xin Wang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| | - Bo Shen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu, Nanjing 21000, China
| |
Collapse
|
5
|
Tian H, Wei R, Xiao C, Fan T, Che Y, Liu T, Zheng B, Li C, He J. Tumor-derived KLK8 predicts inferior survival and promotes an immune-suppressive tumor microenvironment in lung squamous cell carcinoma. BMC Pulm Med 2024; 24:53. [PMID: 38273291 PMCID: PMC10809653 DOI: 10.1186/s12890-023-02770-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/17/2023] [Indexed: 01/27/2024] Open
Abstract
Lung squamous cell carcinoma (LUSC) is the second most common lung cancer worldwide, leading to millions of deaths annually. Although immunotherapy has expanded the therapeutic choices for LUSC and achieved considerable efficacy in a subset of patients, many patients could not benefit, and resistance was pervasive. Therefore, it is significant to investigate the mechanisms leading to patients' poor response to immunotherapies and explore novel therapeutic targets. Using multiple public LUSC datasets, we found that Kallikrein-8 (KLK8) expression was higher in tumor samples and was correlated with inferior survival. Using a LUSC cohort (n = 190) from our center, we validated the bioinformatic findings about KLK8 and identified high KLK8 expression as an independent risk factor for LUSC. Function enrichment showed that several immune signaling pathways were upregulated in the KLK8 low-expression group and downregulated in the KLK8 high-expression group. For patients with low KLK8 expression, they were with a more active TME, which was both observed in the TCGA database and immune marker immunohistochemistry, and they had extensive positive relations with immune cells with tumor-eliminating functions. This study identified KLK8 as a risk factor in LUSC and illustrated the associations between KLK8 and cancer immunity, suggesting the potentiality of KLK8 as a novel immune target in LUSC.
Collapse
Affiliation(s)
- He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Ran Wei
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Yun Che
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Tiejun Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Bo Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
6
|
Wu Z, You C, Zhu Z, Wu W, Cao J, Xie Q, Deng C, Huang X, Hu S. SLA2 is a prognostic marker in HNSCC and correlates with immune cell infiltration in the tumor microenvironment. Eur Arch Otorhinolaryngol 2024; 281:427-440. [PMID: 37688682 PMCID: PMC10764518 DOI: 10.1007/s00405-023-08213-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 08/25/2023] [Indexed: 09/11/2023]
Abstract
PURPOSE To investigate Src-like adaptor 2 gene (SLA2) expression in head and neck squamous cell carcinoma (HNSCC), its potential prognostic value, and its effect on immune cell infiltration. METHODS Through a variety of bioinformatics analyses, we extracted and analyzed data sets from the Cancer Genome Atlas (TCGA), Tumor Immune Estimation Resource (TIMER), and Gene Expression Profile Interaction Analysis (GEPIA) to analyze the correlation between SLA2 and the prognosis, immune checkpoint, tumor microenvironment (TME) and immune cell infiltration of HNSCC, and to explore its potential oncogenic mechanism. To further explore the potential role of SLA2 in HNSCC by Gene ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. RESULTS SLA2 messenger ribonucleic acid (mRNA) levels were increased in HNSCC tumor tissues compared with normal tissues. In addition, we found that SLA2 may be an independent prognostic factor for HNSCC, and high SLA2 expression is associated with favorable prognosis in HNSCC. SLA2 expression was positively correlated with B cells, cluster of differentiation 8-positive T cells (CD8 + T cells), cluster of differentiation 4-positive T cells (CD4 + T cells), macrophages, neutrophil and dendritic cells infiltration. SLA2 has also been shown to co-express immune-related genes and immune checkpoints. Significant GO term analysis by Gene Set Enrichment Analysis (GSEA) indicated that genes correlated with SLA2 were located mainly in the side of membrane, receptor complex, secretory granule membrane, endocytic vesicle, membrane region, and endosome membrane, where they were involved in leukocyte cell-cell adhesion, response to interferon-gamma, and regulation of immune effector process. These related genes also served as antigen binding, cytokine receptor activity, phosphatidylinositol 3-kinase activity, peptide receptor activity, Src homology domain 3 (SH3) domain binding, and cytokine receptor binding. KEGG pathway analysis demonstrated that these genes related to SLA2 were mainly enriched in signal pathways, such as hematopoietic cell lineage, cell adhesion molecules (CAMs), natural killer cell mediated cytotoxicity, measles, and chemokine signaling pathway. CONCLUSIONS SLA2 is increased in HNSCC, and high SLA2 expression is associated with favorable prognosis. SLA2 may affect tumor development by regulating tumor infiltrating cells in TME. SLA2 may be a potential target for immunotherapy.
Collapse
Affiliation(s)
- Zhongbiao Wu
- Department of Otolaryngology, Jiangxi Hospital of Integrated Traditional Chinese and Western Medicine, 90 Bayi Avenue, Xihu District, Nanchang, 330003, Jiangxi, China
| | - Chengkun You
- Department of Neurology, Pinghu Hospital of Traditional Chinese Medicine, Jiaxing, 314200, China
| | - Zhongyan Zhu
- Department of Rehabilitation, Jiangxi Hospital of Integrated Traditional Chinese and Western Medicine, Nanchang, 330003, China
| | - Weikun Wu
- Department of Otolaryngology, Jiangxi Hospital of Integrated Traditional Chinese and Western Medicine, 90 Bayi Avenue, Xihu District, Nanchang, 330003, Jiangxi, China
| | - Jian Cao
- Department of Otolaryngology, Jiangxi Hospital of Integrated Traditional Chinese and Western Medicine, 90 Bayi Avenue, Xihu District, Nanchang, 330003, Jiangxi, China
| | - Qiang Xie
- Department of Otolaryngology, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330019, China
| | - Chengcheng Deng
- Department of Otolaryngology, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330019, China
| | - Xinmei Huang
- Department of Otolaryngology, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi, China
| | - Shiping Hu
- Department of Otolaryngology, Jiangxi Hospital of Integrated Traditional Chinese and Western Medicine, 90 Bayi Avenue, Xihu District, Nanchang, 330003, Jiangxi, China.
| |
Collapse
|
7
|
Li Z, Liu Y, Ma T, Lv C, Li Y, Duan H, Zhao X, Wang J, Zhang Y. Smart-seq2 Technology Reveals a Novel Mechanism That Zearalenone Inhibits the In Vitro Maturation of Ovine Oocytes by Influencing TNFAIP6 Expression. Toxins (Basel) 2023; 15:617. [PMID: 37888648 PMCID: PMC10611292 DOI: 10.3390/toxins15100617] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/05/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
Zearalenone (ZEN), a non-steroidal estrogenic fungal toxin widely present in forage, food, and their ingredients, poses a serious threat to animal and human reproductive health. ZEN also threatens ovine, a major source of human food and breeding stock. However, the mechanisms underlying the impact of ZEN on the in vitro maturation (IVM) of ovine oocytes remain unclear. This study aimed to elucidate these mechanisms using the Smart-seq2 technology. A total of 146 differentially expressed genes were obtained, using Smart-seq2, from sheep oocytes cultured in vitro after ZEN treatment. ZEN treatment inhibited RUNX2 and SPP1 expression in the PI3K signaling pathway, leading to the downregulation of THBS1 and ultimately the downregulation of TNFAIP6; ZEN can also decrease TNFAIP6 by reducing PTPRC and ITGAM. Both inhibit in vitro maturation of ovine oocytes and proliferation of cumulus cells by downregulating TNFAIP6. These findings provide data and a theoretical basis for elucidating ZEN's toxicity mechanisms, screening therapeutic drugs, and reducing ZEN-related losses in the ovine industry.
Collapse
Affiliation(s)
- Zongshuai Li
- State Key Laboratory of Grassland Agro–Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Grassland Agriculture Engineering Center, Ministry of Education, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China;
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China; (T.M.); (C.L.); (Y.L.); (H.D.); (X.Z.)
| | - Yali Liu
- Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Tian Ma
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China; (T.M.); (C.L.); (Y.L.); (H.D.); (X.Z.)
| | - Chen Lv
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China; (T.M.); (C.L.); (Y.L.); (H.D.); (X.Z.)
| | - Yina Li
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China; (T.M.); (C.L.); (Y.L.); (H.D.); (X.Z.)
| | - Hongwei Duan
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China; (T.M.); (C.L.); (Y.L.); (H.D.); (X.Z.)
| | - Xingxu Zhao
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China; (T.M.); (C.L.); (Y.L.); (H.D.); (X.Z.)
| | - Jianlin Wang
- State Key Laboratory of Grassland Agro–Ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, Grassland Agriculture Engineering Center, Ministry of Education, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China;
| | - Yong Zhang
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China; (T.M.); (C.L.); (Y.L.); (H.D.); (X.Z.)
| |
Collapse
|
8
|
Lu G, Wang H, Xu R, Xu J, An F, Xu H, Nie H, Mei J, Zhan Q, Zhang Q. Formin protein FMNL1 is a biomarker for tumor-infiltrating immune cells and associated with well immunotherapeutic response. J Cancer 2023; 14:2978-2989. [PMID: 37859818 PMCID: PMC10583584 DOI: 10.7150/jca.86965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/03/2023] [Indexed: 10/21/2023] Open
Abstract
Background: Increased studies on the basis of bulk RNA-sequencing (RNA-seq) data of cancer identify numbers of immune-related genes which may play potential regulatory roles in the tumor microenvironment (TME) without in-depth validation. Methods: In the current study, the immunological correlation and cell subpopulation expression pattern of FMNL1 were analyzed using public data. In addition, the cell subpopulation expression pattern of FMNL1 was also deeply validated using single-cell RNA-sequencing (scRNA-seq) and multiplexed quantitative immunofluorescence (mQIF). Results: Bulk FMNL1 mRNA was related to better prognosis in hepatocellular carcinoma (HCC) and was able to identify immuno-hot tumor in not only HCC but also multiple cancer types. Bulk FMNL1 mRNA also predicted the response to immunotherapy in multiple cancers. Further validation using scRNA-seq and mQIF revealed that FMNL1 was a biomarker for immune cells. Conclusions: FMNL1 is a biomarker for immune cells in not only hepatocellular carcinoma, but also multiple cancer types. Moreover, immune infiltration analysis using the bulk RNA-seq data would be further validated using scRNA-seq and/or mQIF to describe the cell subpopulation expression pattern in tumor tissues for more in-depth and appropriate understanding.
Collapse
Affiliation(s)
- Guomin Lu
- Departments of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Hui Wang
- Departments of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Rui Xu
- The First College of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Junying Xu
- Departments of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Fangmei An
- Departments of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Haoran Xu
- Departments of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - He Nie
- Departments of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Jie Mei
- Departments of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Qiang Zhan
- Departments of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Qinglin Zhang
- Departments of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| |
Collapse
|
9
|
Pudjihartono N, Ho D, Golovina E, Fadason T, Kempa-Liehr AW, O'Sullivan JM. Juvenile idiopathic arthritis-associated genetic loci exhibit spatially constrained gene regulatory effects across multiple tissues and immune cell types. J Autoimmun 2023; 138:103046. [PMID: 37229810 DOI: 10.1016/j.jaut.2023.103046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/15/2023] [Indexed: 05/27/2023]
Abstract
Juvenile idiopathic arthritis (JIA) is an autoimmune, inflammatory joint disease with complex genetic etiology. Previous GWAS have found many genetic loci associated with JIA. However, the biological mechanism behind JIA remains unknown mainly because most risk loci are located in non-coding genetic regions. Interestingly, increasing evidence has found that regulatory elements in the non-coding regions can regulate the expression of distant target genes through spatial (physical) interactions. Here, we used information on the 3D genome organization (Hi-C data) to identify target genes that physically interact with SNPs within JIA risk loci. Subsequent analysis of these SNP-gene pairs using data from tissue and immune cell type-specific expression quantitative trait loci (eQTL) databases allowed the identification of risk loci that regulate the expression of their target genes. In total, we identified 59 JIA-risk loci that regulate the expression of 210 target genes across diverse tissues and immune cell types. Functional annotation of spatial eQTLs within JIA risk loci identified significant overlap with gene regulatory elements (i.e., enhancers and transcription factor binding sites). We found target genes involved in immune-related pathways such as antigen processing and presentation (e.g., ERAP2, HLA class I and II), the release of pro-inflammatory cytokines (e.g., LTBR, TYK2), proliferation and differentiation of specific immune cell types (e.g., AURKA in Th17 cells), and genes involved in physiological mechanisms related to pathological joint inflammation (e.g., LRG1 in arteries). Notably, many of the tissues where JIA-risk loci act as spatial eQTLs are not classically considered central to JIA pathology. Overall, our findings highlight the potential tissue and immune cell type-specific regulatory changes contributing to JIA pathogenesis. Future integration of our data with clinical studies can contribute to the development of improved JIA therapy.
Collapse
Affiliation(s)
- N Pudjihartono
- The Liggins Institute, The University of Auckland, Auckland, New Zealand.
| | - D Ho
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - E Golovina
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - T Fadason
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - A W Kempa-Liehr
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | - J M O'Sullivan
- The Liggins Institute, The University of Auckland, Auckland, New Zealand; The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand; MRC Lifecourse Epidemiology Unit, University of Southampton, United Kingdom; Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, New South Wales, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia; A*STAR Singapore Institute for Clinical Sciences, Singapore, Singapore.
| |
Collapse
|
10
|
Hamilton F, Mentzer AJ, Parks T, Baillie JK, Smith GD, Ghazal P, Timpson NJ. Variation in ERAP2 has opposing effects on severe respiratory infection and autoimmune disease. Am J Hum Genet 2023; 110:691-702. [PMID: 36889308 PMCID: PMC10119032 DOI: 10.1016/j.ajhg.2023.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/06/2023] [Indexed: 03/09/2023] Open
Abstract
ERAP2 is an aminopeptidase involved in immunological antigen presentation. Genotype data in human samples from before and after the Black Death, an epidemic due to Yersinia pestis, have marked changes in allele frequency of the single-nucleotide polymorphism (SNP) rs2549794, with the T allele suggested to be deleterious during this period, while ERAP2 is also implicated in autoimmune diseases. This study explored the association between variation at ERAP2 and (1) infection, (2) autoimmune disease, and (3) parental longevity. Genome-wide association studies (GWASs) of these outcomes were identified in contemporary cohorts (UK Biobank, FinnGen, and GenOMICC). Effect estimates were extracted for rs2549794 and rs2248374, a haplotype tagging SNP. Additionally, cis expression and protein quantitative trait loci (QTLs) for ERAP2 were used in Mendelian randomization (MR) analyses. Consistent with decreased survival in the Black Death, the T allele of rs2549794 showed evidence of association with respiratory infection (odds ratio; OR for pneumonia 1.03; 95% CI 1.01-1.05). Effect estimates were larger for more severe phenotypes (OR for critical care admission with pneumonia 1.08; 95% CI 1.02-1.14). In contrast, opposing effects were identified for Crohn disease (OR 0.86; 95% CI 0.82-0.90). This allele was shown to associate with decreased ERAP2 expression and protein levels, independent of haplotype. MR analyses suggest that ERAP2 expression may be mediating disease associations. Decreased ERAP2 expression is associated with severe respiratory infection with an opposing association with autoimmune diseases. These data support the hypothesis of balancing selection at this locus driven by autoimmune and infectious disease.
Collapse
Affiliation(s)
- Fergus Hamilton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Infection Science, North Bristol NHS Trust, Bristol, UK.
| | | | - Tom Parks
- Wellcome Centre For Human Genetics, University of Oxford, Oxford, UK; Department of Infectious Disease, Imperial College London, London, UK
| | - J Kenneth Baillie
- Baillie Gifford Pandemic Science Hub, Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK; Roslin Institute, University of Edinburgh, Edinburgh, UK; Intensive Care Unit, Royal Infirmary of Edinburgh, Edinburgh, UK
| | | | | | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
11
|
A Comparative Review of Pregnancy and Cancer and Their Association with Endoplasmic Reticulum Aminopeptidase 1 and 2. Int J Mol Sci 2023; 24:ijms24043454. [PMID: 36834865 PMCID: PMC9965492 DOI: 10.3390/ijms24043454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The fundamental basis of pregnancy and cancer is to determine the fate of the survival or the death of humanity. However, the development of fetuses and tumors share many similarities and differences, making them two sides of the same coin. This review presents an overview of the similarities and differences between pregnancy and cancer. In addition, we will also discuss the critical roles that Endoplasmic Reticulum Aminopeptidase (ERAP) 1 and 2 may play in the immune system, cell migration, and angiogenesis, all of which are essential for fetal and tumor development. Even though the comprehensive understanding of ERAP2 lags that of ERAP1 due to the lack of an animal model, recent studies have shown that both enzymes are associated with an increased risk of several diseases, including pregnancy disorder pre-eclampsia (PE), recurrent miscarriages, and cancer. The exact mechanisms in both pregnancy and cancer need to be elucidated. Therefore, a deeper understanding of ERAP's role in diseases can make it a potential therapeutic target for pregnancy complications and cancer and offer greater insight into its impact on the immune system.
Collapse
|
12
|
Yu P, Luo S, Cai J, Li J, Peng C. ERAP2 as a potential biomarker for predicting gemcitabine response in patients with pancreatic cancer. Aging (Albany NY) 2022; 14:7941-7958. [PMID: 36214762 PMCID: PMC9596206 DOI: 10.18632/aging.204324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022]
Abstract
Objective: Pancreatic cancer is one of the most malignant tumors, with rapid metastasis, high mortality rate, and difficult early screening. Currently, gemcitabine is a first-line drug for pancreatic cancer patients, but its clinical effect is limited due to drug resistance. It is particularly important to further identify biomarkers associated with gemcitabine resistance to improve the sensitivity of gemcitabine treatment. Methods: Drug sensitivity data and the corresponding transcript data derived from the Genomics of Drug Sensitivity in Cancer (GDSC) database for correlation analysis was adopted to obtain genes related to gemcitabine sensitivity. Moreover, the survival model of pancreatic cancer patients treated with gemcitabine in The Cancer Genome Atlas (TCGA) database was utilized to obtain key genes. Multiple in vitro assays were performed to verify the function of the key biomarker. Results: Endoplasmic Reticulum Aminopeptidase 2 (ERAP2) was identified as a biomarker promoting gemcitabine resistance, and its high expression resulted in a worse prognosis. Besides, gemcitabine significantly increased the mRNA and protein levels of ERAP2 in pancreatic cancer cells. Additionally, ERAP2 knockdown suppressed tumorigenesis and potentiated gemcitabine-induced growth, migration and invasion inhibition in human pancreatic cancer cells. Conclusions: ERAP2 may be a novel key biomarker for gemcitabine sensitivity and diagnosis, thus providing an effective therapeutic strategy for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Pian Yu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Changsha 410000, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410000, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha 410000, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha 410000, Hunan, China
| | - Shifu Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Jiaxin Cai
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Jie Li
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Changsha 410000, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410000, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha 410000, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha 410000, Hunan, China
| | - Cong Peng
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Changsha 410000, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410000, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha 410000, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha 410000, Hunan, China
| |
Collapse
|
13
|
Overexpression of CDCP1 is Associated with Poor Prognosis and Enhanced Immune Checkpoints Expressions in Breast Cancer. JOURNAL OF ONCOLOGY 2022; 2022:1469354. [PMID: 36090897 PMCID: PMC9452972 DOI: 10.1155/2022/1469354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022]
Abstract
CUB-domain containing protein 1 (CDCP1) is a transmembrane protein acting as an effector of SRC family kinases, which play an oncogenic role in multiple human cancers. However, its clinical and immune correlations in breast cancer (BrCa) have not been explored. To define the expression, prognostic value, and potential molecular role of CDCP1 in BrCa, multiple public datasets, and an in-house cohort were used. Compared with paratumor tissue, CDCP1 was remarkably upregulated in the tumor tissues at both mRNA and protein levels. In the in-house cohort, CDCP1 protein expression was related to several clinicopathological parameters, including age, ER status, PR status, molecular type, and survival status. Kaplan–Meier analysis and Cox regression analysis exhibited that CDCP1 was an important prognostic biomarker in BrCa. In addition, enrichment analysis uncovered that CDCP1 was not only involved in multiple oncogenic pathways, but correlated with overexpression of immune checkpoints. Overall, we reported that increased expression of CDCP1 is a favorable prognostic factor in patients with BrCa. In addition, the correlations between CDCP1 and immune checkpoints provide a novel insight into the adjuvant treatment for immune checkpoint blockade via targeting CDCP1.
Collapse
|