1
|
Zhang X, Wang Z, Zhao Y, Ye H, Li T, Wang H, Sun G, Liang F, Dai L, Wang P, Liu X. Multi-omics analysis unveils a four-gene prognostic signature in esophageal squamous carcinoma and the therapeutic potential of PKP1. BMC Cancer 2025; 25:777. [PMID: 40281492 PMCID: PMC12032815 DOI: 10.1186/s12885-025-14150-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most common malignancies, characterized by high heterogeneity and poor outcomes. Effective classification for patient stratification and identifying reliable markers for prognosis prediction and treatment choice are crucial. METHODS Integration of single-cell RNA-sequencing (RNA-seq) and bulk RNA-seq analyses were used to characterize ESCC. Non-negative matrix factorization (NMF) clustering was performed to stratify the ESCC patients into different subtypes and the clinical and pathological features of the ESCC subtypes were compared. Cox regression analysis and LASSO regression analysis were used to select key genes and construct a risk model for ESCC. The associations of the key genes with anti-cancer drug sensitivities in ESCC cell lines were investigated. RT-qRCR experiments, proteomics analysis, and multiplex immunohistochemistry (mIHC) experiments were used to validate the results. Furthermore, one identified gene was selected to investigate its correlation with EGFR expression and the gene effect scores of various potential gene targets across pan-cancer. RESULTS The study identified the dysregulated distributions of epithelial cells and fibroblasts as characteristic of ESCC. ESCC patients could be classified into four distinct subtypes with unique cell type features and prognoses. With the gene makers of the cell type features, a four-gene prognostic signature for ESCC was constructed. The CCND1-PKP1-JUP-ANKRD12 model could effectively discriminate the survival status of ESCC patients, independent of various pathological and clinical features. The risk score for the samples was correlated with the expression levels of immunoregulatory genes. The prognostic effects of CCND1, PKP1, and JUP were confirmed at the protein level. The phosphorylation levels of PKP1, JUP, and ANKRD12 were found to be dysregulated in ESCC tumors. Their expression dysregulation and heterogeneity were demonstrated in ESCC cell lines. All four genes were significantly correlated with at least one of the anti-cancer drug sensitivities in ESCC cell lines. PKP1 expression was significantly correlated with EGFR expression and gene effect scores in multiple cancers. CONCLUSIONS We conclude that the CCND1-PKP1-JUP-ANKRD12 signature may serve as a novel indicator for ESCC prognosis and diagnosis. PKP1 expression might provide new clues for gene therapy efficacy in multiple cancers.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
| | - Zhi Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Yutong Zhao
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Hua Ye
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Tiandong Li
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
| | - Han Wang
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
| | - Guiying Sun
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China
| | - Feifei Liang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| | - Peng Wang
- College of Public Health, Zhengzhou University, Zhengzhou, 4500001, China.
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450052, China.
| | - Xiaoli Liu
- Laboratory Department, Henan Provincial People's Hospital, Zhengzhou, 450003, China.
| |
Collapse
|
2
|
Owecki W, Wojtowicz K, Nijakowski K. Salivary Extracellular Vesicles in Detection of Cancers Other than Head and Neck: A Systematic Review. Cells 2025; 14:411. [PMID: 40136660 PMCID: PMC11941535 DOI: 10.3390/cells14060411] [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: 02/12/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Cancer is one of the leading causes of death worldwide. Evidence indicates that extracellular vesicles are involved in cancer development and may be used as promising biomarkers in cancer detection. Concomitantly, saliva constitutes a non-invasive and inexpensive source of biomarkers. This systematic review investigates the use of salivary extracellular vesicles in detecting cancers located outside of the head and neck. PubMed, Web of Science, Scopus, and Embase were thoroughly searched from database inception to 16 July 2024. Data from sixteen eligible studies were analyzed, including glioblastoma, lung, esophageal, gastric, prostate, hepatocellular, breast, and pancreatobiliary tract cancers. The findings highlight strong diagnostic potential for lung and esophageal cancers, where specific exosomal RNAs and proteins demonstrated high accuracy in distinguishing cancer patients from healthy individuals. Additionally, biomarkers in glioblastoma showed prognostic value, while those in hepatocellular and pancreatobiliary cancers exhibited potential for early detection. However, gastric and prostate cancer biomarkers showed limited reliability, and breast cancer biomarkers require further validation. In conclusion, salivary extracellular vesicles present potential in non-invasive detection across multiple cancer types; however, their diagnostic power needs further research, including standardization and large-scale validation.
Collapse
Affiliation(s)
- Wojciech Owecki
- Student’s Scientific Group in Department of Conservative Dentistry and Endodontics, Poznan University of Medical Sciences, 60-812 Poznan, Poland;
- The Student Scientific Society, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Karolina Wojtowicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Kacper Nijakowski
- Department of Conservative Dentistry and Endodontics, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| |
Collapse
|
3
|
Li C, Liu C, Ma H, Zhang Z, Zhang J. Lymphocytes-Associated Extracellular Vesicles Activate Natural Killer Cells in HNSCC. Cancer Sci 2025; 116:633-642. [PMID: 39749376 PMCID: PMC11875761 DOI: 10.1111/cas.16440] [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: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Small extracellular vesicles (sEVs) facilitate intercellular communication and play a pivotal role in tumor progression. Accumulated evidence has indicated the diversity of sEVs but with limited results revealing the landscape of heterogeneity of sEVs. The heterogeneity of cargo RNA in sEVs presents the different cell origins and indicates different functions. Here, we analyzed the heterogeneity of sEVs at droplet levels from single-cell RNA sequencing results of head and neck squamous cell carcinoma (HNSCC) with the previously reported algorithm SEVtras. With the sEVs secretion activity calculated by SEVtras, we also found that the T cells held the major role of sEVs secretion. In addition, we found these sEVs secreted by T cells increased the cytotoxic ability of natural killer cells (NK cells), which illustrated an indirect manner for the anti-tumor function of T cells. These results revealed the heterogeneity of cargo RNA of sEVs in HNSCC and underlined a sEVs-dependent manner in which T cells act on NK cells and anti-tumor immunity.
Collapse
Affiliation(s)
- Chuwen Li
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Chun Liu
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Hailong Ma
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
- Research Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghaiPeople's Republic of China
| | - Jianjun Zhang
- Department of Oral and Maxillofacial‐Head and Neck Oncology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
- College of StomatologyShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- National Center for StomatologyShanghaiPeople's Republic of China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghai Center of Head and Neck Oncology Clinical and Translational ScienceShanghaiPeople's Republic of China
| |
Collapse
|
4
|
Yu Y, Zhao Q, Cui H, Song L. CCR7 in esophageal squamous cell carcinoma: an identification from single-cell and bulk transcriptome sequencing. Discov Oncol 2025; 16:183. [PMID: 39953263 PMCID: PMC11828771 DOI: 10.1007/s12672-025-01927-3] [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: 10/22/2024] [Accepted: 02/04/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Considering the involvement of CC-chemokine receptor type 7 (CCR7) in diverse tumors, the purpose of our current study is to reveal the specific mechanisms of CCR7 in esophageal squamous cell carcinoma (ESCC). METHODS We processed single-cell RNA sequencing data based on the Gene Expression Omnibus (GEO) database and utilizing Seurat software, and performed filtering and annotation to obtain different cell types. Genes related to gene ontology biological process and Hallmark were collected, and the enrichment of genes of interested in both single cell and TCGA-ESCA was quantified. Besides, genes related to CCR7 and KRAS signaling up were uploaded to construct the protein-protein interaction network. A series of cellular assays were incorporated to test the effects of CCR7 in ESCC cells. RESULTS 28,281 cells were categorized into 4 non-immune cell classes (epithelial cells, smooth muscle cells, fibroblasts, endothelial cells) and 6 immune cell classes (mast cells, plasma B cells, B cells, neutrophils, macrophages, NK/T cells). CCR7 evidently expressed higher in epithelial cells of ESCA and was positively correlated with KRAS signaling up, inflammatory response and TNFA signaling via NFKB, with the most significant correlation witnessed between CCR7 and KRAS signaling up. Meanwhile, the positive correlation between KRAS signaling up and positive regulation of epithelial cell migration was observed. 12 common genes were related to both KRAS signaling up and positive regulation of epithelial cell migration, 3 of which (SOX9, FLT4 and HDAC9) were higher-expressed in M1 group of TCGA-ESCA. Besides, CCR7 as well as its ligands CCL19 and CCL21 was shown to express higher in ESCC cells, where increased level of immune response-related cytokines was seen. CCR7 knockdown diminished migration and proliferation as well as Macrophage M2 polarization. CONCLUSION CCR7 was highly-expressed in ESCC and positively correlated with KRAS signaling up, which may contribute to the migration of ESCC cells.
Collapse
Affiliation(s)
- Yang Yu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Qian Zhao
- Nursing Department, Shandong Medical College, Jinan, 250000, China
| | - Hongguang Cui
- Department of Anatomy, Weifang Nursing Vocational College, Qingzhou, 262500, China
| | - Liang Song
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| |
Collapse
|
5
|
Guo W, Zhou B, Dou L, Guo L, Li Y, Qin J, Wang Z, Huai Q, Xue X, Li Y, Ying J, Xue Q, Gao S, He J. Single-cell RNA sequencing and spatial transcriptomics of esophageal squamous cell carcinoma with lymph node metastases. Exp Mol Med 2025; 57:59-71. [PMID: 39741182 PMCID: PMC11799171 DOI: 10.1038/s12276-024-01369-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 01/02/2025] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) patients often face a grim prognosis due to lymph node metastasis. However, a comprehensive understanding of the cellular and molecular characteristics of metastatic lymph nodes in ESCC remains elusive. In this study involving 12 metastatic ESCC patients, we employed single-cell sequencing, spatial transcriptomics (ST), and multiplex immunohistochemistry (mIHC) to explore the spatial and molecular attributes of primary tumor samples, adjacent tissues, metastatic and non-metastatic lymph nodes. The analysis of 161,333 cells revealed specific subclusters of epithelial cells that were significantly enriched in metastatic lymph nodes, suggesting pro-metastatic characteristics. Furthermore, stromal cells in the tumor microenvironment, including MMP3+IL24+ fibroblasts, APLN+ endothelial cells, and CXCL12+ pericytes, were implicated in ESCC metastasis through angiogenesis, collagen production, and inflammatory responses. Exhausted CD8+ T cells in a cycling status were notably prevalent in metastatic lymph nodes, indicating their potential role in facilitating metastasis. We identified distinct cell-cell communication networks and specific ligand-receptor pathways. Our findings were validated through a spatial transcriptome map and mIHC. This study enhances our comprehension of the cellular and molecular aspects of metastatic lymph nodes in ESCC patients, offering potential insights into novel therapeutic strategies for these individuals.
Collapse
Affiliation(s)
- Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Minimally Invasive Therapy Research for Lung Cancer, Chinese Academy of Medical Sciences, Beijing, China
| | - Bolun Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lizhou Dou
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianjun Qin
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhen Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qilin Huai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuemin Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yin Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Minimally Invasive Therapy Research for Lung Cancer, Chinese Academy of Medical Sciences, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Key Laboratory of Minimally Invasive Therapy Research for Lung Cancer, Chinese Academy of Medical Sciences, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Wang H, Ma S, Yang Z, Niu R, Zhu H, Li S, Gao S, Li Z, Tian Y. Revolutionizing ESCC prognosis: the efficiency of tumor-infiltrating immune cells (TIIC) signature score. Discov Oncol 2025; 16:65. [PMID: 39833504 PMCID: PMC11747060 DOI: 10.1007/s12672-024-01709-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Patients suffer from esophageal squamous cell carcinoma (ESCC), which is the ninth highly aggressive malignancy. Tumor-infiltrating immune cells (TIIC) exert as major component of the tumor microenvironment (TME), showing possible prognostic value in ESCC. METHODS Transcriptome data and scRNA-seq data of ESCC samples were extracted from the GEO and TCGA databases. Tissue Specific Index (TSI) was defined to identify potential TIIC-RNAs from the TME. Twenty machine learning algorithms were further applied to evaluate the prognostic efficacy of TIIC signature score. Gene colocalization analysis was performed. Differences in CNV on chromosomes and SNP sites of prognostic model genes were calculated. RESULTS The most reliable model of TIIC signature score was developed based on three prognostic TIIC-RNAs. It showed a higher C-index than any other reported prognostic models. ESCC patients with high TIIC signature score showed poorer survival outcomes than low TIIC signature score. The activity of most immune cells decreased with the increase of TIIC score. TIIC signature score showed difference in the expression levels and methylation levels of DEGs. There was also significant different correlation with the degree of CNV amplification and CNV deletion of the immune checkpoint genes. Gene colocalization analysis showed two prognostic model genes (ATP6V0E1 and BIRC2). MR analysis found that rs148710154 and rs75146099 SNP sites of TIIC-RNA gene had a significant correlation between them gastro-oesophageal reflux and ESCC. CONCLUSION TIIC signature score was the first time developed which provided a novel strategy and guidance for the prognosis and immunotherapy of ESCC. It also gave the evidence in the important role of immune cells from the TME in the treatment of cancers.
Collapse
Affiliation(s)
- Haixia Wang
- Department of Radiation Oncology, The Fifth Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou People's Hospital, Zhengzhou, 450003, China
| | - Shaowei Ma
- Department of Gastrointestinal Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Zixin Yang
- Second Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Ren Niu
- Second Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Haiyong Zhu
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Shujun Li
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Shaolin Gao
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Zhirong Li
- Clinical Laboratory Center, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Yanhua Tian
- Second Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
7
|
Shao F, Wang Z, Ye L, Wu R, Wang J, Yu QX, Wusiman D, Tuo Z, Yoo KH, Shu Z, Wei W, Li D, Cho WC, Liu Z, Feng D. Basic helix-loop-helix ARNT like 1 regulates the function of immune cells and participates in the development of immune-related diseases. BURNS & TRAUMA 2025; 13:tkae075. [PMID: 39830193 PMCID: PMC11741524 DOI: 10.1093/burnst/tkae075] [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: 07/06/2024] [Revised: 10/26/2024] [Accepted: 11/01/2024] [Indexed: 01/22/2025]
Abstract
The circadian clock is an internal timekeeper system that regulates biological processes through a central circadian clock and peripheral clocks controlling various genes. Basic helix-loop-helix ARNT-like 1 (BMAL1), also known as aryl hydrocarbon receptor nuclear translocator-like protein 1 (ARNTL1), is a key component of the circadian clock. The deletion of BMAL1 alone can abolish the circadian rhythms of the human body. BMAL1 plays a critical role in immune cell function. Dysregulation of BMAL1 is linked to immune-related diseases such as autoimmune diseases, infectious diseases, and cancer, and vice versa. This review highlights the significant role of BMAL1 in governing immune cells, including their development, differentiation, migration, homing, metabolism, and effector functions. This study also explores how dysregulation of BMAL1 can have far-reaching implications and potentially contribute to the onset of immune-related diseases such as autoimmune diseases, infectious diseases, cancer, sepsis, and trauma. Furthermore, this review discusses treatments for immune-related diseases that target BMAL1 disorders. Understanding the impact of BMAL1 on immune function can provide insights into the pathogenesis of immune-related diseases and help in the development of more effective treatment strategies. Targeting BMAL1 has been demonstrated to achieve good efficacy in immune-related diseases, indicating its promising potential as a targetable therapeutic target in these diseases.
Collapse
Affiliation(s)
- Fanglin Shao
- Chengdu Basebio Company, Tianfu Third Street, High-Tech Zone, Chengdu 610041, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, First Ring Road, Qingyang District, Chengdu 610041, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, West Gate Street, Linhai City 317000, Zhejiang Province, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Qing-Xin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Huancheng North Road, Jiangbei District, Ningbo, Zhejiang Province, 315211, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue University, 615 W. State Street, West Lafayette, IN 47907, USA
| | - Zhouting Tuo
- Chengdu Basebio Company, Tianfu Third Street, High-Tech Zone, Chengdu 610041, China
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Jinzhai South Road, Shushan District, Hefei, Anhui 230032, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, South Korea
| | - Ziyu Shu
- Department of Earth Science and Engineering, Imperial College London, Exhibition Road, South Kensington, London SW7 2AZ, UK
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), Chongqing University, Shapingba Street, Shapingba District, Chongqing 400044, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Gascoigne Road, Yau Ma Tei, Kowloon, Hong Kong SAR, China
| | - Zhihong Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Renmin South Road, Wuhou District, Chengdu 610041, China
- Division of Surgery & Interventional Science, University College London, Gower Street, London W1T 6JF, London W1W 7TS, UK
| |
Collapse
|
8
|
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
|
9
|
Yang J, Wu B, Li G, Zhang C, Xie Y, Kong W, Zeng Z. Landscape of epithelial cell subpopulations in the human esophageal squamous cell carcinoma microenvironment. Heliyon 2024; 10:e38091. [PMID: 39391485 PMCID: PMC11466536 DOI: 10.1016/j.heliyon.2024.e38091] [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: 07/09/2024] [Revised: 08/19/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Aims We sought to reveal the landscape of epithelial cell subpopulations in the human esophageal squamous cell carcinoma microenvironment and investigate their parts on esophageal squamous carcinoma (ESCC) development. Background Epithelial cells play an important role in the occurrence and development of ESCC through multiple mechanisms. While the landscape of epithelial cell subpopulations in ESCC, remains unclear. Objective Exploring the role of epithelial cell subpopulations in ESCC progression. Methods Seurat R package was used for single-cell RNA sequencing (scRNA-seq) data filtering, dimensionality reduction, clustering and differentially expressed genes analysis. Cellmarker database was adopted for cell cluster annotation. Functional enrichment analysis was carried out by Gene Ontology (GO) analysis. InferCNV package was conducted for copy number variation (CNV) of epithelial cell subpopulations in all chromosomal regions. Pseudotime trajectory analysis was implemented for exploring differentiation trajectory of epithelial cells subgroups during the cancer progression. CellChat analysis was used for probing the interactions between epithelial cells and NK/T cells. cellular experiments were performed using Quantitative Real-Time Polymerase Chain Reaction (RT-qPCR), Wound-Healing Assay and transwell. Results 11 major cell subpopulations were identified in ESCC and adjunct tissues. Further reclassification of epithelial cells uncovered 4 subpopulations. Enrichment analysis revealed that highly expressed genes in 4 epithelial cell subpopulations were related to cell proliferation, immune response and angiogenesis. CNV analysis found that UBD + epithelial cells and GAS2L3+ epithelial cells had a higher proportion of CNV. Cell differentiation trajectories disclosed that KRT6C+ and GSTA1+ epithelial cells were in an intermediate state of differentiation, while UBD+ and GAS2L3+ epithelial cells are in an end state of differentiation during ESCC progression. Finally, we found that four epithelial cell subpopulations all inhibited NK/T cells through NECTIN2-TIGIT and CLEC2B-KLRB1. Low ATF3 and DDIT3 mRNA expression inhibited ESCC cell migration and invasion. Conclusion Here, we obtained a through epithelial cell atlas of ESCC at single-cell resolution, explored the role of epithelial cell in ESCC progression, and unveiled immunosuppressive signals to NK/T cells in promoting ESCC. Our findings expand the comprehension of epithelial cells and offer a theoretical guidance for future anti-epithelial cell treatment of ESCC.
Collapse
Affiliation(s)
- Jingrong Yang
- Department of Cardiothoracic Surgery, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
| | - Bo Wu
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
- Department of Emergency, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
| | - Guo Li
- Department of Cardiothoracic Surgery, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
| | - Chenxi Zhang
- Department of Cardiothoracic Surgery, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
| | - Yongwei Xie
- Department of Cardiothoracic Surgery, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
| | - Wencui Kong
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
- Department of Respiratory Medicine and Critical Care Medicine, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
| | - Zhiyong Zeng
- Department of Cardiothoracic Surgery, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, 350025, China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350122, China
| |
Collapse
|
10
|
Huang Q, Zhong X, Li J, Hu R, Yi J, Sun J, Xu Y, Zhou X. Exosomal ncRNAs: Multifunctional contributors to the immunosuppressive tumor microenvironment of hepatocellular carcinoma. Biomed Pharmacother 2024; 173:116409. [PMID: 38460375 DOI: 10.1016/j.biopha.2024.116409] [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: 12/05/2023] [Revised: 02/23/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant liver cancer characterized by aggressive progression, unfavorable prognosis, and an increasing global health burden. Therapies that precisely target immunological checkpoints and immune cells have gained significant attention as possible therapeutics in recent years. In truth, the efficacy of immunotherapy is heavily contingent upon the tumor microenvironment (TME). Recent studies have indicated that exosomes serve as a sophisticated means of communication among biomolecules, executing an essential part in the TME of immune suppression. Exosomal non-coding RNAs (ncRNAs) can induce the activation of tumor cells and immunosuppressive immune cells that suppress the immune system, such as cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), CD+8 T cells, regulatory T cells (Tregs), and regulatory B cells (Bregs). This cell-cell crosstalk triggered by exosomal ncRNAs promotes tumor proliferation and metastasis, angiogenesis, malignant phenotype transformation, and drug resistance. Hence, it is imperative to comprehend how exosomal ncRNAs regulate tumor cells or immune cells within the TME to devise more comprehensive and productive immunotherapy programs. This study discusses the features of exosomal ncRNAs in HCC and how the activation of the exosomes redefines the tumor's immunosuppressive microenvironment, hence facilitating the advancement of HCC. Furthermore, we also explored the potential of exosomal ncRNAs as a viable biological target or natural vehicle for HCC therapy.
Collapse
Affiliation(s)
- Qi Huang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao PR China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Xin Zhong
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Jing Li
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao PR China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Rui Hu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao PR China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Jinyu Yi
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao PR China; Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Jialing Sun
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao PR China.
| | - Xiaozhou Zhou
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, PR China; Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, PR China.
| |
Collapse
|
11
|
Guo W, Zhou B, Zhao L, Huai Q, Tan F, Xue Q, Lv F, Gao S, He J. Plasma extracellular vesicle long RNAs predict response to neoadjuvant immunotherapy and survival in patients with non-small cell lung cancer. Pharmacol Res 2023; 196:106921. [PMID: 37709184 DOI: 10.1016/j.phrs.2023.106921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
Neoadjuvant immunotherapy has brought new hope for patients with non-small cell lung cancer (NSCLC). However, limited by the lack of clinically feasible markers, it is still difficult to select NSCLC patients who respond well and to predict patients' clinical outcomes before the treatment. Before the treatment, we isolated plasma extracellular vesicles (EVs) from three cohorts (discovery, training and validation) of 78 NSCLC patients treated with neoadjuvant immunotherapy. To identify differentially-expressed EV long RNAs (exLRs), we employed RNA-seq in the discovery cohort. And we subsequently used qRT-PCR to establish and validate the predictive signature in the other two cohorts. We have identified 8 candidate exLRs from 27 top-ranked exLRs differentially expressed between responders and non-responders, and tested their expression with qRT-PCR in the training cohort. We finally identified H3C2 (P = 0.029), MALAT1 (P = 0.043) and RPS3 (P = 0.0086) significantly expressed in responders for establishing the predictive signature. Integrated with PD-L1 expression, our signature performed well in predicting immunotherapeutic responses in the training (AUC=0.892) and validation cohorts (AUC=0.747). Furthermore, our signature was proven to be a predictor for favorable prognosis of patients treated with neoadjuvant immunotherapy, which demonstrates the feasibility of our signature in clinical practices (P = 0.048). Our results demonstrate that the exLR-based signature could accurately predict responses to neoadjuvant immunotherapy and prognosis in NSCLC patients.
Collapse
Affiliation(s)
- Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China; Key Laboratory of Minimally Invasive Therapy Research for Lung Cancer, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Bolun Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Liang Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qilin Huai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China; Key Laboratory of Minimally Invasive Therapy Research for Lung Cancer, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China; Key Laboratory of Minimally Invasive Therapy Research for Lung Cancer, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Fang Lv
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China; Key Laboratory of Minimally Invasive Therapy Research for Lung Cancer, Chinese Academy of Medical Sciences, Beijing, People's Republic of China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|