1
|
Yuan Y, Jiang H, Xue R, Feng X, Liu B, Li L, Peng B, Ren C, Li S, Li N, Li M, Wang D, Zhang X. Identification of a Biomarker Panel in Extracellular Vesicles Derived From Non-Small Cell Lung Cancer (NSCLC) Through Proteomic Analysis and Machine Learning. J Extracell Vesicles 2025; 14:e70078. [PMID: 40366616 PMCID: PMC12077270 DOI: 10.1002/jev2.70078] [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: 09/20/2024] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Antigen fingerprint profiling of tumour-derived extracellular vesicles (TDEVs) in the body fluids is a promising strategy for identifying tumour biomarkers. In this study, proteomic and immunological assays reveal significantly higher CD155 levels in plasma extracellular vesicles (EVs) from patients with non-small cell lung cancer (NSCLC) than from healthy individuals. Utilizing CD155 as a bait protein on the EV membrane, CD155+ TDEVs are enriched from NSCLC patient plasma EVs. In the discovery cohort, 281 differentially expressed proteins are identified in TDEVs of the NSCLC group compared with the healthy control group. In the verification cohort, 49 candidate biomarkers are detected using targeted proteomic analysis. Of these, a biomarker panel of seven frequently and stably detected proteins-MVP, GYS1, SERPINA3, HECTD3, SERPING1, TPM4, and APOD-demonstrates good diagnostic performance, achieving an area under the curve (AUC) of 1.0 with 100% sensitivity and specificity in receiver operating characteristic (ROC) curve analysis, and 92.3% sensitivity and 88.9% specificity in confusion matrix analysis. Western blotting results confirm upregulation trends for MVP, GYS1, SERPINA3, HECTD3, SERPING1 and APOD, and TPM4 is downregulated in EVs of NSCLC patients compared with healthy individuals. These findings highlight the potential of this biomarker panel for the clinical diagnosis of NSCLC.
Collapse
Affiliation(s)
- Ye Yuan
- College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanP. R. China
- Key Laboratory of Biomacromolecules (CAS), Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Hai Jiang
- Renmin HospitalHubei University of MedicineShiyanP. R. China
| | - Rui Xue
- Renmin HospitalHubei University of MedicineShiyanP. R. China
| | - Xiao‐Jun Feng
- College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanP. R. China
| | - Bi‐Feng Liu
- College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanP. R. China
| | - Lian Li
- Renmin HospitalHubei University of MedicineShiyanP. R. China
| | - Bo Peng
- Key Laboratory of Biomacromolecules (CAS), Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Chen‐Shuo Ren
- Key Laboratory of Biomacromolecules (CAS), Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Shi‐Min Li
- Key Laboratory of Biomacromolecules (CAS), Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Na Li
- Key Laboratory of Biomacromolecules (CAS), Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Min Li
- Key Laboratory of Biomacromolecules (CAS), Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Dian‐Bing Wang
- Key Laboratory of Biomacromolecules (CAS), Institute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Xian‐En Zhang
- Key Laboratory of Biomacromolecules (CAS), Institute of BiophysicsChinese Academy of SciencesBeijingChina
- Faculty of Synthetic BiologyShenzhen University of Advanced TechnologyShenzhenChina
| |
Collapse
|
2
|
Liu YQ, Zheng ZJ, Fang WK, Li YS, Li C, Yang M, Han DC, Zhou JH, Xie YH, Zhang YY, Kang ZY, Xu YW, Xie JJ. Interplay and cooperation between GLI2 and master transcription factors promote progression of esophageal squamous cell carcinoma. Am J Hum Genet 2025; 112:1039-1061. [PMID: 40157366 PMCID: PMC12120187 DOI: 10.1016/j.ajhg.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025] Open
Abstract
The establishment of gene expression programs that drive cell identity is governed by tightly regulated transcription factors (TFs) that engage in auto- and cross-regulation in a feedforward manner, forming core regulatory circuitries (CRCs). Here, we identify and validate an important interconnected CRC formed by three master TFs-GLI2, TP63, and RUNX1-in esophageal squamous cell carcinoma (ESCC). Furthermore, master TFs co-bind to their own and each other's super-enhancers, forming an interconnected auto-regulatory loop. Mechanistically, these master TFs occupy the majority of ESCC super-enhancers and cooperatively orchestrate the ESCC transcription program. Functionally, GLI2, a master TF, is essential for ESCC viability, migration, invasion, and the growth of xenograft tumors. Moreover, the overexpression of GLI2 is significantly associated with shorter overall survival of patients with ESCC. Downstream, this CRC apparatus coordinately regulates gene expression networks in ESCC, controlling important cancer-promoting pathways, including Hedgehog, glycolysis, and epidermal growth factor receptor signaling pathways. Together, these findings offer significant mechanistic insights into the transcriptional dysregulation in ESCC and recognize GLI2 as a potential therapeutic target and prognostic marker for ESCC. More importantly, CRC-downstream genes and signaling pathways may contain potential therapeutic targets for this malignancy.
Collapse
Affiliation(s)
- Yin-Qiao Liu
- Department of Clinical Laboratory Medicine, Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, the Cancer Hospital of Shantou University Medical College, Shantou 515041, China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Ze-Jun Zheng
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Institute of Biochemistry & Molecular Biology, Institute of Aging Research, Guangdong Medical University, Dongguan 523808, P.R. China
| | - Wang-Kai Fang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Yan-Shang Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Chun Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Min Yang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Dong-Chen Han
- Beijing University of Chinese Medicine, Fangshan 102400, China
| | - Jun-Hua Zhou
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Ying-Hua Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Yu-Ying Zhang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Zhuo-Ying Kang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, the Cancer Hospital of Shantou University Medical College, Shantou 515041, China.
| | - Jian-Jun Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China; Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Institute of Biochemistry & Molecular Biology, Institute of Aging Research, Guangdong Medical University, Dongguan 523808, P.R. China.
| |
Collapse
|
3
|
Pan Y, Sun Y, Xiao Y, Ding J, Hu G, Lin Z, Chen C. DOCK9 as a predictive biomarker linked to angiogenesis and immune response in esophageal squamous cell carcinoma. Clin Exp Med 2025; 25:126. [PMID: 40272582 PMCID: PMC12021961 DOI: 10.1007/s10238-025-01653-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/26/2025] [Indexed: 04/25/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) remains a serious health concern due to its high prevalence and mortality rates. Identifying prognostic biomarkers is essential to improving patient outcomes and treatment strategies. DOCK9, a gene implicated in various cellular functions, may play a significant role in ESCC progression and prognosis. We analyzed RNA microarray datasets and single-cell RNA sequencing data to identify survival-associated genes in ESCC. Using protein expression analysis, we examined DOCK9 in ESCC tissues and assessed its functional impact on human umbilical vein endothelial cells to understand its role in angiogenesis. Additionally, we developed a 21-gene prognostic risk model, focusing on the relevance of DOCK9. Our findings revealed that DOCK9 expression is significantly reduced in ESCC tissues and correlates with poor survival outcomes. Functionally, DOCK9 was found to regulate angiogenesis and modulate the tumor-associated fibroblast environment in ESCC. Furthermore, the DOCK9/CD31 ratio emerged as a potential marker to predict immune therapy response in ESCC. DOCK9 serves as a prognostic biomarker in ESCC, influencing both angiogenesis and immune response, and could guide future therapeutic strategies, particularly in immunotherapy. This study highlights DOCK9's relevance in ESCC prognosis, supporting its potential role in tailored therapies aimed at angiogenesis and immune modulation.
Collapse
Affiliation(s)
- Yaqiang Pan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People's Republic of China
| | - Yangyong Sun
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Ying Xiao
- Department of Emergency, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, 211100, Jiangsu, People's Republic of China
| | - Jifei Ding
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Ge Hu
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Zhiqiang Lin
- Department of Otolaryngology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Suzhou, 215008, Jiangsu, People's Republic of China.
| | - Chang Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People's Republic of China.
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
4
|
Zhao LL, Liu YJ, Guo QJ, Yan N, Yang J, Han JQ, Xie XH, Luo YS. TPM4 influences the initiation and progression of gastric cancer by modulating ferroptosis via SCD1. Clin Exp Med 2025; 25:115. [PMID: 40214825 PMCID: PMC11991984 DOI: 10.1007/s10238-025-01629-8] [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: 01/31/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025]
Abstract
Gastric cancer (GC) is a deadly disease with poor prognosis and few treatment options. Tropomyosin 4 (TPM4) is an actin-binding protein that stabilizes the cytoskeleton of cells and has an unclear role in GC. This study aimed to elucidate the role and underlying mechanisms of TPM4 in GC pathogenesis. The expression and diagnostic and prognostic value of TPM4 in GC were analyzed using bioinformatics. A nomogram based on TPM4 expression was created and validated with an external cohort. TPM4-knockdown GC cells and xenograft models in nude mice were used to study the function of TPM4 in vitro and in vivo. Proteomic and rescue experiments confirmed the regulatory effect of TPM4 on stearoyl-CoA desaturase 1 (SCD1) in GC. Immunohistochemistry verified the expression and correlation of the TPM4 and SCD1 proteins in GC tissues. Our study identified TPM4 as an oncogene in GC, suggesting its potential diagnostic and prognostic value. The TPM4-based nomogram showed potential prognostic value for clinical use. TPM4 knockdown inhibited GC cell proliferation, induced ferroptosis, and slowed tumor growth in vivo, which is achieved by inhibiting SCD1 expression. Immunohistochemical analysis of GC tissues revealed elevated expression levels of both TPM4 and SCD1 proteins, with a positive correlation observed between their expression. TPM4 is a promising target for new diagnostic, prognostic, and therapeutic strategies for GC. Downregulation of TPM4 inhibits GC cell growth and induces ferroptosis by suppressing SCD1 expression.
Collapse
Affiliation(s)
- Ling-Lin Zhao
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
- Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Yu-Jun Liu
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, 810001, China
| | - Qi-Jing Guo
- Department of Oncology, Air Force Medical Center, PLA, Beijing, 100142, China
| | - Nan Yan
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
| | - Jie Yang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China
| | - Jing-Qi Han
- Department of Pathology, Affiliated Hospital of Qinghai University, Xining, 810001, China
| | - Xiao-Hong Xie
- Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Yu-Shuang Luo
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, China.
- Department of Oncology, Affiliated Hospital of Qinghai University, Xining, 810001, China.
| |
Collapse
|
5
|
Huang J, Zhang H, Lin X, Wu X, Chen X, Chen W, Liang S, Chen Y, Luo Q, Xu C, Liu S, Liu X, Zhang S. Regulatory T Cell Infiltration-Driven Single-Cell Transcriptomic Analysis Identifies SAP18 as a Prognostic Marker for Esophageal Squamous Cell Carcinoma. J Gastrointest Cancer 2025; 56:97. [PMID: 40208395 DOI: 10.1007/s12029-025-01174-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Advanced esophageal squamous cell carcinoma (ESCC) is characterized by molecular heterogeneity and distinct patterns of immune cell infiltration. Regulatory T cells (Tregs), in particular, play a critical role in shaping an immunosuppressive tumor microenvironment (TME), which is associated with poor clinical outcomes. METHODS We developed a prognostic model by integrating GEO-derived bulk RNA sequencing data and single-cell transcriptome. Model predictions were confirmed through RT-qPCR, Western blot, and immunohistochemistry on clinical specimens, while in vitro assays (CCK8, transwell invasion, scratch, colony formation, and immunofluorescence) validated the function of SAP18 in cell proliferation, invasion, and ECM remodeling. RESULTS Expression patterns of the 5 Tregs-associated genes in clinical specimens aligned with model predictions, underscoring the model's robustness. The high-risk subgroup was associated with upregulated extracellular matrix (ECM) remodeling, an abundance of immune-suppressive cells, higher TP53 mutation rate, and limited benefit from immunotherapy. In contrast, the low-risk subgroup exhibited anti-tumor immunity. Cell-cell communication analysis also implicated the collagen pathway in Tregs-mediated immune evasion in ESCC. Functional assays indicated that SAP18 in the prognostic model significantly promotes proliferation, invasion, and ECM reconstruction, further highlighting its potential as a therapeutic target. CONCLUSION Our findings elucidate the role of Tregs in the TME, underscoring significant potential of SAP18, which is essential for assessing patient prognosis and may facilitate the development of personalized therapies for ESCC.
Collapse
Affiliation(s)
- Jianxiang Huang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China
| | - Hanshuo Zhang
- Gastrointestinal Anorectal Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Xinyue Lin
- Department of Pharmacology, Medical College of Shantou University, Shantou, 515063, PR China
| | - Xiaolong Wu
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China
| | - Xiaoshan Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Wang Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Shanshan Liang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Yun Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Qianhua Luo
- Department of Pharmacy, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China
| | - Chengcheng Xu
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China
| | - Shaojie Liu
- Gastrointestinal Anorectal Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China
| | - Xingmei Liu
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China.
- Department of Nursing, Guangzhou Red Cross Hospital of Jinan University, Haizhu District, No. 396, Tongfuzhong Road, Guangzhou, 510220, PR China.
| | - Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, PR China.
- College of Pharmacy, Jinan University, Guangzhou, 510220, PR China.
| |
Collapse
|
6
|
Harris A, Burnham K, Pradhyumnan R, Jaishankar A, Häkkinen L, Góngora-Rosero RE, Piazza Y, Andl CD, Andl T. Human-Specific Organization of Proliferation and Stemness in Squamous Epithelia: A Comparative Study to Elucidate Differences in Stem Cell Organization. Int J Mol Sci 2025; 26:3144. [PMID: 40243939 PMCID: PMC11989042 DOI: 10.3390/ijms26073144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
The mechanisms that influence human longevity are complex and operate on cellular, tissue, and organismal levels. To better understand the tissue-level mechanisms, we compared the organization of cell proliferation, differentiation, and cytoprotective protein expression in the squamous epithelium of the esophagus between mammals with varying lifespans. Humans are the only species with a quiescent basal stem cell layer that is distinctly physically separated from parabasal transit-amplifying cells. In addition to these stark differences in the organization of proliferation, human squamous epithelial stem cells express DNA repair-related markers, such as MECP2 and XPC, which are absent or low in mouse basal cells. Furthermore, we investigated whether the transition from basal to suprabasal is different between species. In humans, the parabasal cells seem to originate from cells detaching from the basement membrane, and these can already begin to proliferate while delaminating. In most other species, delaminating cells have been rare or their proliferation rate is different from that of their human counterparts, indicating an alternative mode of how stem cells maintain the tissue. In humans, the combination of an elevated cytoprotective signature and novel tissue organization may enhance resistance to aging and prevent cancer. Our results point to enhanced cellular cytoprotection and a tissue architecture which separates stemness and proliferation. These are both potential factors contributing to the increased fitness of human squamous epithelia to support longevity by suppressing tumorigenesis. However, the organization of canine oral mucosa shows some similarities to that of human tissue and may provide a useful model to understand the relationship between tissue architecture, gene expression regulation, tumor suppression, and longevity.
Collapse
Affiliation(s)
- Ashlee Harris
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA (K.B.); (R.P.); (R.E.G.-R.)
| | - Kaylee Burnham
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA (K.B.); (R.P.); (R.E.G.-R.)
| | - Ram Pradhyumnan
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA (K.B.); (R.P.); (R.E.G.-R.)
| | - Arthi Jaishankar
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA (K.B.); (R.P.); (R.E.G.-R.)
| | - Lari Häkkinen
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada;
| | - Rafael E. Góngora-Rosero
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA (K.B.); (R.P.); (R.E.G.-R.)
| | - Yelena Piazza
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Claudia D. Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA (K.B.); (R.P.); (R.E.G.-R.)
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA (K.B.); (R.P.); (R.E.G.-R.)
| |
Collapse
|
7
|
Jang J, Ko KP, Zhang J, Jun S, Park JI. Deciphering Precursor Cell Dynamics in Esophageal Preneoplasia via Genetic Barcoding and Single-Cell Transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.637920. [PMID: 40060545 PMCID: PMC11888434 DOI: 10.1101/2025.02.26.637920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Cancer cells exhibit high heterogeneity and lineage plasticity, complicating studies of tumorigenesis and development of therapies. Recently, preneoplastic cells, although histologically normal, have been shown to possess high plasticity and early genetic alterations, yet their origins and lineage trajectories remain unclear. Herein, we introduce a lineage-tracing tool integrating genetic barcoding with single-cell RNA sequencing to map preneoplastic esophageal cell lineages. We identified preneoplastic precursor cells (PNPCs) as a distinct progenitor-like population with unique transcriptional profiles and high plasticity, contributing to proliferative and basal cell populations. To enhance lineage mapping, we developed the eXamined Ridge (XR) score, accurately identifying high-plasticity cells. Nfib and Qk emerged as conserved PNPC markers, peaking in early preneoplasia and declining after malignant transformation. These findings reveal PNPCs as key players in early tumorigenesis and highlight their potential as biomarkers for early cancer detection and therapeutic intervention, offering new strategies for preventing esophageal cancer progression.
Collapse
Affiliation(s)
- Jinho Jang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kyung-Pil Ko
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sohee Jun
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
8
|
Zhang J, Jia F, Li C, Song S, Gong A. Unveiling SSR4: a promising biomarker in esophageal squamous cell carcinoma. Front Immunol 2025; 16:1544154. [PMID: 40066443 PMCID: PMC11891195 DOI: 10.3389/fimmu.2025.1544154] [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: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 05/13/2025] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) represents a frequent cancer with a poor prognosis. Altered glucose metabolism contributes factor to ESCC progression. In our previous study, signal sequence receptor subunit delta (SSR4) was included in an ESCC prognostic model; however, the mechanisms underlying SSR4 implication in ESCC remain ambiguous. Accordingly, we aim to determine the interconnection between SSR4 expression and clinical characteristics of ESCC. Methods This differential expression and prognostic significance of SSR4 was performed using bulk RNA-seq data and 110 patients with complete follow-up information. The ESCC cell subsets with the highest gene expression levels were identified with single-cell data. Gene function and enrichment, immune infiltration, cell communication, and molecular docking analyses were performed. Results Unlike adjacent non-cancerous tissues, SSR4 was overexpressed in ESCC tissues, validated by both reverse transcription-qPCR and IHC staining. SSR4 expression was related to the N stage, lymph node metastasis, and AJCC TNM classification stage. Patients exhibiting low SSR4 expression had a more favorable prognosis. The highest SSR4 expression was recognized in tumor plasma cells. Continued exploration of immune infiltration highlighted a close association between SSR4 gene expression and the infiltration of immune cells such as plasma cells. On dividing cells into SSR4-positive and -negative groups, CellChat analysis indicated that SSR4 may regulate the interactions that existed between ESCC tumor plasma cells and the tumor microenvironment (TME) by modulating the MIF/CD74/CXCR4 axis. Conclusion The SSR4 gene may have significant relevance with clinical pathological factors, and play a critical role in the regulation of tumor microenvironment of ESCC patients. Overall, SSR4 may be a promising ESCC biomarker with prospective applicability in clinical diagnosis as well as the development of targeted treatment approaches in patients of ESCC.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Department of Digestive Endoscopy, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Fang Jia
- Department of Digestive Endoscopy, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chuqiao Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Shunzhe Song
- Department of Digestive Endoscopy, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Aixia Gong
- Department of Digestive Endoscopy, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
9
|
Maleka MN, Mbita Z, Morafo V. Regulation of Granzymes A and B by High-Risk HPV: Impact on Immune Evasion and Carcinogenesis. Viruses 2025; 17:221. [PMID: 40006976 PMCID: PMC11861749 DOI: 10.3390/v17020221] [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: 12/06/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
The number of new cancer cases is soaring, and currently, there are 440.5 per 100,000 new cases reported every year. A quarter of these are related to human papillomavirus (HPV) infections, particularly types 16 and 18. These include oropharyngeal, anal, vaginal, and penile cancers. A critical aspect of their oncogenic potential lies in their ability to manipulate host immune responses, facilitating immune evasion and carcinogenesis. High-risk HPVs target key immune components like granzymes A and B and MHC-I, which are crucial for the elimination of virus-infected and transformed cells, thereby weakening immune surveillance. Evidence suggests that high-risk HPVs downregulate the expression of tumor suppressors, such as p53 and pRB, and the activity of these immune components, weakening CTL and NK cell responses, thus enabling persistent infection and carcinogenesis. We discuss the implications of granzyme and MHC-I dysregulation for immune evasion, tumor progression, and potential therapeutic strategies. This review further explores the regulation of granzyme A, B, and MHC-I by high-risk HPVs, focusing on how viral oncoproteins, E6 and E7, interfere with granzyme-mediated cytotoxicity and antigen presentation. The complex interplay between high-risk HPVs, granzyme A, granzyme B, and MHC-I may provide insights into novel approaches for targeting HPV-associated cancers.
Collapse
Affiliation(s)
| | | | - Vivian Morafo
- Department of Biochemistry, Microbiology and Biotechnology, School of Molecular and Life Sciences, Private Bag X 1106, Sovenga, Polokwane 0727, South Africa; (M.N.M.); (Z.M.)
| |
Collapse
|
10
|
She Y, Wang C, Zhao Q. Improving doublet cell removal efficiency through multiple algorithm runs. Comput Struct Biotechnol J 2025; 27:451-460. [PMID: 39911841 PMCID: PMC11794957 DOI: 10.1016/j.csbj.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 02/07/2025] Open
Abstract
Doublets are a key confounding factor in the analysis of scRNA-seq data, as they can interfere with differential expression analysis and disrupt developmental trajectories. However, due to the randomness of the algorithms, most doublet removal methods still leave a certain proportion of doublets after application. In this study, we proposed a multi-round doublet removal (MRDR) strategy, that ran the algorithm in cycles multiple times to effectively reduce randomness while enhancing the effectiveness of doublet removal. We evaluated the MRDR strategy in 14 real-world datasets, 29 barcoded scRNA-seq datasets, and 106 synthetic datasets with four popular doublet detection tools, including DoubletFinder, cxds, bcds, and hybrid. We found that in real-world datasets, the DoubletFinder had a better performance in MRDR strategy compared to a single removal of doublets and the recall rate improved by 50 % for two rounds of doublet removal compared to one round, and the performance of the other three doublet algorithms improved the ROC by about 0.04. In barcoded scRNA-seq datasets, we found that using cxds for two rounds of doublet removal yielded the best results. Subsequently, in simulated datasets, we proved that the multi-round removal strategy was more effective in removing doublets than a single removal, with cxds showing the best results when applied twice, and the ROC of the four methods during the two rounds of removal improved by at least 0.05 compared to single removal. Finally, compared to running the algorithm once, we found that the MRDR strategy was more beneficial for differential gene expression analysis and cell trajectory inference when using default analysis parameters. Overall, we proved that the MRDR strategy was more effective in removing doublets and advantageous for downstream analyses, and the strategy could be incorporated into the standard analysis pipeline for scRNA-seq experiments and recommend using cxds to remove doublets through two rounds of algorithm iteration.
Collapse
Affiliation(s)
- Yong She
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Chaoye Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Qi Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| |
Collapse
|
11
|
Chen X, Agustinus AS, Li J, DiBona M, Bakhoum SF. Chromosomal instability as a driver of cancer progression. Nat Rev Genet 2025; 26:31-46. [PMID: 39075192 DOI: 10.1038/s41576-024-00761-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/31/2024]
Abstract
Chromosomal instability (CIN) refers to an increased propensity of cells to acquire structural and numerical chromosomal abnormalities during cell division, which contributes to tumour genetic heterogeneity. CIN has long been recognized as a hallmark of cancer, and evidence over the past decade has strongly linked CIN to tumour evolution, metastasis, immune evasion and treatment resistance. Until recently, the mechanisms by which CIN propels cancer progression have remained elusive. Beyond the generation of genomic copy number heterogeneity, recent work has unveiled additional tumour-promoting consequences of abnormal chromosome segregation. These mechanisms include complex chromosomal rearrangements, epigenetic reprogramming and the induction of cancer cell-intrinsic inflammation, emphasizing the multifaceted role of CIN in cancer.
Collapse
Affiliation(s)
- Xuelan Chen
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Albert S Agustinus
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Jun Li
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melody DiBona
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samuel F Bakhoum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
12
|
Li R, Li N, Yang Q, Tong X, Wang W, Li C, Zhao J, Jiang D, Huang H, Fang C, Xie K, Yuan J, Chen S, Li G, Luo H, Gao Z, Wu D, Cui X, Jiang W, Guo L, Ma H, Feng Y. Spatial transcriptome profiling identifies DTX3L and BST2 as key biomarkers in esophageal squamous cell carcinoma tumorigenesis. Genome Med 2024; 16:148. [PMID: 39696540 DOI: 10.1186/s13073-024-01422-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Understanding the stepwise progression of esophageal squamous cell carcinoma (ESCC) is crucial for developing customized strategies for early detection and optimal clinical management. Herein, we aimed to unravel the transcriptional and immunologic alterations occurring during malignant transformation and identify clinically significant biomarkers of ESCC. METHODS Digital spatial profiling (DSP) was performed on 11 patients with early-stage ESCC (pT1) to explore the transcriptional alterations in epithelial, immune cell, and non-immune cell stromal compartments across regions of distinct histology, including normal tissues, low- and high-grade dysplasia, and cancerous tissues. Furthermore, single-cell spatial transcriptomics was performed using the CosMx Spatial Molecular Imaging (SMI) system on 4 additional patients with pT1 ESCC. Immunohistochemical (IHC) analysis was performed on consecutive histological sections of 20 pT1 ESCCs. Additionally, public bulk and single-cell RNA-sequencing (scRNA-seq) datasets were analyzed, and in vitro and in vivo functional studies were conducted. RESULTS Spatial transcriptional reprogramming and dynamic cell signaling pathways that determined ESCC progression were delineated. Increased infiltration of macrophages from normal tissues through dysplasia to cancerous tissues occurred. Macrophage subtypes were characterized using the scRNA-seq dataset. Cell-cell communication analysis of scRNA-seq and SMI data indicated that the migration inhibitory factor (MIF)-CD74 axis may exhibit pro-tumor interactions between macrophages and epithelial cells. DSP, SMI, and IHC data demonstrated that DTX3L expression in epithelial cells and BST2 expression in stromal cells increased gradually with ESCC progression. Functional studies demonstrated that DTX3L or BST2 knockdown inhibited ESCC proliferation and migration and decreased M2 polarization of tumor-associated macrophages. CONCLUSIONS Spatial profiling comprehensively characterized the molecular and immunological hallmarks from normal tissue to ESCC, guiding the way to a deeper understanding of the tumorigenesis and progression of this disease and contributing to the prevention of ESCC. Within this exploration, we uncovered biomarkers that exhibit a robust correlation with ESCC progression, offering potential new avenues for insightful therapeutic approaches.
Collapse
Affiliation(s)
- Rutao Li
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Institute of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Department of Thoracic Surgery, the Fourth Affiliated Hospital Affiliated to Soochow University, Suzhou, 215000, China
| | - Na Li
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, 518000, China.
| | - Qianqian Yang
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Xing Tong
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Wei Wang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, 518000, China
| | - Chang Li
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Institute of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Jun Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Institute of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Dong Jiang
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Institute of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Haitao Huang
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Institute of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Chen Fang
- Department of Thoracic Surgery, the Fourth Affiliated Hospital Affiliated to Soochow University, Suzhou, 215000, China
| | - Kai Xie
- Department of Thoracic Surgery, the Fourth Affiliated Hospital Affiliated to Soochow University, Suzhou, 215000, China
| | - Jiamin Yuan
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Shaomu Chen
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Institute of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Guangbin Li
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Institute of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Haitao Luo
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, 518000, China
| | - Zhibo Gao
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, 518000, China
| | - Dongfang Wu
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, 518000, China
| | - Xiaoli Cui
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, 518000, China
| | - Wei Jiang
- Department of Thoracic Surgery, the Fourth Affiliated Hospital Affiliated to Soochow University, Suzhou, 215000, China
| | - Lingchuan Guo
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
| | - Haitao Ma
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
- Department of Thoracic Surgery, the Fourth Affiliated Hospital Affiliated to Soochow University, Suzhou, 215000, China.
| | - Yu Feng
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
- Institute of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
| |
Collapse
|
13
|
Zhang N, Zhao L, Li J, Li H, Chen Y. Harnessing Nanotechnology for Gout Therapy: Colchicine-Loaded Nanoparticles Regulate Macrophage Polarization and Reduce Inflammation. Biomater Res 2024; 28:0089. [PMID: 39665079 PMCID: PMC11632155 DOI: 10.34133/bmr.0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 12/13/2024] Open
Abstract
Gout is a disease caused by hyperuricemia, characterized by inflammation reactions triggered by macrophage polarization. Colchicine is a commonly used drug for gout treatment, but its mechanism of action remains unclear. The aim of this study was to investigate the regulatory effect of colchicine on macrophage polarization to enhance the therapeutic effectiveness against gout inflammation. To accomplish this, a mouse model was established, and peripheral blood mononuclear cell samples were collected. Single-cell RNA sequencing was employed to reveal cellular heterogeneity and identify key genes. Molecular docking and experimental validation were performed to confirm the binding between the key genes and colchicine. Lentiviral intervention and biochemical indicator detection were conducted to assess the impact of key genes on gout mice. Additionally, the therapeutic effect of colchicine incorporated into neutrophil membrane-coated nanoparticles was investigated. The study found that macrophage polarization plays a critical role in gout, and AHNAK was identified as the key gene through which colchicine affects macrophage polarization. Lentiviral intervention to decrease AHNAK expression was shown to alleviate joint swelling in gout mice and regulate macrophage polarization. Colchicine encapsulated in R4F peptide-modified neutrophil membrane-coated Pluronic F127 nanoparticle (R4F-NM@F127) nanocarriers inhibited M1 macrophage polarization, induced M2 macrophage polarization, alleviated gout, and minimized toxicity to normal tissues. Colchicine suppressed M1 macrophage polarization and induced M2 macrophage polarization by binding to AHNAK protein, thereby alleviating gout. Colchicine incorporated into R4F-NM@F127 nanocarriers can serve as a targeted therapeutic drug to regulate macrophage polarization, alleviate gout, and reduce toxicity to normal tissues.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Rheumatology and Immunology,
Shengjing Hospital Affiliated to China Medical University, Shenyang 110000, China
| | - Lanqing Zhao
- Department of Sleep Medicine Center, The Shengjing Affiliated Hospital,
China Medical University, Shenyang 110000, Liaoning, China
| | - Jinwei Li
- Department of Neurology/Stroke Center, the First Affiliated Hospital ofChina Medical University, China Medical University, Shenyang 110000, Liaoning, China
| | - Hongxi Li
- Department of Pain Management,
Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Yu Chen
- Department of The Fourth Otolaryngology Head and Neck Surgery,
Shengjing Hospital of China Medical University, Shenyang 110000, China
| |
Collapse
|
14
|
Liao Y, Gao IH, Kusakabe T, Lin WY, Grier A, Pan X, Morzhanaeva O, Shea TP, Yano H, Karo-Atar D, Olsen KA, Oh JH, Vandegrift KJ, King IL, Cuomo CA, Artis D, Rehermann B, Lipman N, Iliev ID. Fungal symbiont transmitted by free-living mice promotes type 2 immunity. Nature 2024; 636:697-704. [PMID: 39604728 PMCID: PMC11733984 DOI: 10.1038/s41586-024-08213-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/14/2024] [Indexed: 11/29/2024]
Abstract
The gut mycobiota is crucial for intestinal homeostasis and immune function1. Yet its variability and inconsistent fungal colonization of laboratory mice hinders the study of the evolutionary and immune processes that underpin commensalism2,3. Here, we show that Kazachstania pintolopesii is a fungal commensal in wild urban and rural mice, with an exceptional ability to colonize the mouse gastrointestinal tract and dominate the gut mycobiome. Kazachstania pintolopesii colonization occurs in a bacteria-independent manner, results in enhanced colonization resistance to other fungi and is shielded from host immune surveillance, allowing commensal presence. Following changes in the mucosal environment, K. pintolopesii colonization triggers a type 2 immune response in mice and induces gastrointestinal eosinophilia. Mechanistically, we determined that K. pintolopesii activates type 2 immunity via the induction of epithelial IL-33 and downstream IL-33-ST2 signalling during mucus fluctuations. Kazachstania pintolopesii-induced type 2 immunity enhanced resistance to helminth infections or aggravated gastrointestinal allergy in a context-dependent manner. Our findings indicate that K. pintolopesii is a mouse commensal and serves as a valuable model organism for studying gut fungal commensalism and immunity in its native host. Its unnoticed presence in mouse facilities highlights the need to evaluate its influence on experimental outcomes and phenotypes.
Collapse
Affiliation(s)
- Yun Liao
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Iris H Gao
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Takato Kusakabe
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Woan-Yu Lin
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Alexander Grier
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Xiangyu Pan
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Olga Morzhanaeva
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Terrance P Shea
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hiroshi Yano
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Allen Discovery Center for Neuroimmune Interactions, New York, NY, USA
| | - Danielle Karo-Atar
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, McGill University, Montreal, Quebec, Canada
| | - Kaitlin A Olsen
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, McGill University, Montreal, Quebec, Canada
| | - Ji Hoon Oh
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Kurt J Vandegrift
- Department of Biology, The Pennsylvania State University, University Park, PA, USA
| | - Irah L King
- Department of Microbiology and Immunology, Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, McGill University, Montreal, Quebec, Canada
| | - Christina A Cuomo
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - David Artis
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Allen Discovery Center for Neuroimmune Interactions, New York, NY, USA
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Neil Lipman
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | - Iliyan D Iliev
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
15
|
Lei XY, He KY, Li QT, Zhang L, Wu DH, Yang JY, Guo JR, Liu MJ, Zhao ZL, Li JQ, Liu H, Zhao Y, Li YJ, Sun QH, Wu CG, Wang YF, Cao GS, Wang G, Jian YP, Xu ZX. PARylation of HMGA1 desensitizes esophageal squamous cell carcinoma to olaparib. Clin Transl Med 2024; 14:e70111. [PMID: 39690136 DOI: 10.1002/ctm2.70111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 11/01/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
As a chromatin remodelling factor, high mobility group A1 (HMGA1) plays various roles in both physiological and pathological conditions. However, its role in DNA damage response and DNA damage-based chemotherapy remains largely unexplored. In this study, we report the poly ADP-ribosylation (PARylation) of HMGA1 during DNA damage, leading to desensitization of esophageal squamous cell carcinoma (ESCC) cells to the poly(ADP-ribose) polymerase 1 (PARP1) inhibitor, olaparib. We found that HMGA1 accumulates at sites of DNA damage, where it interacts with PARP1 and undergoes PARylation at residues E47 and E50 in its conserved AT-hook domain. This modification enhances the accumulation of Ku70/Ku80 at the site of DNA damage and activates the DNA-dependent protein kinase catalytic subunit, facilitating nonhomologous end-joining repair. In both subcutaneous tumour models and genetically engineered mouse models of in situ esophageal cancer, HMGA1 interference increased tumour sensitivity to olaparib. Moreover, HMGA1 was highly expressed in ESCC tissues and positively correlated with PARP1 levels as well as poor prognosis in ESCC patients. Taken together, these findings reveal a mechanistic link between HMGA1 and PARP1 in regulating cell responses to DNA damage and suggest that targeting HMGA1 could be a promising strategy to increase cancer cell sensitivity to olaparib.
Collapse
Affiliation(s)
- Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, China
| | - Qiu-Tong Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, China
| | - Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Zi-Long Zhao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Huai Liu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yuan Zhao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yu-Jia Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Qian-Hui Sun
- School of Life Sciences, Henan University, Kaifeng, China
| | - Chen-Guang Wu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yun-Fan Wang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Geng-Sheng Cao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Gang Wang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, China
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, China
| |
Collapse
|
16
|
Fan L, Li M, Zhou X, Jia X, Tian H, Wen Q. T cell-related circRNA pairs to predict prognosis of patients with esophageal squamous cell carcinoma. Int Immunopharmacol 2024; 141:112909. [PMID: 39154531 DOI: 10.1016/j.intimp.2024.112909] [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: 05/08/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 08/20/2024]
Abstract
The prognosis for esophageal squamous cell carcinoma (ESCC), a prevalent and aggressive form of cancer, remains poor despite advancements in treatment options. Addressing the gap in comprehensive prognostic information derived from circRNA expression profiles for ESCC, our study aimed to establish a linkage between circRNA expressions and ESCC prognosis. To achieve this, we first developed an optimized prognostic model named T cell-related risk score (TRRS), which integrates T cell-associated features with machine learning algorithms. In parallel, we re-analyzed existing RNA-seq datasets to redefine the expression profiles of circRNAs and mRNAs. Utilizing the TRRS as a foundational "bridge," we identified circRNAs correlated with TRRS, leading to the development of a novel circRNA pair-based prognostic model, the TCRS, which is independent of specific expression levels. Further investigations uncovered two circRNAs, circNLK(5,6,7).1 and circRC3H1(2).1, with potential functional significance. These findings underscore the utility of these risk scores as tools for predicting overall survival and identifying potential therapeutic targets for ESCC patients.
Collapse
Affiliation(s)
- Liyuan Fan
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China.
| | - Mian Li
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xinyuan Zhou
- College of Artificial Intelligence and Big Data For Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiao Jia
- School of Control Science and Engineering, Shandong University, Jinan, China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Qiang Wen
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, China.
| |
Collapse
|
17
|
Zhang Y, Zhu Z, Ren N, Wang D, Zeng X, Zhang N. Implication of CCNG1 in radiosensitivity via the Wnt/β-catenin pathway in esophageal squamous cells. Sci Rep 2024; 14:27037. [PMID: 39511268 PMCID: PMC11544242 DOI: 10.1038/s41598-024-77811-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
Esophageal cancer (EC) poses a substantial threat to human health. The development of radioresistance in esophageal cancer cells is a critical factor contributing to local treatment failure and an unfavorable prognosis in affected patients. A comprehensive analysis was performed using bulk RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq) data from esophageal squamous cell carcinoma (ESCC) samples. Radioresistant ESCC cell lines were generated to explore the functional role of CCNG1. Various techniques, including gene knockdown, flow cytometry, and apoptosis assays, were utilized to evaluate alterations in radiosensitivity, cell cycle progression, and cell survival in response to CCNG1 modulation. Elevated CCNG1 expression was associated with poor clinical outcomes in ESCC patients and contributed to various malignant phenotypes in ESCC cells. In radioresistant ESCC cell lines, CCNG1 knockdown markedly increased radiosensitivity, as demonstrated by enhanced G2/M phase arrest and apoptosis following radiation exposure. CellChat analysis indicated a correlation between CCNG1 and the Wnt/β-catenin signaling pathway, while western blot (WB) analysis confirmed that CCNG1 functions as a downstream effector of Wnt/β-catenin. Our study has identified CCNG1 as a key regulator of radiosensitivity in ESCC, mediated through its interaction with the Wnt/β-catenin signaling pathway. Targeting the Wnt/β-catenin/CCNG1 axis presents a promising therapeutic strategy to enhance the efficacy of radiotherapy in ESCC, potentially overcoming radioresistance and improving patient outcomes.
Collapse
Affiliation(s)
- Yuxiang Zhang
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
- Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhiman Zhu
- Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Naihan Ren
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
| | - Dongni Wang
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
| | - Xue Zeng
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
| | - Na Zhang
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China.
| |
Collapse
|
18
|
Reyes Hueros RA, Gier RA, Shaffer SM. Non-genetic differences underlie variability in proliferation among esophageal epithelial clones. PLoS Comput Biol 2024; 20:e1012360. [PMID: 39466790 PMCID: PMC11573201 DOI: 10.1371/journal.pcbi.1012360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/18/2024] [Accepted: 07/24/2024] [Indexed: 10/30/2024] Open
Abstract
Individual cells grown in culture exhibit remarkable differences in their growth, with some cells capable of forming large clusters, while others are limited or fail to grow at all. While these differences have been observed across cell lines and human samples, the growth dynamics and associated cell states remain poorly understood. In this study, we performed clonal tracing through imaging and cellular barcoding of an in vitro model of esophageal epithelial cells (EPC2-hTERT). We found that about 10% of clones grow exponentially, while the remaining have cells that become non-proliferative leading to a halt in the growth rate. Using mathematical models, we demonstrate two distinct growth behaviors: exponential and logistic. Further, we discovered that the propensity to grow exponentially is largely heritable through four doublings and that the less proliferative clones can become highly proliferative through increasing plating density. Combining barcoding with single-cell RNA-sequencing (scRNA-seq), we identified the cellular states associated with the highly proliferative clones, which include genes in the WNT and PI3K pathways. Finally, we identified an enrichment of cells resembling the highly proliferative cell state in the proliferating healthy human esophageal epithelium.
Collapse
Affiliation(s)
- Raúl A. Reyes Hueros
- Department of Biochemistry and Molecular Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rodrigo A. Gier
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sydney M. Shaffer
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
19
|
Wong CN, Zhang Y, Ru B, Wang S, Zhou H, Lin J, Lyu Y, Qin Y, Jiang P, Lee VH, Guan X. Identification and Characterization of Metastasis-Initiating Cells in ESCC in a Multi-Timepoint Pulmonary Metastasis Mouse Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401590. [PMID: 38864342 PMCID: PMC11321633 DOI: 10.1002/advs.202401590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/23/2024] [Indexed: 06/13/2024]
Abstract
Metastasis is the biggest obstacle to esophageal squamous cell carcinoma (ESCC) treatment. Single-cell RNA sequencing analyses are applied to investigate lung metastatic ESCC cells isolated from pulmonary metastasis mouse model at multiple timepoints to characterize early metastatic microenvironment. A small population of parental KYSE30 cell line (Cluster S) resembling metastasis-initiating cells (MICs) is identified because they survive and colonize at lung metastatic sites. Differential expression profile comparisons between Cluster S and other subpopulations identified a panel of 7 metastasis-initiating signature genes (MIS), including CD44 and TACSTD2, to represent MICs in ESCC. Functional studies demonstrated MICs (CD44high) exhibited significantly enhanced cell survival (resistances to oxidative stress and apoptosis), migration, invasion, stemness, and in vivo lung metastasis capabilities, while bioinformatics analyses revealed enhanced organ development, stress responses, and neuron development, potentially remodel early metastasis microenvironment. Meanwhile, early metastasizing cells demonstrate quasi-epithelial-mesenchymal phenotype to support both invasion and anchorage. Multiplex immunohistochemistry (mIHC) staining of 4 MISs (CD44, S100A14, RHOD, and TACSTD2) in ESCC clinical samples demonstrated differential MIS expression scores (dMISs) predict lymph node metastasis, overall survival, and risk of carcinothrombosis.
Collapse
Affiliation(s)
- Ching Ngar Wong
- Department of Clinical OncologyCentre for Cancer MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongSAR999077China
| | - Yu Zhang
- Department of Pediatric OncologySun Yat‐sen University Cancer CenterGuangzhou510060China
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Beibei Ru
- Cancer Data Science LabCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20814USA
| | - Songna Wang
- Department of Clinical OncologyCentre for Cancer MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongSAR999077China
| | - Hongyu Zhou
- Department of Clinical OncologyCentre for Cancer MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongSAR999077China
| | - Jiarun Lin
- Department of Clinical OncologyCentre for Cancer MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongSAR999077China
| | - Yingchen Lyu
- Department of Clinical OncologyCentre for Cancer MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongSAR999077China
| | - Yanru Qin
- Department of Clinical Oncologythe First Affiliated HospitalZhengzhou UniversityZhengzhou450052China
| | - Peng Jiang
- Cancer Data Science LabCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMD20814USA
| | - Victor Ho‐Fun Lee
- Department of Clinical OncologyCentre for Cancer MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongSAR999077China
| | - Xin‐Yuan Guan
- Department of Clinical OncologyCentre for Cancer MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongSAR999077China
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterGuangzhou510060China
- Department of Clinical OncologyThe University of Hong Kong ‐ Shenzhen HospitalShenzhen518053China
- Shenzhen Key Laboratory for cancer metastasis and personalized therapyThe University of Hong Kong‐Shenzhen HospitalShenzhen518053China
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhou516029China
- MOE Key Laboratory of Tumor Molecular BiologyJinan UniversityGuangzhou510610China
| |
Collapse
|
20
|
Liu MJ, Zhao Y, Li QT, Lei XY, He KY, Guo JR, Yang JY, Yan ZH, Wu DH, Zhang L, Jian YP, Xu ZX. HMGA1 promotes the progression of esophageal squamous cell carcinoma by elevating TKT-mediated upregulation of pentose phosphate pathway. Cell Death Dis 2024; 15:541. [PMID: 39080260 PMCID: PMC11289123 DOI: 10.1038/s41419-024-06933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) possesses a poor prognosis and treatment outcome. Dysregulated metabolism contributes to unrestricted growth of multiple cancers. However, abnormal metabolism, such as highly activated pentose phosphate pathway (PPP) in the progression of ESCC remains largely unknown. Herein, we report that high-mobility group AT-hook 1 (HMGA1), a structural transcriptional factor involved in chromatin remodeling, promoted the development of ESCC by upregulating the PPP. We found that HMGA1 was highly expressed in ESCC. Elevated HMGA1 promoted the malignant phenotype of ESCC cells. Conditional knockout of HMGA1 markedly reduced 4-nitroquinoline-1-oxide (4NQO)-induced esophageal tumorigenesis in mice. Through the metabolomic analysis and the validation assay, we found that HMGA1 upregulated the non-oxidative PPP. With the transcriptome sequencing, we identified that HMGA1 upregulated the expression of transketolase (TKT), which catalyzes the reversible reaction in non-oxidative PPP to exchange metabolites with glycolytic pathway. HMGA1 knockdown suppressed the PPP by downregulating TKT, resulting in the reduction of nucleotides in ESCC cells. Overexpression of HMGA1 upregulated PPP and promoted the survival of ESCC cells by activating TKT. We further characterized that HMGA1 promoted the transcription of TKT by interacting with and enhancing the binding of transcription factor SP1 to the promoter of TKT. Therapeutics targeting TKT with an inhibitor, oxythiamine, reduced HMGA1-induced ESCC cell proliferation and tumor growth. Together, in this study, we identified a new role of HMGA1 in ESCCs by upregulating TKT-mediated activation of PPP. Our results provided a new insight into the role of HMGA1/TKT/PPP in ESCC tumorigenesis and targeted therapy.
Collapse
Affiliation(s)
- Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yuan Zhao
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Qiu-Tong Li
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Zhen-Hua Yan
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| |
Collapse
|
21
|
Zhang D, Zhao F, Li J, Guo P, Liu H, Lu T, Li S, Li Z, Li Y. Comprehensive single-cell transcriptomic profiling reveals molecular subtypes and prognostic biomarkers with implications for targeted therapy in esophageal squamous cell carcinoma. Transl Oncol 2024; 44:101948. [PMID: 38582059 PMCID: PMC11004200 DOI: 10.1016/j.tranon.2024.101948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 03/26/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a genetically heterogeneous disease with poor clinical outcomes. Identification of biomarkers linked to DNA replication stress may enable improved prognostic risk stratification and guide therapeutic decision making. We performed integrated single-cell RNA sequencing and computational analyses to define the molecular determinants and subtypes underlying ESCC heterogeneity. METHODS Single-cell RNA sequencing was performed on ESCC samples and analyzed using Seurat. Differential gene expression analysis was used to identify esophageal cell phenotypes. DNA replication stress-related genes were intersected with single-cell differential expression data to identify potential prognostic genes, which were used to generate a DNA replication stress (DRS) score. This score and associated genes were evaluated in survival analysis. Putative prognostic biomarkers were evaluated by Cox regression and consensus clustering. Mendelian randomization analyses assessed the causal role of PRKCB. RESULTS High DRS score associated with poor survival. Four genes (CDKN2A, NUP155, PPP2R2A, PRKCB) displayed prognostic utility. Three molecular subtypes were identified with discrete survival and immune properties. A 12-gene signature displayed robust prognostic performance. PRKCB was overexpressed in ESCC, while PRKCB knockdown reduced ESCC cell migration. CONCLUSIONS This integrated single-cell sequencing analysis provides new insights into the molecular heterogeneity and prognostic determinants underlying ESCC. The findings identify potential prognostic biomarkers and a gene expression signature that may enable improved patient risk stratification in ESCC. Experimental validation of the role of PRKCB substantiates the potential clinical utility of our results.
Collapse
Affiliation(s)
- Dengfeng Zhang
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Fangchao Zhao
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Jing Li
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Pengfei Guo
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Haitao Liu
- College of Life Science, Inner Mongolia University, Hohhot 010000, China
| | - Tianxing Lu
- 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.
| | - Zhirong Li
- Provincial Center for Clinical Laboratories, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| | - Yishuai Li
- Department of Thoracic Surgery, Hebei Chest Hospital, Shijiazhuang 050000, China; Hebei Provincial Key Laboratory of Pulmonary Diseases, Shijiazhuang 050000, China.
| |
Collapse
|
22
|
Huang H, Jiang Y, Liu J, Luo D, Yuan J, Mu R, Yu X, Sun D, Lin J, Chen Q, Li X, Jiang M, Xu J, Chu B, Yin C, Zhang L, Ye Y, Cao B, Wang Q, Zhang Y. Jag1/2 maintain esophageal homeostasis and suppress foregut tumorigenesis by restricting the basal progenitor cell pool. Nat Commun 2024; 15:4124. [PMID: 38750026 PMCID: PMC11096375 DOI: 10.1038/s41467-024-48347-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 04/28/2024] [Indexed: 05/18/2024] Open
Abstract
Basal progenitor cells are crucial for maintaining foregut (the esophagus and forestomach) homeostasis. When their function is dysregulated, it can promote inflammation and tumorigenesis. However, the mechanisms underlying these processes remain largely unclear. Here, we employ genetic mouse models to reveal that Jag1/2 regulate esophageal homeostasis and foregut tumorigenesis by modulating the function of basal progenitor cells. Deletion of Jag1/2 in mice disrupts esophageal and forestomach epithelial homeostasis. Mechanistically, Jag1/2 deficiency impairs activation of Notch signaling, leading to reduced squamous epithelial differentiation and expansion of basal progenitor cells. Moreover, Jag1/2 deficiency exacerbates the deoxycholic acid (DCA)-induced squamous epithelial injury and accelerates the initiation of squamous cell carcinoma (SCC) in the forestomach. Importantly, expression levels of JAG1/2 are lower in the early stages of human esophageal squamous cell carcinoma (ESCC) carcinogenesis. Collectively, our study demonstrates that Jag1/2 are important for maintaining esophageal and forestomach homeostasis and the onset of foregut SCC.
Collapse
Affiliation(s)
- Haidi Huang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Yu Jiang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Jiangying Liu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Dan Luo
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Jianghong Yuan
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Rongzi Mu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Xiang Yu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Donglei Sun
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Jihong Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, PR China
| | - Qiyue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, PR China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, PR China
| | - Xinjing Li
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Ming Jiang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310030, Zhejiang, PR China
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Bo Chu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, PR China
| | - Chengqian Yin
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, Guangdong, PR China
| | - Lei Zhang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, Guangdong, PR China
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, Guangdong, PR China
| | - Youqiong Ye
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Bo Cao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Qiong Wang
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China.
| | - Yongchun Zhang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| |
Collapse
|
23
|
Zhang P, Wen B, Gong J, Liu Z, Zhang M, Zhou G, Zhang L, Zhang Z. Clinical prognostication and immunotherapy response prediction in esophageal squamous cell carcinoma using the DNA damage repair-associated signature. ENVIRONMENTAL TOXICOLOGY 2024; 39:2803-2816. [PMID: 38287713 DOI: 10.1002/tox.24155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/06/2024] [Accepted: 01/18/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND The relationship between DNA damage repair (DDR) and cancer is intricately intertwined; however, its specific role in esophageal squamous cell carcinoma (ESCC) remains enigmatic. METHODS Employing single-cell analysis, we delineated the functionality of DDR-related genes within the tumor microenvironment (TME). A diverse array of scoring mechanisms, including AUCell, UCell, singscore, ssgsea, and AddModuleScore, were harnessed to scrutinize the activity of DDR-related genes across different cell types. Differential pathway alterations between high-and low-DDR activity cell clusters were compared. Furthermore, leveraging multiple RNA-seq datasets, we constructed a robust DDR-associated signature (DAS), and through integrative multiomics analysis, we explored differences in prognosis, pathways, mutational landscapes, and immunotherapy predictions among distinct DAS groups. RESULTS Notably, high-DDR activity cell subpopulations exhibited markedly enhanced cellular communication. The DAS demonstrated uniformity across multiple datasets. The low-DAS group exhibited improved prognoses, accompanied by heightened immune infiltration and elevated immune checkpoint expression. SubMap analysis of multiple immunotherapy datasets suggested that low-DAS group may experience enhanced immunotherapy responses. The "oncopredict" R package analyzed and screened sensitive drugs for different DAS groups. CONCLUSION Through the integration of single-cell and bulk RNA-seq data, we have developed a DAS associated with prognosis and immunotherapy response. This signature holds promise for the future stratification and personalized treatment of ESCC patients in clinical settings.
Collapse
Affiliation(s)
- Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Bing Wen
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Cardiothoracic Surgery, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Jialin Gong
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zuo Liu
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Mengzhe Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Guangyao Zhou
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lianmin Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhenfa Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
24
|
Liang J, Lei K, Liang R, Huang J, Tan B, Lin H, Wang M. Single-cell RNA sequencing reveals the MIF-ACKR3 receptor-ligand interaction between iCAFs and tumor cells in esophageal squamous cell carcinoma. Cell Signal 2024; 117:111093. [PMID: 38336189 DOI: 10.1016/j.cellsig.2024.111093] [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: 12/01/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a malignant tumor of the gastrointestinal tract with a high morbidity and mortality rate. The heterogeneity of ESCC poses challenges in treatment and contributes to the poor prognosis of patients. Therefore, it is crucial to gain a better understanding of the tumor microenvironment (TME) heterogeneity and identify novel therapeutic targets. METHODS To solve this problem, we performed a single-cell RNA sequencing (scRNA-seq) analysis of ESCC samples obtained from the GEO database. RESULTS A total of 31,283 single cells were categorized into nine cell types, which included four non-immune cells (epithelial cells, endothelial cells, fibroblasts, schwann cells) and five immune cells (T cells, macrophages, mast cells, neutrophils, B cells). Our study revealed the presence of immunosuppressive tumor microenvironments in ESCC. We have also identified not only inflammatory cancer-associated fibroblast (iCAFs) and myofibroblastic cancer-associated fibroblasts (myCAFs) but also a subset of antigen presenting cancer-associated fibroblasts (apCAFs) which express high levels of HLA class II molecules in ESCC. Furthermore, our analysis of cell communication showed up-regulation of MIF-ACKR3 interaction between iCAFs and tumor cells in tumors compared to normal tissues. Finally, it was demonstrated that macrophage migration inhibitory factor (MIF) facilitates tumor cell migration and invasion through interacting with ACKR3 in vitro. CONCLUSIONS This study exposes the features of the tumor microenvironment of ESCC via scRNA-seq and examines the dynamics of various cellular subpopulations, thus facilitating the identification of future therapeutic targets for ESCC.
Collapse
Affiliation(s)
- Jialu Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Lei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruihao Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Binhua Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huayue Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Minghui Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
25
|
Zhang Z, Wang J, Shi Y, Wang B, Wang D. Cathepsin L promotes oesophageal squamous cell carcinoma development and may be associated with tumour-associated macrophages. Heliyon 2024; 10:e29273. [PMID: 38601581 PMCID: PMC11004422 DOI: 10.1016/j.heliyon.2024.e29273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Abstract
Background Oesophageal squamous cell carcinoma (ESCC) is a leading cause of cancer-related deaths worldwide because existing treatments are often insufficient. Therefore, new, reliable biomarkers must be identified. CTSL overexpression is closely associated with tumour progression and poor prognosis. However, the role and mechanism of CTSL as an oncogene in ESCC remain unclear. Methods Genome-wide association study (GWAS) data were used for Mendelian randomization analysis to investigate the possible relationships between CTSL and ESCC. The correlation between CTSL expression and prognosis was analysed using GEO, TCGA, and GEPIA data. We compared CTSL expression among the cell types using single-cell sequencing. Correlations between CTSL and the tumour microenvironment, immune cell infiltration, tumour mutational load, immunological checkpoints, and treatment sensitivity in patients with ESCC were investigated. Finally, using mouse models and cellular investigations, we assessed the effects of CTSL on the growth, apoptosis, and metastasis of ESCC tumour cells. Results CTSL was overexpressed in ESCC and correlated with prognosis. We also discovered its close association with cell immunity, especially with tumour-associated macrophages and immune checkpoints in the tumour microenvironment. CTSL may play a key role in ESCC development by affecting M2 macrophage polarisation. CTSL and the M2 macrophage marker genes showed significant positive correlations. Mendelian randomization analysis confirmed a relationship between CTSL and ESCC. Finally, our in vitro and in vivo experiments demonstrated that CTSL promoted the proliferation and migration of ESCC cells, validating our bioinformatic analysis. Conclusion CTSL emerged as a crucial gene in ESCC that influences patient prognosis and immunity, particularly in association with M2 macrophages. Therefore, targeting or modulating CTSL levels may provide new therapeutic strategies for patients with ESCC.
Collapse
Affiliation(s)
- Zhenhu Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Jianyu Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yamin Shi
- School of Foreign Languages, Shandong University of Finance and Economics, Jinan, 250014, China
| | - Ben Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Dong Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| |
Collapse
|
26
|
Tan Y, Song L, Ma J, Pan M, Niu S, Yue X, Li Y, Gu L, Liu S, Chang J. Single-cell analysis identified POSTN + cells associated with the aggressive phenotype and risk of esophageal squamous cell carcinoma. HGG ADVANCES 2024; 5:100278. [PMID: 38369754 PMCID: PMC10924139 DOI: 10.1016/j.xhgg.2024.100278] [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: 12/16/2023] [Revised: 02/14/2024] [Accepted: 02/14/2024] [Indexed: 02/20/2024] Open
Abstract
Tumors are intricate and heterogeneous systems characterized by mosaic cancer cell populations with diverse expression profiles. Leveraging single-cell technologies, we employed the Scissor algorithm to delineate an epithelial subpopulation associated with the aggressive phenotype in esophageal squamous cell carcinoma (ESCC). This identified subpopulation exhibited elevated expression of genes involved in critical pathways, such as epithelial-mesenchymal transition and PI3K-Akt. Key signature genes within this subpopulation, namely CAV1, COL3A1, COL6A1, POSTN, and TAGLN, demonstrated significant upregulation concomitant with both tumorigenesis and tumor progression across independent single-cell datasets. Furthermore, we selected 1,450 expression quantitative trait loci of the top 62 signature genes of this cell subpopulation to investigate their potential in predicting ESCC risk. The results showed that the POSTN loci were predominantly associated with ESCC susceptibility. Through functional annotation and replication analyses, we identified that the rs1028728 in the POSTN promoter was significantly associated with increased ESCC risk in 7,049 ESCC cases and 8,063 controls (odds ratio = 1.29, 95% confidence interval: 1.18-1.42, p = 4.03 × 10-8). Subsequent biochemical experiments showed that the rs1028728[T] allele enhanced POSTN expression by affecting the binding of PRRX1 in the POSTN promoter. In summary, our meticulous single-cell analysis delineates an invasive epithelial subpopulation in ESCC, with POSTN emerging as an important marker for the aggressive phenotype. These findings offer more insights into potential strategies for the prevention and intervention of ESCC, enriching our understanding of this complex cancer landscape.
Collapse
Affiliation(s)
- Yuqian Tan
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lina Song
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jialing Ma
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Miaoxin Pan
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Siyuan Niu
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinying Yue
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yueping Li
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Linglong Gu
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shasha Liu
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiang Chang
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
27
|
Chen Q, Mo S, Zhu L, Tang M, Cheng J, Ye P, Zheng W, Hu J. Prognostic implication of UBE2C + CD8 + T cell in neoadjuvant immune checkpoint blockade plus chemotherapy for locally advanced esophageal cancer. Int Immunopharmacol 2024; 130:111696. [PMID: 38412672 DOI: 10.1016/j.intimp.2024.111696] [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: 12/25/2023] [Revised: 02/04/2024] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Immune checkpoint blockers (ICBs) plus chemotherapy as neoadjuvant therapy for patients with esophageal cancer (EC) has gained substantial attention. This study aimed to investigate the early and mid-term outcome of neoadjuvant ICBs plus chemotherapy and discover immune-associated predictors of major pathological response (MPR) for locally advanced EC. METHOD Patients with locally advanced EC who received neoadjuvant ICBs plus chemotherapy were retrospectively included between June 2019 to December 2021. Conjoint analysis of Bulk-RNA seq (GSE165252) and scRNA seq (GSE188900) were used to investigate potential prognostic factors and immunological mechanisms, then multiplexed immunofluorescence was applied to validate. RESULTS 76 patients were included. A total of 21 (27.6 %) patients achieved MPR, with 13 (17.1 %) attaining a pathological complete response. Over a median follow-up of 1.8 years, 6 (7.9 %) patients died and 21 (27.6 %) experienced disease recurrence within 0.6 to 2.1 years after surgery. The overall survival rate and recurrence-free survival rate were 93.3 + 2.9 % and 84.8 + 4.2 % at 12 months, 90.8 + 3.7 % and 67.1 + 6.4 % at 24 months, and 90.8 + 3.7 % and 62.9 + 7.2 % at 36 months, respectively. Patients achieving MPR had a significantly lower risk of recurrence compared to non-responders (9.5 % vs 34.5 %, P = 0.017). Analysis of bulk-RNA seq and scRNA-seq revealed that UBE2C and UBE2C + CD8 + T cells were adverse prognostic factors. Immunohistochemistry demonstrated that the non-MPR group had a higher infiltration of UBE2C + immune cells than MPR group after neoadjuvant treatment. Multiplexed immunofluorescence confirmed that infiltrating UBE2C + CD8 + T cells in MPR group were significantly fewer than non-MPR group after neoadjuvant treatment, indicating their poor prognostic role for EC. CONCLUSIONS Neoadjuvant ICBs plus chemotherapy shows promising efficacy in locally advanced EC, with MPR being a significant predictor of lower recurrence risk. Immunological analyses identified UBE2C + CD8 + T cells as adverse prognostic factors, suggesting their potential as biomarkers for patient stratification and treatment response.
Collapse
Affiliation(s)
- Qiuming Chen
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Shaocong Mo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Linhai Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Muhu Tang
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jun Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peng Ye
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wanwei Zheng
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
28
|
Yang JY, Lei XY, He KY, Guo JR, Liu MJ, Li JQ, Li QT, Jiang ZH, Zhang L, Wu DH, Li YJ, Sun QH, Jian YP, Xu ZX. HMGA1 drives chemoresistance in esophageal squamous cell carcinoma by suppressing ferroptosis. Cell Death Dis 2024; 15:158. [PMID: 38383528 PMCID: PMC10881472 DOI: 10.1038/s41419-024-06467-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 02/23/2024]
Abstract
Chemotherapy is a primary treatment for esophageal squamous cell carcinoma (ESCC). Resistance to chemotherapeutic drugs is an important hurdle to effective treatment. Understanding the mechanisms underlying chemotherapy resistance in ESCC is an unmet medical need to improve the survival of ESCC. Herein, we demonstrate that ferroptosis triggered by inhibiting high mobility group AT-hook 1 (HMGA1) may provide a novel opportunity to gain an effective therapeutic strategy against chemoresistance in ESCC. HMGA1 is upregulated in ESCC and works as a key driver for cisplatin (DDP) resistance in ESCC by repressing ferroptosis. Inhibition of HMGA1 enhances the sensitivity of ESCC to ferroptosis. With a transcriptome analysis and following-up assays, we demonstrated that HMGA1 upregulates the expression of solute carrier family 7 member 11 (SLC7A11), a key transporter maintaining intracellular glutathione homeostasis and inhibiting the accumulation of malondialdehyde (MDA), thereby suppressing cell ferroptosis. HMGA1 acts as a chromatin remodeling factor promoting the binding of activating transcription factor 4 (ATF4) to the promoter of SLC7A11, and hence enhancing the transcription of SLC7A11 and maintaining the redox balance. We characterized that the enhanced chemosensitivity of ESCC is primarily attributed to the increased susceptibility of ferroptosis resulting from the depletion of HMGA1. Moreover, we utilized syngeneic allograft tumor models and genetically engineered mice of HMGA1 to induce ESCC and validated that depletion of HMGA1 promotes ferroptosis and restores the sensitivity of ESCC to DDP, and hence enhances the therapeutic efficacy. Our finding uncovers a critical role of HMGA1 in the repression of ferroptosis and thus in the establishment of DDP resistance in ESCC, highlighting HMGA1-based rewiring strategies as potential approaches to overcome ESCC chemotherapy resistance. Schematic depicting that HMGA1 maintains intracellular redox homeostasis against ferroptosis by assisting ATF4 to activate SLC7A11 transcription, resulting in ESCC resistance to chemotherapy.
Collapse
Affiliation(s)
- Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Qiu-Tong Li
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Zhi-Hao Jiang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yu-Jia Li
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Qian-Hui Sun
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| |
Collapse
|
29
|
Li C, Song W, Zhang J, Luo Y. Single-cell transcriptomics reveals heterogeneity in esophageal squamous epithelial cells and constructs models for predicting patient prognosis and immunotherapy. Front Immunol 2023; 14:1322147. [PMID: 38098487 PMCID: PMC10719955 DOI: 10.3389/fimmu.2023.1322147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC), characterized by its high invasiveness and malignant potential, has long been a formidable challenge in terms of treatment. Methods A variety of advanced analytical techniques are employed, including single-cell RNA sequencing (scRNA-seq), cell trajectory inference, transcription factor regulatory network analysis, GSVA enrichment analysis, mutation profile construction, and the inference of potential immunotherapeutic drugs. The purpose is to conduct a more comprehensive exploration of the heterogeneity among malignant squamous epithelial cell subgroups within the ESCC microenvironment and establish a model for predicting the prognosis and immunotherapy outcomes of ESCC patients. Results An analysis was conducted through scRNA-seq, and three Cluster of malignant epithelial cells were identified using the infer CNV method. Cluster 0 was found to exhibit high invasiveness, whereas Cluster 1 displayed prominent characteristics associated with epithelial-mesenchymal transition. Confirmation of these findings was provided through cell trajectory analysis, which positioned Cluster 0 at the initiation stage of development and Cluster 1 at the final developmental stage. The abundance of Cluster 0-2 groups in TCGA-LUAD samples was assessed using ssGSEA and subsequently categorized into high and low-expression groups. Notably, it was observed that Cluster 0-1 had a significant impact on survival (p<0.05). Furthermore, GSVA enrichment analysis demonstrated heightened activity in hallmark pathways for Cluster 0, whereas Cluster 1 exhibited notable enrichment in pathways related to cell proliferation. It is noteworthy that a prognostic model was established utilizing feature genes from Cluster 0-1, employing the Lasso and stepwise regression methods. The results revealed that in TCGA and GSE53624 cohorts, the low-risk group demonstrated significantly higher overall survival and increased levels of immune infiltration. An examination of four external immunotherapy cohorts unveiled that the low-risk group exhibited improved immunotherapeutic efficacy. Additionally, more meaningful treatment options were identified for the low-risk group. Conclusion The findings revealed distinct interactions between malignant epithelial cells of ESCC and subgroups within the tumor microenvironment. Two cell clusters, strongly linked to survival, were pinpointed, and a signature was formulated. This signature is expected to play a crucial role in identifying and advancing precision medicine approaches for the treatment of ESCC.
Collapse
Affiliation(s)
- Chenglin Li
- Department of Cardiothoracic Surgery, The Affiliated Huaian No.1 People’s Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Wei Song
- Department of Gastroenterology, The Affiliated Huaian No.1 People’s Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Jialing Zhang
- Department of Gastroenterology, The Affiliated Huaian No.1 People’s Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Yonggang Luo
- Department of Cardiothoracic Surgery, The Affiliated Huaian No.1 People’s Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| |
Collapse
|
30
|
Zhang P, Dong S, Sun W, Zhong W, Xiong J, Gong X, Li J, Lin H, Zhuang Y. Deciphering Treg cell roles in esophageal squamous cell carcinoma: a comprehensive prognostic and immunotherapeutic analysis. Front Mol Biosci 2023; 10:1277530. [PMID: 37842637 PMCID: PMC10568469 DOI: 10.3389/fmolb.2023.1277530] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Background: Esophageal squamous cell carcinoma (ESCC) is a prevalent and aggressive form of cancer that poses significant challenges in terms of prognosis and treatment. Regulatory T cells (Treg cells) have gained attention due to their influential role in immune modulation within the tumor microenvironment (TME). Understanding the intricate interactions between Treg cells and the tumor microenvironment is essential for unraveling the mechanisms underlying ESCC progression and for developing effective prognostic models and immunotherapeutic strategies. Methods: A combination of single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq analysis was utilized to explore the role of Treg cells within the TME of ESCC. The accuracy and applicability of the prognostic model were assessed through multi-dimensional evaluations, encompassing an examination of the model's performance across various dimensions, such as the mutation landscape, clinical relevance, enrichment analysis, and potential implications for immunotherapy strategies. Results: The pivotal role of the macrophage migration inhibitory factor (MIF) signaling pathway within the ESCC TME was investigated, with a focus on its impact on Treg cells and other subpopulations. Through comprehensive integration of bulk sequencing data, a Treg-associated signature (TAS) was constructed, revealing that ESCC patients with elevated TAS (referred to as high-TAS individuals) experienced significantly improved prognoses. Heightened immune infiltration and increased expression of immune checkpoint markers were observed in high-TAS specimens. The model's validity was established through the IMvigor210 dataset, demonstrating its robustness in predicting prognosis and responsiveness to immunotherapy. Heightened therapeutic benefits were observed in immune-based interventions for high-TAS ESCC patients. Noteworthy differences in pathway enrichment patterns emerged between high and low-TAS cohorts, highlighting potential avenues for therapeutic exploration. Furthermore, the clinical relevance of key model genes was substantiated by analyzing clinical samples from ten paired tumor and adjacent tissues, revealing differential expression levels. Conclusion: The study established a TAS that enables accurate prediction of patient prognosis and responsiveness to immunotherapy. This achievement holds significant implications for the clinical management of ESCC, offering valuable insights for informed therapeutic interventions.
Collapse
Affiliation(s)
- Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shiyang Dong
- Department of General Surgery, Fuyang Tumour Hospital, Fuyang, China
| | - Wei Sun
- Department of Thoracic Surgery, The Second Hospital of Nanjing, Nanjing, China
| | - Wan Zhong
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingwen Xiong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Xiangjin Gong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Jun Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haoran Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Zhuang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing, China
- Afliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Zhong J, Xiao C, Chen Q, Pan X, Xu T, Wang Y, Hou W, Liu L, Cao F, Wang Y, Li X, Zhou L, Yang H, Yang Y, Zhao C. Zebrafish functional xenograft vasculature platform identifies PF-502 as a durable vasculature normalization drug. iScience 2023; 26:107734. [PMID: 37680473 PMCID: PMC10480778 DOI: 10.1016/j.isci.2023.107734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/21/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Tumor vasculature often exhibits disorder and inefficiency. Vascular normalization offers potential for alleviating hypoxia and optimizing drug delivery in tumors. However, identifying effective agents is hindered by a lack of robust screening. We aimed to establish a comprehensive method using the zebrafish functional xenograft vasculature platform (zFXVP) to visualize and quantify tumor vasculature changes. Employing zFXVP, we systematically screened compounds, identifying PF-502 as a robust vascular normalization agent. Mechanistic studies showed PF-502 induces endothelial cell-cycle arrest, streamlines vasculature, and activates Notch1 signaling, enhancing stability and hemodynamics. In murine models, PF-502 exhibited pronounced vascular normalization and improved drug delivery at a sub-maximum tolerated dose. These findings highlight zFXVP's utility and suggest PF-502 as a promising adjunctive for vascular normalization in clinical settings.
Collapse
Affiliation(s)
- Jian Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Chaoxin Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Qin Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Xiangyu Pan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Tongtong Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yiyun Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Wanting Hou
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Lu Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Fujun Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yulin Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Xiaoying Li
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Lin Zhou
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yu Yang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| |
Collapse
|
32
|
Meng Y, Huang K, Shi M, Huo Y, Han L, Liu B, Li Y. Research Advances in the Role of the Tropomyosin Family in Cancer. Int J Mol Sci 2023; 24:13295. [PMID: 37686101 PMCID: PMC10488083 DOI: 10.3390/ijms241713295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Cancer is one of the most difficult diseases for human beings to overcome. Its development is closely related to a variety of factors, and its specific mechanisms have been a hot research topic in the field of scientific research. The tropomyosin family (Tpm) is a group of proteins closely related to the cytoskeleton and actin, and recent studies have shown that they play an important role in various cancers, participating in a variety of biological activities, including cell proliferation, invasion, and migration, and have been used as biomarkers for various cancers. The purpose of this review is to explore the research progress of the Tpm family in tumorigenesis development, focusing on the molecular pathways associated with them and their relevant activities involved in tumors. PubMed and Web of Science databases were searched for relevant studies on the role of Tpms in tumorigenesis and development and the activities of Tpms involved in tumors. Data from the literature suggest that the Tpm family is involved in tumor cell proliferation and growth, tumor cell invasion and migration, tumor angiogenesis, tumor cell apoptosis, and immune infiltration of the tumor microenvironment, among other correlations. It can be used as a potential biomarker for early diagnosis, follow-up, and therapeutic response of some tumors. The Tpm family is involved in cancer in a close relationship with miRNAs and LncRNAs. Tpms are involved in tumor tissue invasion and migration as a key link. On this basis, TPM is frequently used as a biomarker for various cancers. However, the specific molecular mechanism of its involvement in cancer progression has not been explained clearly, which remains an important direction for future research.
Collapse
Affiliation(s)
- Yucheng Meng
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Ke Huang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China
| | - Mingxuan Shi
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Yifei Huo
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Liang Han
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Bin Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Yi Li
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| |
Collapse
|
33
|
Chen J, Dai S, Zhao L, Peng Y, Sun C, Peng H, Zhong Q, Quan Y, Li Y, Chen X, Pan X, Zhong A, Wang M, Zhang M, Yang S, Lu Y, Lian Z, Liu Y, Zhou S, Li Z, Na F, Chen C. A New Type of Endometrial Cancer Models in Mice Revealing the Functional Roles of Genetic Drivers and Exploring their Susceptibilities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300383. [PMID: 37340596 PMCID: PMC10460855 DOI: 10.1002/advs.202300383] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/12/2023] [Indexed: 06/22/2023]
Abstract
Endometrial cancer (EC) is the most common female reproductive tract cancer and its incidence has been continuously increasing in recent years. The underlying mechanisms of EC tumorigenesis remain unclear, and efficient target therapies are lacking, for both of which feasible endometrial cancer animal models are essential but currently limited. Here, an organoid and genome editing-based strategy to generate primary, orthotopic, and driver-defined ECs in mice is reported. These models faithfully recapitulate the molecular and pathohistological characteristics of human diseases. The authors names these models and similar models for other cancers as organoid-initiated precision cancer models (OPCMs). Importantly, this approach can conveniently introduce any driver mutation or a combination of driver mutations. Using these models,it is shown that the mutations in Pik3ca and Pik3r1 cooperate with Pten loss to promote endometrial adenocarcinoma in mice. In contrast, the Kras G12D mutati led to endometrial squamous cell carcinoma. Then, tumor organoids are derived from these mouse EC models and performed high-throughput drug screening and validation. The results reveal distinct vulnerabilities of ECs with different mutations. Taken together, this study develops a multiplexing approach to model EC in mice and demonstrates its value for understanding the pathology of and exploring the potential treatments for this malignancy.
Collapse
Affiliation(s)
- Jingyao Chen
- Precision Medicine Research CenterState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Siqi Dai
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Lei Zhao
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Yiman Peng
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Chongen Sun
- West China Second HospitalSichuan UniversityChengdu610041China
| | - Hongling Peng
- West China Second HospitalSichuan UniversityChengdu610041China
| | - Qian Zhong
- West China Second HospitalSichuan UniversityChengdu610041China
| | - Yuan Quan
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Yue Li
- Department of DermatologyState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Xuelan Chen
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Xiangyu Pan
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Ailing Zhong
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Manli Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Mengsha Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - You Lu
- Division of Thoracic Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengdu610041China
- Laboratory of Clinical Cell Therapy, West China HospitalSichuan UniversityChengdu610041China
| | - Zhong Lian
- Department of DermatologyState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Yu Liu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Shengtao Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
- West China Second HospitalSichuan UniversityChengdu610041China
| | - Zhengyu Li
- West China Second HospitalSichuan UniversityChengdu610041China
| | - Feifei Na
- Division of Thoracic Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengdu610041China
| | - Chong Chen
- Precision Medicine Research CenterState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
34
|
Xia Y, Sun T, Li G, Li M, Wang D, Su X, Ye J, Ji C. Spatial single cell analysis of tumor microenvironment remodeling pattern in primary central nervous system lymphoma. Leukemia 2023; 37:1499-1510. [PMID: 37120690 PMCID: PMC10317840 DOI: 10.1038/s41375-023-01908-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 05/01/2023]
Abstract
To determine the overall tumor microenvironment (TME), characteristics, and transition mechanisms in primary central nervous system lymphoma (PCNSL), we performed spatial transcriptomics and matched the corresponding single-cell sequencing data of PCNSL patients. We found that tumor cells may achieve a "TME remodeling pattern" through an "immune pressure-sensing model", in which they could choose to reshape the TME into a barrier environment or a cold environment according to the immune pressure. A key FKBP5+ tumor subgroup was found to be responsible for pushing tumors into the barrier environment, which provides a possible way to evaluate the stage of PCNSL. The specific mechanism of the TME remodeling pattern and the key molecules of the immune pressure-sensing model were identified through the spatial communication analysis. Finally, we discovered the spatial and temporal distributions and variation characteristics of immune checkpoint molecules and CAR-T target molecules in immunotherapy. These data clarified the TME remodeling pattern of PCNSL, provided a reference for its immunotherapy, and provided suggestions for the TME remodeling mechanism of other cancers.
Collapse
Affiliation(s)
- Yuan Xia
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Tao Sun
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Guosheng Li
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
- Shandong Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Mingying Li
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Dongmei Wang
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Xiuhua Su
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Jingjing Ye
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
- Shandong Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| | - Chunyan Ji
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
- Shandong Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| |
Collapse
|
35
|
Wang R, Cheng X, Chi D, Liu S, Li Q, Chen B, Xi M. M 1A and m 7G modification-related genes are potential biomarkers for survival prognosis and for deciphering the tumor immune microenvironment in esophageal squamous cell carcinoma. Discov Oncol 2023; 14:99. [PMID: 37314494 DOI: 10.1007/s12672-023-00710-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is the most common esophageal malignancy, and RNA methylation has been reported to be involved in the tumorigenesis of ESCC. However, no study has explored methylation modifications in m1A and m7G as prognostic markers for survival prediction in ESCC. METHODS Public gene-expression data and clinical annotation of 254 patients obtained from The Cancer Genome Atlas and the Gene Expression Omnibus databases were analyzed to identify potential consensus clusters of m1A and m7G modification-related genes. The RNA-seq of 20 patients in Sun Yat-Sen University Cancer Center was used as the validation set. Following screening for relevant differentially expressed genes (DEGs) and enrichment pathways were elucidated. DEGs were used to construct risk models using the randomForest algorithm, and the prognostic role of the models was assessed by applying Kaplan-Meier analysis. Extent of immune cell infiltration, drug resistance, and response to cancer treatment among different clusters and risk groups were also evaluated. RESULTS Consensus clustering analysis based on m1A and m7G modification patterns revealed three potential clusters. In total, 212 RNA methylation-related DEGs were identified. The methylation-associated signature consisting of 6 genes was then constructed to calculate methylation-related score (MRScore) and patients were dived into MRScore-high and MRScore-low groups. This signature has satisfied prognostic value for survival of ESCC (AUC = 0.66, 0.67, 0.64 for 2-, 3-, 4- year OS), and has satisfied performance in the validation SYSUCC cohort (AUC = 0.66 for 2- and 3-year OS). Significant correlation between m1A and m7G modification-related genes and immune cell infiltration, and drug resistance was also observed. CONCLUSIONS Transcriptomic prognostic signatures based on m1A and m7G modification-related genes are closely associated with immune cell infiltration in ESCC patients and have important correlations with the therapeutic sensitivity of multiple chemotherapeutic agents.
Collapse
Affiliation(s)
- Ruixi Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Xingyuan Cheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Dongmei Chi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Shiliang Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Qiaoqiao Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Baoqing Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China.
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou, 510060, China.
| | - Mian Xi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China.
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou, 510060, China.
| |
Collapse
|
36
|
Zheng J, Wang Z, Pan X, Zhang Z, Li H, Deng X, Liu P, Zhang Q, Na F, Chen C, Niu T, Liu Y. DNMT3A R882H accelerates angioimmunoblastic T-cell lymphoma in mice. Oncogene 2023:10.1038/s41388-023-02699-2. [PMID: 37127775 DOI: 10.1038/s41388-023-02699-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 05/03/2023]
Abstract
DNA methylation-related genes, including TET2, IDH2, and DNMT3A are highly frequently mutated in angioimmunoblastic T-cell lymphoma (AITL), an aggressive malignancy of T follicular helper (Tfh) cells associated with aberrant immune features. It has been shown that TET2 loss cooperates with RHOAG17V to promote AITL in mice but the functional role of DNMT3A mutations in AITL remains unclear. Here, we report that DNMT3AR882H, the most common mutation of DNMT3A in AITL, accelerates the development of Tet2-/-; RHOAG17V AITL in mice, indicated by the expansion of malignant Tfh cells and aberrant B cells, skin rash, and significantly shortened disease-free survival. To understand the underlying cellular and molecular mechanisms, we performed single-cell transcriptome analyses of lymph nodes of mice transplanted with Tet2-/-, Tet2-/-; RHOAG17V or DNMT3AR882H; Tet2-/-; RHOAG17V hematopoietic stem and progenitor cells. These single-cell landscapes reveal that DNMT3A mutation further activates Tfh cells and leads to rapid and terminal differentiation of B cells, probably through enhancing the interacting PD1/PD-L1, ICOS/ICOSL, CD28/CD86, and ICAM1/ITGAL pairs. Our study establishes the functional roles of DNMT3A mutation in AITL and sheds light on the molecular mechanisms of this disease.
Collapse
Affiliation(s)
- Jianan Zheng
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongwang Wang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangyu Pan
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhixin Zhang
- Department of Technology, Chengdu ExAb Biotechnology, LTD, Chengdu, China
| | - He Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xintong Deng
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Pengpeng Liu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qi Zhang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feifei Na
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chong Chen
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China.
| | - Yu Liu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
37
|
Lu Z, Zhong A, Liu H, Zhang M, Chen X, Pan X, Wang M, Deng X, Gao L, Zhao L, Wang J, Yang Y, Zhang Q, Wu B, Zheng J, Wang Y, Song X, Liu K, Zhang W, Chen X, Yang K, Chen X, Zhao Y, Zhao C, Wang Y, Chen L, Zhou Z, Hu J, Liu Y, Chen C. Dissecting the genetic and microenvironmental factors of gastric tumorigenesis in mice. Cell Rep 2022; 41:111482. [PMID: 36261019 DOI: 10.1016/j.celrep.2022.111482] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 07/21/2022] [Accepted: 09/19/2022] [Indexed: 11/03/2022] Open
Abstract
Gastric cancer (GC) is one of the most frequent and lethal malignancies in the world. However, our understanding of the mechanisms underlying its initiation and progression is limited. Here, we generate a series of primary GC models in mice with genome-edited gastric organoids, which elucidate the genetic drivers for sequential transformation from dysplasia to well-differentiated and poorly differentiated GC. Further, we find that the orthotopic GC, but not the subcutaneous GC even with the same genetic drivers, display remote metastasis, suggesting critical roles of the microenvironment in GC metastasis. Through single-cell RNA-seq analyses and functional studies, we show that the interaction between fibronectin 1 on stomach-specific macrophages and integrin a6β4 on GC cells promotes remote metastases. Taken together, our studies propose a strategy to model GC and dissect the genetic and microenvironmental factors driving the full-range gastric tumorigenesis.
Collapse
Affiliation(s)
- Zhenghao Lu
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ailing Zhong
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongyu Liu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mengsha Zhang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuelan Chen
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiangyu Pan
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Manli Wang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xintong Deng
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Limin Gao
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linyong Zhao
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jian Wang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yi Yang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qi Zhang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Baohong Wu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianan Zheng
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yigao Wang
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaohai Song
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kai Liu
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weihan Zhang
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaolong Chen
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Kun Yang
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinzu Chen
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - YingLan Zhao
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Chen
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zongguang Zhou
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiankun Hu
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yu Liu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Chong Chen
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
38
|
Wang M, Chen X, Tan P, Wang Y, Pan X, Lin T, Jiang Y, Wang B, Xu H, Wang Y, Yang Y, Wang J, Zhao L, Zhang J, Zhong A, Peng Y, Du J, Zhang Q, Zheng J, Chen J, Dai S, Na F, Lu Z, Liu J, Zheng X, Yang L, Zhang P, Han P, Gong Q, Zhong Q, Xiao K, Yang H, Deng H, Zhao Y, Shi H, Man J, Gou M, Zhao C, Dai L, Xue Z, Chen L, Wang Y, Zeng M, Huang C, Wei Q, Wei Y, Liu Y, Chen C. Acquired semi-squamatization during chemotherapy suggests differentiation as a therapeutic strategy for bladder cancer. Cancer Cell 2022; 40:1044-1059.e8. [PMID: 36099882 DOI: 10.1016/j.ccell.2022.08.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/16/2022] [Accepted: 08/11/2022] [Indexed: 02/06/2023]
Abstract
Cisplatin-based chemotherapy remains the primary treatment for unresectable and metastatic muscle-invasive bladder cancers (MIBCs). However, tumors frequently develop chemoresistance. Here, we established a primary and orthotopic MIBC mouse model with gene-edited organoids to recapitulate the full course of chemotherapy in patients. We found that partial squamous differentiation, called semi-squamatization, is associated with acquired chemoresistance in both mice and human MIBCs. Multi-omics analyses showed that cathepsin H (CTSH) is correlated with chemoresistance and semi-squamatization. Cathepsin inhibition by E64 treatment induces full squamous differentiation and pyroptosis, and thus specifically restrains chemoresistant MIBCs. Mechanistically, E64 treatment activates the tumor necrosis factor pathway, which is required for the terminal differentiation and pyroptosis of chemoresistant MIBC cells. Our study revealed that semi-squamatization is a type of lineage plasticity associated with chemoresistance, suggesting that differentiation via targeting of CTSH is a potential therapeutic strategy for the treatment of chemoresistant MIBCs.
Collapse
Affiliation(s)
- Manli Wang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuelan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Tan
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yiyun Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiangyu Pan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tianhai Lin
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yong Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bo Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huan Xu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuying Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yucen Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jian Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lei Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiapeng Zhang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ailing Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yiman Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiajia Du
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qi Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianan Zheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingyao Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Siqi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Feifei Na
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenghao Lu
- Chengdu OrganoidMed Medical Laboratory, West China Health Valley, Chengdu, Sichuan 610041, China
| | - Jiaming Liu
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaonan Zheng
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Yang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Peng Zhang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Han
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan 610041, China
| | - Qian Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510000, China
| | - Kai Xiao
- Laboratory of Non-Human Primate Disease Model Research, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yinglan Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hubing Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianghong Man
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing 100850, China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhihong Xue
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Musheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510000, China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yu Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Chong Chen
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|