1
|
Yu M, Si C, Xinjue H, Pan Y, Dai Y, Jin C, Han T, Yu C, Zhang J. Biglycan deficiency alleviates intestinal fibrosis through BMP-7-mediated Smad1/5/8 signaling. J Crohns Colitis 2025; 19:jjaf065. [PMID: 40249230 DOI: 10.1093/ecco-jcc/jjaf065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Indexed: 04/19/2025]
Abstract
BACKGROUND Biglycan (BGN) is a small proteoglycan rich in leucine, which plays a crucial role in the excessive production of extracellular matrix (ECM) and its association with fibrosis across various organs. Nevertheless, the precise contribution of BGN to intestinal fibrosis remains undisclosed. This study aimed to investigate the role and mechanism of BGN in intestinal fibrosis. METHODS Human Crohn's disease (CD) tissue samples were obtained from patients with Crohn's disease who underwent surgical resection of the intestine and were categorized as stenotic/nonstenotic regions. A dextran sodium sulfate (DSS)-induced mouse model of intestinal fibrosis was established. Bgn-/0 (BGN KO) mice and primary human intestinal fibroblasts were applied for the study of experimental fibrosis. Coimmunoprecipitation, immunofluorescence staining, western blot, and qRT-PCR were conducted to identify the regulatory effects of BGN on bone morphogenetic protein-7 (BMP-7) expression and intesinal fibrosis. RESULTS In both human CD samples and the DSS-induced mouse model of intestinal fibrosis, we observed a significant upregulation of BGN in areas activated by fibrosis. The genetic deletion of BGN resulted in the alleviation of intestinal fibrosis in mice administered DSS. The knockdown of BGN through siRNA significantly attenuated TGF-β1-induced ECM deposition and fibroblastic activation in primary human intestinal fibroblasts. Mechanistically, BGN directly interacted with and negatively regulated the anti-fibrotic protein BMP-7. Rescue experiments demonstrated that BGN facilitated intestinal fibrosis by modulating Smad1/5/8 phosphorylation and activating ECM deposition. CONCLUSION Our data indicate that BGN deficiency inhibits intestinal fibrosis through activation of the BMP-7-Smad1/5/8 signaling pathway. BGN and BMP-7 may become new biomarkers of intestinal fibrosis and novel targets for disease prevention and treatment.
Collapse
Affiliation(s)
- Mengli Yu
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
- Department of Gastroenterology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Chenqin Si
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 200054, China
| | - He Xinjue
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Yuanyuan Pan
- Department of Gastroenterology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yiyang Dai
- Department of Gastroenterology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Chengfeng Jin
- Department of Gastroenterology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Tiemei Han
- Department of Gastroenterology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Chaohui Yu
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jie Zhang
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
2
|
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
|
3
|
Mohammad SI, Vasudevan A, Hussein Alzewmel A, Rab SO, Ballal S, Kalia R, Bethanney Janney J, Ray S, Joshi KK, Yasin HA. The mutual effects of stearoyl-CoA desaturase and cancer-associated fibroblasts: A focus on cancer biology. Exp Cell Res 2025; 447:114508. [PMID: 40122505 DOI: 10.1016/j.yexcr.2025.114508] [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: 01/25/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
The tumor microenvironment (TME) 's primary constituents that promote cancer development are cancer-associated fibroblasts (CAFs). Metabolic remodeling has been shown to control CAF activity, particularly aberrant lipid metabolism. SCD1 can be thought of as the primary enzyme controlling the fluidity of lipid bilayers by gradually converting saturated fatty acids into monounsaturated fatty acids. Furthermore, its crucial function in the onset and spread of cancer is well acknowledged. Even with the increasing amount of research on changes in lipid metabolism, this problem remains a relatively understudied aspect of cancer research. Blocking several fatty acid synthesis-related enzymes highly expressed in cancerous cells inhibits cell division and encourages apoptosis. This is the situation with SCD1, whose overexpression has been linked to several changed tumors and cells. Both genetic and pharmacological silencing of SCD1 in cancer cells prevents glucose-mediated lipogenesis and tumor cell growth. However, its role in CAFs, hence, cancer biology, has been less studied. This study aimed to review the role of SCD1 in CAF biology, shedding light on their function in cancer cell biology.
Collapse
Affiliation(s)
- Suleiman Ibrahim Mohammad
- Research Follower, INTI International University, 71800 Negeri Sembilan, Malaysia; Electronic Marketing and Social Media, Economic and Administrative Sciences, Zarqa University, Jordan.
| | - Asokan Vasudevan
- Faculty of Business and Communications, INTI International University, 71800, Negeri Sembilan, Malaysia.
| | - Ahmad Hussein Alzewmel
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rishiv Kalia
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - J Bethanney Janney
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Hatif Abdulrazaq Yasin
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
4
|
Li C, Pan J, Shi Z, Zeng X, Xia X, He X, Wang W, Qiu B, Ding W, Huang D. Engineered Endometrial Clear Cell Cancer-on-a-Chip Reveals Early Invasion-Metastasis Cascade of Cancer Cells. Biomater Res 2025; 29:0177. [PMID: 40231208 PMCID: PMC11994883 DOI: 10.34133/bmr.0177] [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: 12/03/2024] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
Endometrial clear cell cancer (ECCC) is an extremely rare and highly malignant subtype of endometrial cancer. For most ECCC patients, cancer metastasis is the major cause of death. To date, due to the complexity of cancer evolution and the small number of cases, the metastasis of ECCC at the early stage remains largely unknown. Herein, we modeled the early invasion-metastasis cascade of ECCC by coculturing the ECCC patient-derived tumor cells (PDTCs) and primary human vascular endothelial cells on a microfluidic chip. With the chip, we for the first time replicated the dynamic migration of PDTCs into the surrounding stroma, including the intravasation and extravasation of PDTCs through the capillaries/microvessels, and presented the changes in the morphology and permeability of capillaries, with the decreased diameter and the increased permeability after cancer metastasis. We found that PDTCs were more invasive than the common endometrial adenocarcinoma cells. In addition, we preliminarily explored the inhibition of drugs on the early PDTC infiltration. This study provides new ideas for better understanding of ECCC evolution.
Collapse
Affiliation(s)
- Chengpan Li
- Department of Electronic Engineering and Information Science, School of Information Science and Technology,
University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jing Pan
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Zhengdi Shi
- Department of Electronic Engineering and Information Science, School of Information Science and Technology,
University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xinyan Zeng
- Department of Integrated Traditional Chinese and Western Medicine,
Anhui Medical University, Hefei, Anhui 230032, China
| | - Xiaoping Xia
- Department of Obstetrics and Gynecology, Anhui Provincial Children’s Hospital, Children’s Hospital of Fudan University Anhui Hospital, Children’s Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Xiaogang He
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Wei Wang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Bensheng Qiu
- Department of Electronic Engineering and Information Science, School of Information Science and Technology,
University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Weiping Ding
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Dabing Huang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei, Anhui 230001, China
| |
Collapse
|
5
|
Tian Y, Yang Y, He L, Yu X, Zhou H, Wang J. Exploring the tumor microenvironment of breast cancer to develop a prognostic model and predict immunotherapy responses. Sci Rep 2025; 15:12569. [PMID: 40221624 PMCID: PMC11993623 DOI: 10.1038/s41598-025-97784-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
Breast cancer is the most prevalent malignancy in women and exhibits significant heterogeneity. The tumor microenvironment (TME) plays a critical role in tumorigenesis, progression, and response to therapy. However, its impact on the prognosis and immunotherapy responses is incompletely understood. Using public databases, we conducted a comprehensive investigation of transcriptome and single-cell sequencing data. After performing immune infiltration analysis, we conducted consensus clustering, weighted gene co-expression network analysis (WGCNA), Cox regression, and least absolute shrinkage and selection operator (Lasso) regression to identify independent prognostic genes in breast cancer. Subsequently, we developed a prognostic model for patients with breast cancer. Tumor Immune Dysfunction and Exclusion (TIDE) values were used to assess patient's responsiveness to breast cancer. Based on single-cell RNA-sequencing data, we identified various cell types through cluster analysis and investigated the expression of prognostic model genes in each cell type. The drug sensitivity of targeted therapeutic agents for breast cancer treatment was analyzed in different cell types. We identified 12 independent prognostic genes associated with breast cancer and used these genes to construct a prognostic model. The prognostic model accurately discriminated between patients classified as high- and low-risk, providing precise prognostic predictions for individual patients. Additionally, our model exhibited a robust capacity to predict the immunotherapeutic response in breast cancer patients. Our investigation revealed a notable association between the proportion of endothelial cells (ECs) and patient prognosis in breast cancer. A prognostic model for breast cancer was formulated that showed close associations between prognosis and response to immunotherapy. For patients predicted by our model to not respond effectively to immunotherapeutic agents, it may be considered to combine immunotherapeutic agents with targeted therapeutic agents identified through our drug sensitivity analysis, which could potentially enhance treatment efficacy.
Collapse
Affiliation(s)
- Ye Tian
- Department of Thyroid and Breast Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Thyroid and Breast Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei He
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaocheng Yu
- Department of Thyroid and Breast Surgery, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hu Zhou
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Wang
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Zhao Y, Han J, Yang R, Wang S, Zhao X, Wang Z, Lu H. Evaluating the predictive significance of D-dimer in conjunction with CA724 for the postoperative outcomes in gastric cancer: A retrospective cohort analysis. PLoS One 2025; 20:e0320193. [PMID: 40198649 PMCID: PMC11978037 DOI: 10.1371/journal.pone.0320193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/15/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Gastric cancer represents a highly aggressive form of malignant tumor originating from the epithelial cells lining the gastric mucosa. Despite notable improvements in treatment approaches over the last few years, the associated mortality rate continues to be considerably high. Therefore, there is a pressing requirement for dependable biomarkers that can be utilized to predict and monitor prognosis, as well as to formulate targeted treatment strategies for patient groups at high risk. METHODS We conducted an analysis of data collected from patients who were diagnosed with gastric cancer and underwent radical gastrectomy at Shanxi Cancer Hospital from June 2017 to June 2018, with follow-up data gathered over a five-year duration until 2023. Patient follow-up information was sourced from the hospital's monitoring system. The analysis focused on the variances in effectiveness of D-dimer against different tumor markers through Cox stratification analysis. The tumor marker that exhibited the most pronounced impact was selected to formulate a novel combined indicator. Furthermore, we examined how this combined indicator influences five-year overall survival (OS) outcomes following gastric cancer surgery using Cox multivariate regression analysis. RESULTS The Cox multivariate regression analysis revealed that the effect value of the D_Dimer-CA724 Middle group on the overall survival rate post-surgery for gastric cancer was found to be 1.42 (1.13-1.78), p = 0.003 (<0.05), in comparison with the D_Dimer-CA724 Low group. For the D_Dimer-CA724 High group, the effect value on overall survival after gastric cancer surgery was 2.11 (1.65-2.68), p < 0.001. Additionally, the trend test results indicated a value of 1.46 (1.29-1.64) with p < 0.001, demonstrating statistical significance. When compared to the D_Dimer-CA724 Low group, both the D_Dimer-CA724 Middle and High groups showed markedly poorer prognoses, with increased risks of 42% and 111%, respectively, highlighting a highly significant finding in clinical practice. CONCLUSION The integrated measure of D-dimer and CA724, referred to as D-dimer_CA724, serves as an independent predictor for the postoperative outcomes of gastric cancer, demonstrating superior predictive capability compared to the individual markers. In clinical settings, patients with gastric cancer exhibiting elevated levels of D-dimer_CA724 tend to experience worse prognoses following surgery. This measure holds significant potential for widespread application and promotion within clinical practice.
Collapse
Affiliation(s)
- Yuanzheng Zhao
- Fenyang College Shanxi Medical University, Shanxi, China; No.16 College Road, Lvliang City, Shanxi Province, China
| | - Jiaqi Han
- Fenyang College Shanxi Medical University, Shanxi, China; No.16 College Road, Lvliang City, Shanxi Province, China
| | - Rong Yang
- Department of Gastroenterology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Shanxi, ChinaNo. 3, Zhigong New Street, Taiyuan City, Shanxi Province, China
| | - Shuqin Wang
- Fenyang College Shanxi Medical University, Shanxi, China; No.16 College Road, Lvliang City, Shanxi Province, China
| | - Xinran Zhao
- Fenyang College Shanxi Medical University, Shanxi, China; No.16 College Road, Lvliang City, Shanxi Province, China
| | - Ziyuan Wang
- Fenyang College Shanxi Medical University, Shanxi, China; No.16 College Road, Lvliang City, Shanxi Province, China
| | - Hongxia Lu
- Department of Gastroenterology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Shanxi, ChinaNo. 3, Zhigong New Street, Taiyuan City, Shanxi Province, China
| |
Collapse
|
7
|
Qiu Y, Wang Y, Liu J, Liu B, Sun K, Hou Q. Single-cell sequencing uncovers a high ESM1-expression endothelial cell subpopulation associated with bladder cancer progression and the immunosuppressive microenvironment. Sci Rep 2025; 15:10946. [PMID: 40159545 PMCID: PMC11955522 DOI: 10.1038/s41598-025-95731-2] [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: 01/11/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Despite remarkable advancements in therapeutic strategies, a considerable proportion of patients with bladder cancer (BC) still experience disease progression and unfavorable prognosis. The heterogeneity and biological functions of tumor endothelial cells (ECs) during BC progression remain poorly understood. We collected scRNA-seq data from BC samples and identified two EC subpopulations through hierarchical clustering analysis. The activity of signaling pathways in distinct EC subpopulations was assessed utilizing AUCell analysis. Gene regulatory networks (GRN) were constructed and analyzed for different EC subpopulations using the pySCENIC algorithm. Additionally, we investigated the association between the abundance of EC subpopulations and both clinical prognosis and immune cell infiltration. The biological effects of ESM1 protein on BC cells were further validated through EdU and Transwell assays. We analyzed 7,519 CD45-negative single cells from BC tissues and discerned two distinct EC subpopulations. The two subpopulations were characterized by high expression of ESM1 (S1 ECs) and CXCL2 (S2 ECs), respectively. In S1 ECs, we observed significant activation of signaling pathways involved in tumor promotion, including angiogenesis and cell proliferation. Additionally, our GRN analysis uncovered notable differences in transcription factor activity between S1 and S2 ECs. Moreover, ESM1 protein promoted proliferation and migration of BC cells. Patients with higher abundance of the S1 EC subpopulation exhibited more unfavorable clinical outcomes and increased infiltration of inhibitory immune cells. Our findings elucidate the transcriptional profiles and biological roles of the high ESM1-expression endothelial cell subpopulation in BC. This subpopulation is associated with poor prognosis and immunosuppressive tumor microenvironment. Accordingly, targeting endothelial cells with high ESM1 expression may offer a novel therapeutic strategy for patients with BC.
Collapse
Affiliation(s)
- Yifeng Qiu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of ShenzhenUniversity, Shenzhen, China
| | - Yuhan Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Jiahe Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
| | - Kai Sun
- Department of Radiology, the Third People's Hospital of Longgang District, Shenzhen Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518116, China.
| | - Qi Hou
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of ShenzhenUniversity, Shenzhen, China.
| |
Collapse
|
8
|
Xia H, He T, Li X, Zhao K, Zhang Z, Zhu G, Yang H, Yan X, Wang Q, Li Z, Jiang Z, Wang K, Yin X. Study on the mechanism of BGN in progression and metastasis of ccRCC. BMC Med Genomics 2025; 18:55. [PMID: 40108593 PMCID: PMC11924620 DOI: 10.1186/s12920-025-02124-5] [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: 03/24/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE To investigate the role of Biglycan(BGN) in the progression and metastasis of clear cell renal cell carcinoma(ccRCC). METHODS Based on multiple public databases, we investigated the expression level of BGN in ccRCC, its clinical significance, and its association with immune cells. Real-time fluorescence quantitative polymerase chain reaction(PCR) was employed to validate BGN expression in tumor and adjacent normal tissues from ten patients. We utilized RNA sequencing results for further analysis, including differential gene analysis, GO-KEGG analysis, and GSEA analysis, to identify the signaling pathways through which BGN exerts its effects. BGN knockdown cells(786-0 and Caki-1) were generated through lentiviral transfection to examine the impact of BGN on ccRCC. Cell proliferation, migration, and invasion were assessed using CCK8, colony formation, wound healing, Transwell migration, and invasion assays, respectively. RESULTS Our findings from database analysis and PCR revealed a significant upregulation of BGN expression in kidney cancer tissues compared to normal tissues. Further analysis demonstrated a correlation between high BGN expression and ccRCC progression and immune infiltration. In vitro experiments confirmed that BGN silencing effectively inhibited cell proliferation, migration, and invasion of ccRCC. Mechanistically, these effects may be mediated through the MAPK signaling pathway. CONCLUSION BGN potentially plays a pivotal role in the progression and metastasis of ccRCC, possibly acting through the MAPK signaling pathway. Therefore, BGN holds promise as a potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Hanqing Xia
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Tianzhen He
- Institute of Special Environmental Medicine, Nantong University, Nantong, China
| | - Xueyu Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Kai Zhao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zongliang Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Guanqun Zhu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Han Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Xuechuan Yan
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Qinglei Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zhaofeng Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zaiqing Jiang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Ke Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Xinbao Yin
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China.
| |
Collapse
|
9
|
Tong J, Dong X, Martin TA, Yang Y, Dong B, Jiang WG. DRIM modulates Src activation and regulates angiogenic functions in vascular endothelial cells. Cell Biol Int 2025; 49:277-287. [PMID: 39648301 PMCID: PMC11811745 DOI: 10.1002/cbin.12265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 12/10/2024]
Abstract
Downregulated in Metastasis Protein (DRIM) was discovered in malignant epithelial cells and was thought to be mainly a nucleus protein affecting cancer cells. Recent single-cell sequencing analysis suggests that DRIM is abundantly expressed in vascular endothelial cells. There has been no knowledge of the role of DRIM in the endothelium. In the present study, using protein fraction method and cell imaging, we identified that the DRIM protein was abundantly present in both nucleus and the cytoskeletal fractions of human vascular endothelial cells. Knockdown of DRIM in the endothelial cells significantly affected growth, migration, and angiogenic tubule formation. Proteomics analyses revealed that Src was an important direct target protein of DRIM, a finding further confirmed by protein interaction assay. Silencing DRIM activated the tyrosine 419 site phosphorylation of Src kinase in endothelial cells, thereby affecting the downstream proteins of Src including p-FAK and p-STAT3, and exerting biological effects. To conclude, our results provide evidence of DRIM being a nuclear and cytoskeletal-associated protein, having a novel key role of the protein in vascular endothelial cells.
Collapse
Affiliation(s)
- Jia Tong
- Department of Geriatric Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityShandong First Medical UniversityJinanChina
- Cardiff China Medical Research CollaborativeDivision of Cancer and Genetics, Cardiff University School of MedicineCardiffUK
| | - Xuefei Dong
- Cardiff China Medical Research CollaborativeDivision of Cancer and Genetics, Cardiff University School of MedicineCardiffUK
| | - Tracey A. Martin
- Cardiff China Medical Research CollaborativeDivision of Cancer and Genetics, Cardiff University School of MedicineCardiffUK
| | - Yiming Yang
- Cardiff China Medical Research CollaborativeDivision of Cancer and Genetics, Cardiff University School of MedicineCardiffUK
| | - Bo Dong
- Department of Geriatric Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityShandong First Medical UniversityJinanChina
- Department of Cardiology, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Wen G. Jiang
- Cardiff China Medical Research CollaborativeDivision of Cancer and Genetics, Cardiff University School of MedicineCardiffUK
| |
Collapse
|
10
|
Wang R, Liu G, Wang K, Pan Z, Pei Z, Hu X. Hypoxia signature derived from tumor-associated endothelial cells predict prognosis in gastric cancer. Front Cell Dev Biol 2025; 13:1515681. [PMID: 39901877 PMCID: PMC11788339 DOI: 10.3389/fcell.2025.1515681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/03/2025] [Indexed: 02/05/2025] Open
Abstract
Background A hypoxic metabolism environment in the tumors is often associated with poor prognostic events such as tumor progression and treatment resistance. In gastric cancer, the mechanism of how hypoxia metabolism affects the tumor microenvironment and immunotherapy efficacy remains to be elucidated. Methods We used the bulk-mapping method to analyze the signatures correlated with the response of immunotherapy in the single-cell dataset. Cellular, pathway, and gene were systematically analyzed in both single-cell and bulk validation datasets. Results The most significant cell proportion difference between the response and non-response groups was in endothelial cells, which represent the malignant cells. VWF was specifically overexpressed in endothelial cells and was the hub gene of differential genes. EPAS1 was a VWF trans-regulated gene and highly positively correlated with VWF in expression. Knockdown experiments demonstrated that siVWF reduced the expression of VWF, EPAS1, and HIF1A, as well as the synthesis of lactate and adenosine which are indicators of hypoxic metabolism. These results suggest that the overexpression of core malign endothelial genes such as VWF drives hypoxic metabolism in tumors and creates an immunosuppressive environment that reduces the efficacy of immunotherapy. The adverse prognosis of the hypoxia signature was validated in the bulk cohort and significance was further enhanced after selecting core genes and combined survival weight scoring. Conclusion In summary, high expression of the malignant endothelial cell driver genes VWF and EPAS1 enhances hypoxic metabolism, and malignant cell-immune cell interactions suppress the immune response. Therefore, the two core genes of hypoxic metabolism might represent potential therapeutic and predicting biomarkers for immunotherapy of gastric cancer in the future.
Collapse
Affiliation(s)
- Ruiheng Wang
- Surgical Ward, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Guijun Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of administrative, The Fourth Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ke Wang
- Endoscopy Room, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Zhanglei Pan
- Surgical Ward, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhihua Pei
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Xijiao Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Postdoctoral Research Station of Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
11
|
Liu X. Effects of Shenmai Injection on proliferation, migration, invasion and angiogenesis of colorectal carcinoma vascular endothelial cells. Medicine (Baltimore) 2025; 104:e41307. [PMID: 39833060 PMCID: PMC11749673 DOI: 10.1097/md.0000000000041307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 10/24/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Colorectal cancer, being 1 of the most significant malignant tumors globally, poses a substantial risk to human health. Unfortunately, its 5-year survival rate stands at a mere 65%. There remains an urgent need for the development of novel treatments to combat this detrimental malignancy effectively. The Shenmai Injection (SMI) is a Chinese medicine that has been proven to have significant clinical efficacy in the treatment of cardiovascular diseases. This study aimed to examine the impact of SMI on the proliferation, migration, invasion, and angiogenesis of tumor-derived endothelial cells (Td-EC). METHODS Human umbilical vein endothelial cells (HUVEC) induced Td-EC, and HUVEC were treated with conditioned media from the human colorectal carcinoma cells (HCT116). The effects of HCT116 on the proliferation, migration, and invasion of hepatocellular carcinoma cells after treatment of SMI were observed by MTS assay and Transwell techniques. Additionally, an angiogenesis experiment was used to investigate Td-EC tube formation capacity. RESULTS SMI had a significant inhibiting effect on the proliferation, migration, and invasion of HCT116. SMI was also able to inhibit the angiogenesis of Td-EC. Notably, SMI did not have any effect on the normal endothelium. CONCLUSION SMI has obvious antiproliferation, migration, infiltration, and neogenesis effects on HCT116.
Collapse
Affiliation(s)
- Xiangyu Liu
- Lanzhou University Second Hospital, Chengguan District, Lanzhou, China
| |
Collapse
|
12
|
Ji B, Wang X, Wang X, Xu L, Peng S. scDCA: deciphering the dominant cell communication assembly of downstream functional events from single-cell RNA-seq data. Brief Bioinform 2024; 26:bbae663. [PMID: 39694816 DOI: 10.1093/bib/bbae663] [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: 10/21/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cell-cell communications (CCCs) involve signaling from multiple sender cells that collectively impact downstream functional processes in receiver cells. Currently, computational methods are lacking for quantifying the contribution of pairwise combinations of cell types to specific functional processes in receiver cells (e.g. target gene expression or cell states). This limitation has impeded understanding the underlying mechanisms of cancer progression and identifying potential therapeutic targets. Here, we proposed a deep learning-based method, scDCA, to decipher the dominant cell communication assembly (DCA) that have a higher impact on a particular functional event in receiver cells from single-cell RNA-seq data. Specifically, scDCA employed a multi-view graph convolution network to reconstruct the CCCs landscape at single-cell resolution, and then identified DCA by interpreting the model with the attention mechanism. Taking the samples from advanced renal cell carcinoma as a case study, the scDCA was successfully applied and validated in revealing the DCA affecting the crucial gene expression in immune cells. The scDCA was also applied and validated in revealing the DCA responsible for the variation of 14 typical functional states of malignant cells. Furthermore, the scDCA was applied and validated to explore the alteration of CCCs under clinical intervention by comparing the DCA for certain cytotoxic factors between patients with and without immunotherapy. In summary, scDCA provides a valuable and practical tool for deciphering the cell type combinations with the most dominant impact on a specific functional process of receiver cells, which is of great significance for precise cancer treatment. Our data and code are free available at a public GitHub repository: https://github.com/pengsl-lab/scDCA.git.
Collapse
Affiliation(s)
- Boya Ji
- College of Computer Science and Electronic Engineering, Hunan University, Yuelu, 410006 Changsha, China
| | - Xiaoqi Wang
- College of Computer Science and Electronic Engineering, Hunan University, Yuelu, 410006 Changsha, China
| | - Xiang Wang
- The Second Xiangya Hospital, Central South University, Yuelu, 410006 Changsha, China
| | - Liwen Xu
- College of Computer Science and Electronic Engineering, Hunan University, Yuelu, 410006 Changsha, China
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, Yuelu, 410006 Changsha, China
| |
Collapse
|
13
|
Tang Y, Li Q, Zhou Z, Bai H, Xiao N, Xie J, Li C. Nitric oxide-based multi-synergistic nanomedicine: an emerging therapeutic for anticancer. J Nanobiotechnology 2024; 22:674. [PMID: 39497134 PMCID: PMC11536969 DOI: 10.1186/s12951-024-02929-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/11/2024] [Indexed: 11/06/2024] Open
Abstract
Gas therapy has emerged as a promising approach for treating cancer, with gases like NO, H2S, and CO showing positive effects. Among these, NO is considered a key gas molecule with significant potential in stopping cancer progression. However, due to its high reactivity and short half-life, delivering NO directly to tumors is crucial for enhancing cancer treatment. NO-driven nanomedicines (NONs) have been developed to effectively deliver NO donors to tumors, showing great progress in recent years. This review provides an overview of the latest advancements in NO-based cancer nanotherapeutics. It discusses the types of NO donors used in current research, the mechanisms of action behind NO therapy for cancer, and the different delivery systems for NO donors in nanotherapeutics. It also explores the potential of combining NO donors with other treatments for enhanced cancer therapy. Finally, it examines the future prospects and challenges of using NONs in clinical settings for cancer treatment.
Collapse
Affiliation(s)
- Yaqin Tang
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Qiyu Li
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Ziwei Zhou
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Huayang Bai
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Nianting Xiao
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Jing Xie
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, People's Republic of China.
| | - Chong Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
14
|
Tomassetti C, Insinga G, Gimigliano F, Morrione A, Giordano A, Giurisato E. Insights into CSF-1R Expression in the Tumor Microenvironment. Biomedicines 2024; 12:2381. [PMID: 39457693 PMCID: PMC11504891 DOI: 10.3390/biomedicines12102381] [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: 08/22/2024] [Revised: 09/30/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
The colony-stimulating factor 1 receptor (CSF-1R) plays a pivotal role in orchestrating cellular interactions within the tumor microenvironment (TME). Although the CSF-1R has been extensively studied in myeloid cells, the expression of this receptor and its emerging role in other cell types in the TME need to be further analyzed. This review explores the multifaceted functions of the CSF-1R across various TME cellular populations, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), cancer-associated fibroblasts (CAFs), endothelial cells (ECs), and cancer stem cells (CSCs). The activation of the CSF-1R by its ligands, colony-stimulating factor 1 (CSF-1) and Interleukin-34 (IL-34), regulates TAM polarization towards an immunosuppressive M2 phenotype, promoting tumor progression and immune evasion. Similarly, CSF-1R signaling influences MDSCs to exert immunosuppressive functions, hindering anti-tumor immunity. In DCs, the CSF-1R alters antigen-presenting capabilities, compromising immune surveillance against cancer cells. CSF-1R expression in CAFs and ECs regulates immune modulation, angiogenesis, and immune cell trafficking within the TME, fostering a pro-tumorigenic milieu. Notably, the CSF-1R in CSCs contributes to tumor aggressiveness and therapeutic resistance through interactions with TAMs and the modulation of stemness features. Understanding the diverse roles of the CSF-1R in the TME underscores its potential as a therapeutic target for cancer treatment, aiming at disrupting pro-tumorigenic cellular crosstalk and enhancing anti-tumor immune responses.
Collapse
Affiliation(s)
- Caterina Tomassetti
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | - Gaia Insinga
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.I.); (F.G.)
| | - Francesca Gimigliano
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.I.); (F.G.)
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| |
Collapse
|
15
|
Guo M, Sun Y, Wang X, Wang Z, Yuan X, Chen X, Yuan X, Wang L. The MCIB Model: A Novel Theory for Describing the Spatial Heterogeneity of the Tumor Microenvironment. Int J Mol Sci 2024; 25:10486. [PMID: 39408814 PMCID: PMC11476373 DOI: 10.3390/ijms251910486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The tumor microenvironment (TME) can be regarded as a complex and dynamic microecosystem generated by the interactions of tumor cells, interstitial cells, the extracellular matrix, and their products and plays an important role in the occurrence, progression and metastasis of tumors. In a previous study, we constructed an IEO model (prI-, prE-, and pOst-metastatic niche) according to the chronological sequence of TME development. In this paper, to fill the theoretical gap in spatial heterogeneity in the TME, we defined an MCIB model (Metabolic, Circulatory, Immune, and microBial microenvironment). The MCIB model divides the TME into four subtypes that interact with each other in terms of mechanism, corresponding to the four major links of metabolic reprogramming, vascular remodeling, immune response, and microbial action, providing a new way to assess the TME. The combination of the MCIB model and IEO model comprehensively depicts the spatiotemporal evolution of the TME and can provide a theoretical basis for the combination of clinical targeted therapy, immunotherapy, and other comprehensive treatment modalities for tumors according to the combination and crosstalk of different subtypes in the MCIB model and provide a powerful research paradigm for tumor drug-resistance mechanisms and tumor biological behavior.
Collapse
Affiliation(s)
- Minghao Guo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.Y.); (X.C.)
| | - Yinan Sun
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.S.)
| | - Xiaohui Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Zikun Wang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.S.)
| | - Xun Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.Y.); (X.C.)
| | - Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.Y.); (X.C.)
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.Y.); (X.C.)
| | - Lu Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (M.G.); (X.Y.); (X.C.)
| |
Collapse
|
16
|
Feng Q, Liu Q, Liu Z, Xu J, Yang Y, Zhu Y, Lu G, Xu G, Wu D, Wang F, Liu B, Wang W, Ding X. USP9X inhibits metastasis in pulmonary sarcomatoid carcinoma by regulating epithelial-mesenchymal transition, angiogenesis and immune infiltration. Transl Oncol 2024; 47:101950. [PMID: 38964032 PMCID: PMC11283126 DOI: 10.1016/j.tranon.2024.101950] [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: 01/16/2024] [Revised: 03/06/2024] [Accepted: 03/26/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Pulmonary sarcomatoid carcinoma (PSC) is a highly invasive pulmonary malignancy with an extremely poor prognosis. The results of previous studies suggest that ubiquitin-specific peptidase 9X (USP9X) contributes to the progression of numerous types of cancer. Nevertheless, there is little knowledge about the molecular mechanisms and functions of USP9X in the metastasis of PSC. METHODS Immunohistochemistry and western blotting were used to detect USP9X expression levels in PSC tissues and cells. Wound healing, transwell, enzyme-linked immunosorbent assay (ELISA), tube formation, and aortic ring assays were used to examine the function and mechanism of USP9X in the metastasis of PSC. RESULTS Expression of USP9X was markedly decreased and significantly correlated with metastasis and prognosis of patients with PSC. Then we revealed that USP9X protein levels were negatively associated with the levels of epithelial-mesenchymal transition (EMT) markers and the migration of PSC cells. It was confirmed that USP9X in PSC cells reduced VEGF secretion and inhibited tubule formation of human umbilical vein endothelial cells (HUVEC) in vitro. USP9X was detected to downregulate MMP9. Meanwhile, MMP9 was positively related to EMT, angiogenesis and was negatively related to immune infiltration in the public databases. USP9X was significantly negatively associated with the expression of MMP9, EMT markers, CD31, and positively associated with CD4, and CD8 in PSC tissues. CONCLUSION The present study reveals the vital role of USP9X in regulating EMT, angiogenesis and immune infiltration and inhibiting metastasis of PSC via downregulating MMP9, which provides a new effective therapeutic target for PSC.
Collapse
Affiliation(s)
- Qin Feng
- Medical Science and Technology Innovation Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Minicipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Qian Liu
- Department of Pathology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zi Liu
- Medical Science and Technology Innovation Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Minicipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Jianyu Xu
- Medical Science and Technology Innovation Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Minicipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Yang Yang
- Department of Pathology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Ying Zhu
- Medical Science and Technology Innovation Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Minicipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Guangxian Lu
- Medical Science and Technology Innovation Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Minicipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Guangjuan Xu
- Medical Science and Technology Innovation Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Minicipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Dan Wu
- Medical Science and Technology Innovation Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Minicipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Feng Wang
- Medical Science and Technology Innovation Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Minicipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China
| | - Biao Liu
- Department of Pathology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| | - Wenjuan Wang
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, China.
| | - Xinyuan Ding
- Medical Science and Technology Innovation Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Minicipal Hospital, Gusu School of Nanjing Medical University, Suzhou, China.
| |
Collapse
|
17
|
Choi SH, Kim DY. Regulation of Tumor Microenvironment through YAP/TAZ under Tumor Hypoxia. Cancers (Basel) 2024; 16:3030. [PMID: 39272887 PMCID: PMC11394240 DOI: 10.3390/cancers16173030] [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: 07/16/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
In solid tumors such as hepatocellular carcinoma (HCC), hypoxia is one of the important mechanisms of cancer development that closely influences cancer development, survival, and metastasis. The development of treatments for cancer was temporarily revolutionized by immunotherapy but continues to be constrained by limited response rates and the resistance and high costs required for the development of new and innovative strategies. In particular, solid tumors, including HCC, a multi-vascular tumor type, are sensitive to hypoxia and generate many blood vessels for metastasis and development, making it difficult to treat HCC, not only with immunotherapy but also with drugs targeting blood vessels. Therefore, in order to develop a treatment strategy for hypoxic tumors, various mechanisms must be explored and analyzed to treat these impregnable solid tumors. To date, tumor growth mechanisms linked to hypoxia are known to be complex and coexist with various signal pathways, but recently, mechanisms related to the Hippo signal pathway are emerging. Interestingly, Hippo YAP/TAZ, which appear during early tumor and normal tumor growth, and YAP/TAZ, which appear during hypoxia, help tumor growth and proliferation in different directions. Peculiarly, YAP/TAZ, which have different phosphorylation directions in the hypoxic environment of tumors, are involved in cancer proliferation and metastasis in various carcinomas, including HCC. Analyzing the mechanisms that regulate the function and expression of YAP in addition to HIF in the complex hypoxic environment of tumors may lead to a variety of anti-cancer strategies and combining HIF and YAP/TAZ may develop the potential to change the landscape of cancer treatment.
Collapse
Affiliation(s)
- Sung Hoon Choi
- Institute of Health & Environment, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- KoBioLabs Inc., Seoul 08826, Republic of Korea
| | - Do Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- Yonsei Liver Cancer Center, Yonsei Cancer Hospital, Seoul 03722, Republic of Korea
| |
Collapse
|
18
|
Meijuan C, Fang M, Qian W. Dachsous cadherin related 1 (DCHS1) is a novel biomarker for immune infiltration and epithelial-mesenchymal transition in endometrial cancer via pan-cancer analysis. J Ovarian Res 2024; 17:162. [PMID: 39123216 PMCID: PMC11312386 DOI: 10.1186/s13048-024-01478-1] [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: 08/11/2023] [Accepted: 07/15/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Dachsous cadherin related 1 (DCHS1) is one of calcium-dependent adhesion membrane proteins and is mainly involved in the development of mammalian tissues. There is a lack of more detailed research on the biological function of DCHS1 in pan-cancer. MATERIALS AND METHODS We evaluated the expression, the prognostic value, the diagnostic value and genomic alterations of DCHS1 by using the databases, including TCGA, UALCAN, HPA, GEPIA2.0 and GSCA. We employed the databases of UCSC, TIMER2.0, TISIDB, GSCA to analyze the association between DCHS1 expression and the immune microenvironment, stemness, TMB, MSI and anticancer drug sensitivity. BioGRID, STRING and GEPIA2.0 were used to perform protein interaction and functional enrichment analysis. Real-time quantitative PCR, CCK8, Transwell assay and Western blot were performed to determine the function of DCHS1 in UCEC. RESULTS DCHS1 is differentially expressed in many cancers and its expression is significantly associated with tumor prognosis and diagnosis. DCHS1 expression was significantly correlated with the infiltration of cancer-associated fibroblasts (CAFs), Endothelial cell (ECs), and Hematopoietic stem cell in most cancers. In addition, DCHS1 was significantly associated with sensitivity to many antitumor drugs. Functional enrichment analysis revealed that DCHS1-related proteins were involved in Focal adhesion, Endometrial cancer and Wnt signaling pathway. GSEA results showed that DCHS1 was related to epithelial-mesenchymal transition (EMT) in many cancers. In vitro experiments in UCEC showed that DCHS1 regulated cell proliferation, migration and EMT. CONCLUSIONS Our findings indicated that DCHS1 might be a novel prognostic and diagnostic biomarker and immunotherapy target, and plays an important role in the proliferation, migration and EMT in UCEC.
Collapse
Affiliation(s)
- Cai Meijuan
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), No.758 Hefei Road, Qingdao, 266035, Shandong, China
| | - Min Fang
- Department of Gynecology, Qingdao Women's and Children's Hospital, Qingdao University, Qingdao, Shandong, China
| | - Wang Qian
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), No.758 Hefei Road, Qingdao, 266035, Shandong, China.
| |
Collapse
|
19
|
Takahashi Y, Morimura R, Tsukamoto K, Gomi S, Yamada A, Mizukami M, Naito Y, Irie S, Nagayama S, Shinozaki E, Yamaguchi K, Fujita N, Kitano S, Katayama R, Matsusaki M. In vitro throughput screening of anticancer drugs using patient-derived cell lines cultured on vascularized three-dimensional stromal tissues. Acta Biomater 2024; 183:111-129. [PMID: 38801868 DOI: 10.1016/j.actbio.2024.05.037] [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: 02/04/2024] [Revised: 05/06/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
The development of high-throughput anticancer drug screening methods using patient-derived cancer cell (PDC) lines that maintain their original characteristics in an in vitro three-dimensional (3D) culture system poses a significant challenge to achieving personalized cancer medicine. Because stromal tissue plays a critical role in the composition and maintenance of the cancer microenvironment, in vitro 3D-culture using reconstructed stromal tissues has attracted considerable attention. Here, a simple and unique in vitro 3D-culture method using heparin and collagen together with fibroblasts and endothelial cells to fabricate vascularized 3D-stromal tissues for in vitro culture of PDCs is reported. Whereas co-treatment with bevacizumab, a monoclonal antibody against vascular endothelial growth factor, and 5-fluorouracil significantly reduced the survival rate of 3D-cultured PDCs to 30%, separate addition of each drug did not induce comparable strong cytotoxicity, suggesting the possibility of evaluating the combined effect of anticancer drugs and angiogenesis inhibitors. Surprisingly, drug evaluation using eight PDC lines with the 3D-culture method resulted in a drug efficacy concordance rate of 75% with clinical outcomes. The model is expected to be applicable to in vitro throughput drug screening for the development of personalized cancer medicine. STATEMENT OF SIGNIFICANCE: To replicate the cancer microenvironment, we constructed a cancer-stromal tissue model in which cancer cells are placed above and inside stromal tissue with vascular network structures derived from vascular endothelial cells in fibroblast tissue using CAViTs method. Using this method, we were able to reproduce the invasion and metastasis processes of cancer cells observed in vivo. Using patient-derived cancer cells, we assessed the possibility of evaluating the combined effect with an angiogenesis inhibitor. Further, primary cancer cells also grew on the stromal tissues with the normal medium. These data suggest that the model may be useful for new in vitro drug screening and personalized cancer medicine.
Collapse
Affiliation(s)
- Yuki Takahashi
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan; Division of Clinical Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Rii Morimura
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan; Division of Clinical Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Kei Tsukamoto
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan
| | - Sayaka Gomi
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan
| | - Asuka Yamada
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan; Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Miki Mizukami
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan
| | - Yasuyuki Naito
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan; Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Shinji Irie
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Satoshi Nagayama
- Department of Colorectal Surgery, Gastroenterological Cancer Center, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; Department of Surgery, Uji Tokushukai Medical Center, Kyoto 611-0041, Japan
| | - Eiji Shinozaki
- Department of Gastroenterological Chemotherapy, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Naoya Fujita
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Shiro Kitano
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan; Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan.
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan; Department of Applied Chemistry Graduate School of Engineering Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
20
|
Xu D, Zhuang X, Ma H, Li Z, Wei L, Luo J, Han H. Altered tumor microenvironment heterogeneity of penile cancer during progression from non-lymphatic to lymphatic metastasis. Cancer Med 2024; 13:e70025. [PMID: 39003681 PMCID: PMC11246611 DOI: 10.1002/cam4.70025] [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: 05/20/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Lymphatic metastasis is the major challenge in the treatment of penile cancer. The prognosis of individuals with lymphatic metastasis is extremely poor. Therefore, early identification of disease progression and lymphatic metastasis is an urgent task for researchers in penile cancer worldwide. METHODS In this study, using single-cell RNA sequencing, an immune landscape was established for the cancer ecosystem based on 46,861 cells from six patients with penile cancer (four with lymphatic metastasis [stage IV] and two without lymphatic metastasis [stage I]). Using bulk RNA sequencing, the discrepancy between the cancers and their respective metastatic lymph nodes was depicted based on seven patients with penile cancer. RESULTS The interaction between epithelial cells, fibroblasts, and endothelial cells, and the functional cooperation among invasion, epithelial-mesenchymal transition, and angiogenesis were found to be important landscapes in the penile cancer ecosystem, playing important roles in progression of cancer and lymph node metastasis. CONCLUSIONS This study is the first to investigate the altered tumor microenvironment heterogeneity of penile cancer as it evolves from non-lymphatic to lymphatic metastasis and provides insights into the mechanisms underlying malignant progression, the premetastatic niche, and lymphatic metastasis in penile cancer.
Collapse
Affiliation(s)
- Da‐Ming Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
- Department of UrologySun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Xiao‐Yu Zhuang
- Department of AnesthesiologySecond Affiliated Hospital of Shantou University Medical CollegeShantouP. R. China
| | - Hua‐Li Ma
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
- Department of RadiologySun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Zai‐Shang Li
- Department of Urology, Shenzhen People's HospitalThe Second Clinic Medical College of Jinan UniversityShenzhenP. R. China
| | - Li‐Chao Wei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
- Department of UrologySun Yat‐sen University Cancer CenterGuangzhouP. R. China
| | - Jun‐Hang Luo
- Department of Urology, First Affiliated HospitalSun Yat‐sen UniversityGuangzhouP. R. China
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouP. R. China
| | - Hui Han
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouP. R. China
- Department of UrologySun Yat‐sen University Cancer CenterGuangzhouP. R. China
| |
Collapse
|
21
|
Cui L, Liu T, Huang C, Yang F, Luo L, Sun L, Zhao Y, Wang D, Wang M, Ji Y, Zhu W. Gastric Cancer Mesenchymal Stem Cells Trigger Endothelial Cell Functional Changes to Promote Cancer Progression. Stem Cell Rev Rep 2024; 20:1285-1298. [PMID: 38598065 DOI: 10.1007/s12015-024-10720-8] [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] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
Our previous studies have highlighted the pivotal role of gastric cancer mesenchymal stem cells (GCMSCs) in tumor initiation, progression, and metastasis. In parallel, it is well-documented that endothelial cells (ECs) undergo functional alterations in response to challenging tumor microenvironment. This study aims to elucidate whether functional changes in ECs might be induced by GCMSCs and thus influence cancer progression. Cell proliferation was assessed through CCK-8 and colony formation assays, while cell migration and invasion capabilities were evaluated by wound-healing and Transwell assays. Immunohistochemistry was employed to examine protein distribution and expression levels. Additionally, quantitative analysis of protein and mRNA expression was carried out through Western blotting and qRT-PCR respectively, with gene knockdown achieved using siRNA. Our findings revealed that GCMSCs effectively stimulate cell proliferation, migration, and angiogenesis of human umbilical vein endothelial cells (HUVECs), both in vitro and in vivo. GCMSCs promote the migration and invasion of gastric cancer cells by inducing the expression of Slit2 in HUVECs. Notably, the inhibition of phosphorylated AKT partially mitigates the aforementioned effects. In conclusion, GCMSCs may exert regulatory control over Slit2 expression in ECs via the AKT signaling pathway, thereby inducing functional changes in ECs that promote tumor progression.
Collapse
Affiliation(s)
- Linjing Cui
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu Province, 212013, China
| | - Ting Liu
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu Province, 212013, China
| | - Chao Huang
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu Province, 212013, China
| | - Fumeng Yang
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu Province, 212013, China
| | - Liqi Luo
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu Province, 212013, China
| | - Li Sun
- Department of Clinical Laboratory, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, China
| | - Yuanyuan Zhao
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu Province, 212013, China
| | - Deqiang Wang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Mei Wang
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu Province, 212013, China
| | - Yong Ji
- Department of Surgery, Jingjiang People's Hospital, Jingjiang, Jiangsu Province, China
| | - Wei Zhu
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu Province, 212013, China.
| |
Collapse
|
22
|
Li M, Wang J, Zhao Y, Lin C, Miao J, Ma X, Ye Z, Chen C, Tao K, Zhu P, Hu Q, Sun J, Gu J, Wei S. Identifying and evaluating a disulfidptosis-related gene signature to predict prognosis in colorectal adenocarcinoma patients. Front Immunol 2024; 15:1344637. [PMID: 38962013 PMCID: PMC11220892 DOI: 10.3389/fimmu.2024.1344637] [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: 11/26/2023] [Accepted: 05/28/2024] [Indexed: 07/05/2024] Open
Abstract
Disulfidptosis, a regulated form of cell death, has been recently reported in cancers characterized by high SLC7A11 expression, including invasive breast carcinoma, lung adenocarcinoma, and hepatocellular carcinoma. However, its role in colon adenocarcinoma (COAD) has been infrequently discussed. In this study, we developed and validated a prognostic model based on 20 disulfidptosis-related genes (DRGs) using LASSO and Cox regression analyses. The robustness and practicality of this model were assessed via a nomogram. Subsequent correlation and enrichment analysis revealed a relationship between the risk score, several critical cancer-related biological processes, immune cell infiltration, and the expression of oncogenes and cell senescence-related genes. POU4F1, a significant component of our model, might function as an oncogene due to its upregulation in COAD tumors and its positive correlation with oncogene expression. In vitro assays demonstrated that POU4F1 knockdown noticeably decreased cell proliferation and migration but increased cell senescence in COAD cells. We further investigated the regulatory role of the DRG in disulfidptosis by culturing cells in a glucose-deprived medium. In summary, our research revealed and confirmed a DRG-based risk prediction model for COAD patients and verified the role of POU4F1 in promoting cell proliferation, migration, and disulfidptosis.
Collapse
Affiliation(s)
- Ming Li
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of General Surgery, Changshu Hospital Affiliated to Soochow University, The First People’s Hospital of Changshu, Changshu, Jiangsu, China
| | - Jin Wang
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yuhao Zhao
- Department of Biliary and Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Changjie Lin
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianqing Miao
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoming Ma
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhenyu Ye
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chao Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ke Tao
- Department of General Surgery, Changshu Hospital Affiliated to Soochow University, The First People’s Hospital of Changshu, Changshu, Jiangsu, China
| | - Pengcheng Zhu
- Department of General Surgery, Changshu Hospital Affiliated to Soochow University, The First People’s Hospital of Changshu, Changshu, Jiangsu, China
| | - Qi Hu
- Department of General Surgery, Changshu Hospital Affiliated to Soochow University, The First People’s Hospital of Changshu, Changshu, Jiangsu, China
| | - Jinbing Sun
- Department of General Surgery, Changshu Hospital Affiliated to Soochow University, The First People’s Hospital of Changshu, Changshu, Jiangsu, China
| | - Jianfeng Gu
- Department of General Surgery, Changshu Hospital Affiliated to Soochow University, The First People’s Hospital of Changshu, Changshu, Jiangsu, China
| | - Shaohua Wei
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
23
|
Thakur A, Rana M, Mishra A, Kaur C, Pan CH, Nepali K. Recent advances and future directions on small molecule VEGFR inhibitors in oncological conditions. Eur J Med Chem 2024; 272:116472. [PMID: 38728867 DOI: 10.1016/j.ejmech.2024.116472] [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: 03/07/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024]
Abstract
"A journey of mixed emotions" is a quote that best describes the progress chart of vascular endothelial growth factor receptor (VEGFR) inhibitors as cancer therapeutics in the last decade. Exhilarated with the Food and Drug Administration (FDA) approvals of numerous VEGFR inhibitors coupled with the annoyance of encountering the complications associated with their use, drug discovery enthusiasts are on their toes with an unswerving determination to enhance the rate of translation of VEGFR inhibitors from preclinical to clinical stage. The recently crafted armory of VEGFR inhibitors is a testament to their growing dominance over other antiangiogenic therapies for cancer treatment. This review perspicuously underscores the earnest attempts of the researchers to extract the antiproliferative potential of VEGFR inhibitors through the design of mechanistically diverse structural assemblages. Moreover, this review encompasses sections on structural/molecular properties and physiological functions of VEGFR, FDA-approved VEGFR inhibitors, and hurdles restricting the activity range/clinical applicability of VEGFR targeting antitumor agents. In addition, tactics to overcome the limitations of VEGFR inhibitors are discussed. A clear-cut viewpoint transmitted through this compilation can provide practical directions to push the cart of VEGFR inhibitors to advanced-stage clinical investigations in diverse malignancies.
Collapse
Affiliation(s)
- Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Mandeep Rana
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Anshul Mishra
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Charanjit Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Chun-Hsu Pan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
24
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [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: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
25
|
Xiao B, Li G, Gulizeba H, Liu H, Sima X, Zhou T, Huang Y. Choline metabolism reprogramming mediates an immunosuppressive microenvironment in non-small cell lung cancer (NSCLC) by promoting tumor-associated macrophage functional polarization and endothelial cell proliferation. J Transl Med 2024; 22:442. [PMID: 38730286 PMCID: PMC11084143 DOI: 10.1186/s12967-024-05242-3] [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: 02/06/2024] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
INTRODUCTION Lung cancer is a prevalent malignancy globally, and immunotherapy has revolutionized its treatment. However, resistance to immunotherapy remains a challenge. Abnormal cholinesterase (ChE) activity and choline metabolism are associated with tumor oncogenesis, progression, and poor prognosis in multiple cancers. Yet, the precise mechanism underlying the relationship between ChE, choline metabolism and tumor immune microenvironment in lung cancer, and the response and resistance of immunotherapy still unclear. METHODS Firstly, 277 advanced non-small cell lung cancer (NSCLC) patients receiving first-line immunotherapy in Sun Yat-sen University Cancer Center were enrolled in the study. Pretreatment and the alteration of ChE after 2 courses of immunotherapy and survival outcomes were collected. Kaplan-Meier survival and cox regression analysis were performed, and nomogram was conducted to identify the prognostic and predicted values. Secondly, choline metabolism-related genes were screened using Cox regression, and a prognostic model was constructed. Functional enrichment analysis and immune microenvironment analysis were also conducted. Lastly, to gain further insights into potential mechanisms, single-cell analysis was performed. RESULTS Firstly, baseline high level ChE and the elevation of ChE after immunotherapy were significantly associated with better survival outcomes for advanced NSCLC. Constructed nomogram based on the significant variables from the multivariate Cox analysis performed well in discrimination and calibration. Secondly, 4 choline metabolism-related genes (MTHFD1, PDGFB, PIK3R3, CHKB) were screened and developed a risk signature that was found to be related to a poorer prognosis. Further analysis revealed that the choline metabolism-related genes signature was associated with immunosuppressive tumor microenvironment, immune escape and metabolic reprogramming. scRNA-seq showed that MTHFD1 was specifically distributed in tumor-associated macrophages (TAMs), mediating the differentiation and immunosuppressive functions of macrophages, which may potentially impact endothelial cell proliferation and tumor angiogenesis. CONCLUSION Our study highlights the discovery of ChE as a prognostic marker in advanced NSCLC, suggesting its potential for identifying patients who may benefit from immunotherapy. Additionally, we developed a prognostic signature based on choline metabolism-related genes, revealing the correlation with the immunosuppressive microenvironment and uncovering the role of MTHFD1 in macrophage differentiation and endothelial cell proliferation, providing insights into the intricate workings of choline metabolism in NSCLC pathogenesis.
Collapse
Affiliation(s)
- Bijing Xiao
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Guanjun Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, No. 1023-1063, Shatai Southern Road, Baiyun District, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Haimiti Gulizeba
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Hong Liu
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xiaoxian Sima
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Ting Zhou
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Yan Huang
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
26
|
Kato K, Noda T, Kobayashi S, Sasaki K, Iwagami Y, Yamada D, Tomimaru Y, Takahashi H, Uemura M, Asaoka T, Shimizu J, Doki Y, Eguchi H. KLK10 derived from tumor endothelial cells accelerates colon cancer cell proliferation and hematogenous liver metastasis formation. Cancer Sci 2024; 115:1520-1535. [PMID: 38475666 PMCID: PMC11093189 DOI: 10.1111/cas.16144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/06/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Tumor endothelial cells (TECs), which are thought to be structurally and functionally different from normal endothelial cells (NECs), are increasingly attracting attention as a therapeutic target in hypervascular malignancies. Although colorectal liver metastasis (CRLM) tumors are hypovascular, inhibitors of angiogenesis are a key drug in multidisciplinary therapy, and TECs might be involved in the development and progression of cancer. Here, we analyzed the function of TEC in the CRLM tumor microenvironment. We used a murine colon cancer cell line (CT26) and isolated TECs from CRLM tumors. TECs showed higher proliferation and migration than NECs. Coinjection of CT26 and TECs yielded rapid tumor formation in vivo. Immunofluorescence analysis showed that coinjection of CT26 and TECs increased vessel formation and Ki-67+ cells. Transcriptome analysis identified kallikrein-related peptide 10 (KLK10) as a candidate target. Coinjection of CT26 and TECs after KLK10 downregulation with siRNA suppressed tumor formation in vivo. TEC secretion of KLK10 decreased after KLK10 downregulation, and conditioned medium after KLK10 knockdown in TECs suppressed CT26 proliferative activity. Double immunofluorescence staining of KLK10 and CD31 in CRLM tissues revealed a significant correlation between poor prognosis and positive KLK10 expression in TECs and tumor cells. On multivariate analysis, KLK10 expression was an independent prognostic factor in disease-free survival. In conclusion, KLK10 derived from TECs accelerates colon cancer cell proliferation and hematogenous liver metastasis formation. KLK10 in TECs might offer a promising therapeutic target in CRLM.
Collapse
Affiliation(s)
- Kazuya Kato
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Takehiro Noda
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Kazuki Sasaki
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Yoshifumi Iwagami
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Yoshito Tomimaru
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Mamoru Uemura
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Tadafumi Asaoka
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Junzo Shimizu
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| |
Collapse
|
27
|
Bian F, Goda C, Wang G, Lan YW, Deng Z, Gao W, Acharya A, Reza AA, Gomez-Arroyo J, Merjaneh N, Ren X, Goveia J, Carmeliet P, Kalinichenko VV, Kalin TV. FOXF1 promotes tumor vessel normalization and prevents lung cancer progression through FZD4. EMBO Mol Med 2024; 16:1063-1090. [PMID: 38589650 PMCID: PMC11099127 DOI: 10.1038/s44321-024-00064-8] [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: 07/24/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/10/2024] Open
Abstract
Cancer cells re-program normal lung endothelial cells (EC) into tumor-associated endothelial cells (TEC) that form leaky vessels supporting carcinogenesis. Transcriptional regulators that control the reprogramming of EC into TEC are poorly understood. We identified Forkhead box F1 (FOXF1) as a critical regulator of EC-to-TEC transition. FOXF1 was highly expressed in normal lung vasculature but was decreased in TEC within non-small cell lung cancers (NSCLC). Low FOXF1 correlated with poor overall survival of NSCLC patients. In mice, endothelial-specific deletion of FOXF1 decreased pericyte coverage, increased vessel permeability and hypoxia, and promoted lung tumor growth and metastasis. Endothelial-specific overexpression of FOXF1 normalized tumor vessels and inhibited the progression of lung cancer. FOXF1 deficiency decreased Wnt/β-catenin signaling in TECs through direct transcriptional activation of Fzd4. Restoring FZD4 expression in FOXF1-deficient TECs through endothelial-specific nanoparticle delivery of Fzd4 cDNA rescued Wnt/β-catenin signaling in TECs, normalized tumor vessels and inhibited the progression of lung cancer. Altogether, FOXF1 increases tumor vessel stability, and inhibits lung cancer progression by stimulating FZD4/Wnt/β-catenin signaling in TECs. Nanoparticle delivery of FZD4 cDNA has promise for future therapies in NSCLC.
Collapse
Affiliation(s)
- Fenghua Bian
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Chinmayee Goda
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Guolun Wang
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Ying-Wei Lan
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Zicheng Deng
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Wen Gao
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Anusha Acharya
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Abid A Reza
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Jose Gomez-Arroyo
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Nawal Merjaneh
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, 1919 E Thomas Rd., Phoenix, AZ, 85016, USA
| | - Xiaomeng Ren
- Division of Asthma Research of Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Jermaine Goveia
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, Leuven, 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, Leuven, 3000, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Vladimir V Kalinichenko
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
- Division of Neonatology, Phoenix Children's Hospital, 1919 E Thomas Rd., Phoenix, AZ, 85016, USA
| | - Tanya V Kalin
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA.
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, 1919 E Thomas Rd., Phoenix, AZ, 85016, USA.
- Department of Internal Medicine, Division of Pulmonary and Critical Care, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA.
| |
Collapse
|
28
|
Zhang T, Tai Z, Miao F, Zhang X, Li J, Zhu Q, Wei H, Chen Z. Adoptive cell therapy for solid tumors beyond CAR-T: Current challenges and emerging therapeutic advances. J Control Release 2024; 368:372-396. [PMID: 38408567 DOI: 10.1016/j.jconrel.2024.02.033] [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: 12/14/2023] [Revised: 02/05/2024] [Accepted: 02/23/2024] [Indexed: 02/28/2024]
Abstract
Adoptive cellular immunotherapy using immune cells expressing chimeric antigen receptors (CARs) is a highly specific anti-tumor immunotherapy that has shown promise in the treatment of hematological malignancies. However, there has been a slow progress toward the treatment of solid tumors owing to the complex tumor microenvironment that affects the localization and killing ability of the CAR cells. Solid tumors with a strong immunosuppressive microenvironment and complex vascular system are unaffected by CAR cell infiltration and attack. To improve their efficacy toward solid tumors, CAR cells have been modified and upgraded by "decorating" and "pruning". This review focuses on the structure and function of CARs, the immune cells that can be engineered by CARs and the transformation strategies to overcome solid tumors, with a view to broadening ideas for the better application of CAR cell therapy for the treatment of solid tumors.
Collapse
Affiliation(s)
- Tingrui Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China; Medical Guarantee Center, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China; School of Medicine, Shanghai University, Shanghai 200444, China; Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai 200443, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China; Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai 200443, China; Department of Pharmacy, First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Fengze Miao
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China; Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai 200443, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China; Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai 200443, China
| | - Jiadong Li
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China; Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai 200443, China
| | - Hua Wei
- Medical Guarantee Center, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China; School of Medicine, Shanghai University, Shanghai 200444, China; Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai 200443, China.
| |
Collapse
|
29
|
Wang T, Li X, Ma R, Sun J, Huang S, Sun Z, Wang M. Advancements in colorectal cancer research: Unveiling the cellular and molecular mechanisms of neddylation (Review). Int J Oncol 2024; 64:39. [PMID: 38391033 PMCID: PMC10919758 DOI: 10.3892/ijo.2024.5627] [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: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Neddylation, akin to ubiquitination, represents a post‑translational modification of proteins wherein neural precursor cell‑expressed developmentally downregulated protein 8 (NEDD8) is modified on the substrate protein through a series of reactions. Neddylation plays a pivotal role in the growth and proliferation of animal cells. In colorectal cancer (CRC), it predominantly contributes to the proliferation, metastasis and survival of tumor cells, decreasing overall patient survival. The strategic manipulation of the NEDD8‑mediated neddylation pathway holds immense therapeutic promise in terms of the potential to modulate the growth of tumors by regulating diverse biological responses within cancer cells, such as DNA damage response and apoptosis, among others. MLN4924 is an inhibitor of NEDD8, and its combined use with platinum drugs and irinotecan, as well as cycle inhibitors and NEDD activating enzyme inhibitors screened by drug repurposing, has been found to exert promising antitumor effects. The present review summarizes the recent progress made in the understanding of the role of NEDD8 in the advancement of CRC, suggesting that NEDD8 is a promising anti‑CRC target.
Collapse
Affiliation(s)
- Tianyu Wang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| | - Xiaobing Li
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| | - Ruijie Ma
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Jian Sun
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, P.R. China
| | - Shuhong Huang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
- Science and Technology Innovation Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| | - Zhigang Sun
- Department of Thoracic Surgery, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, P.R. China
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, P.R. China
| | - Meng Wang
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
30
|
Wu H, Geng Q, Shi W, Qiu C. Comprehensive pan-cancer analysis reveals CCDC58 as a carcinogenic factor related to immune infiltration. Apoptosis 2024; 29:536-555. [PMID: 38066393 DOI: 10.1007/s10495-023-01919-0] [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] [Accepted: 11/06/2023] [Indexed: 02/18/2024]
Abstract
CCDC58, a member of the CCDC protein family, has been primarily associated with the malignant progression of hepatocellular carcinoma (HCC) and breast cancer, with limited research conducted on its involvement in other tumor types. We aimed to assess the significance of CCDC58 in pan-cancer. We utilized the TCGA, GTEx, and UALCAN databases to perform the differential expression of CCDC58 at both mRNA and protein levels. Prognostic value was evaluated through univariate Cox regression and Kaplan-Meier methods. Mutation and methylation analyses were conducted using the cBioPortal and SMART databases. We identified genes interacting with and correlated to CCDC58 through STRING and GEPIA2, respectively. Subsequently, we performed GO and KEGG enrichment analyses. To gain insights into the functional status of CCDC58 at the single-cell level, we utilized CancerSEA. We explored the correlation between CCDC58 and immune infiltration as well as immunotherapy using the ESTIMATE package, TIMER2.0, TISIDB, TIDE, TIMSO, and TCIA. We examined the relationship between CCDC58 and tumor heterogeneity, stemness, DNA methyltransferases, and MMR genes. Lastly, we constructed a nomogram based on CCDC58 in HCC and investigated its association with drug sensitivity. CCDC58 expression was significantly upregulated and correlated with poor prognosis across various tumor types. The mutation frequency of CCDC58 was found to be increased in 25 tumors. We observed a negative correlation between CCDC58 expression and the methylation sites in the majority of tumors. CCDC58 showed negative correlations with immune and stromal scores, as well as with NK T cells, Tregs, CAFs, endothelial cells, and immunomodulators. Its value in immunotherapy was comparable to that of tumor mutational burden. CCDC58 exhibited positive correlations with tumor heterogeneity, stemness, DNA methyltransferase genes, and MMR genes. In HCC, CCDC58 was identified as an independent risk factor and demonstrated potential associations with multiple drugs. CCDC58 demonstrates significant clinical value as a prognostic marker and indicator of immune response across various tumor types. Its comprehensive analysis provides insights into its potential implications in pan-cancer research.
Collapse
Affiliation(s)
- Huili Wu
- Department of Endodontics, Zhonglou Hospital, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| | - Qing Geng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Wenxiang Shi
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chenjie Qiu
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China.
| |
Collapse
|
31
|
Yang Z, Zhang X, Bai X, Xi X, Liu W, Zhong W. Anti-angiogenesis in colorectal cancer therapy. Cancer Sci 2024; 115:734-751. [PMID: 38233340 PMCID: PMC10921012 DOI: 10.1111/cas.16063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/16/2023] [Accepted: 12/16/2023] [Indexed: 01/19/2024] Open
Abstract
The morbidity of colorectal cancer (CRC) has risen to third place among malignant tumors worldwide. In addition, CRC is a common cancer in China whose incidence increases annually. Angiogenesis plays an important role in the development of tumors because it can bring the nutrients that cancer cells need and take away metabolic waste. Various mechanisms are involved in the formation of neovascularization, and vascular endothelial growth factor is a key mediator. Meanwhile, angiogenesis inhibitors and drug resistance (DR) are challenges to consider when formulating treatment strategies for patients with different conditions. Thus, this review will discuss the molecules, signaling pathways, microenvironment, treatment, and DR of angiogenesis in CRC.
Collapse
Affiliation(s)
- Zhenni Yang
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
- Department of Gastroenterology and HepatologyXing'an League People's HospitalXing'an LeagueChina
| | - Xuqian Zhang
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
- Department of Gastroenterology and HepatologyChina Aerospace Science and Industry CorporationBeijingChina
| | - Xiaozhe Bai
- Department of Gastroenterology and HepatologyXing'an League People's HospitalXing'an LeagueChina
| | - Xiaonan Xi
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| | - Wentian Liu
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
| | - Weilong Zhong
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
| |
Collapse
|
32
|
Fu Y, Sun S, Shi D, Bi J. Construction of endothelial cell signatures for predicting the diagnosis, prognosis and immunotherapy response of bladder cancer via machine learning. J Cell Mol Med 2024; 28:e18155. [PMID: 38429911 PMCID: PMC10907833 DOI: 10.1111/jcmm.18155] [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: 05/11/2023] [Revised: 01/08/2024] [Accepted: 01/16/2024] [Indexed: 03/03/2024] Open
Abstract
We subtyped bladder cancer (BC) patients based on the expression patterns of endothelial cell (EC) -related genes and constructed a diagnostic signature and an endothelial cell prognostic index (ECPI), which are useful for diagnosing BC patients, predicting the prognosis of BC and evaluating drug sensitivity. Differentially expressed genes in ECs were obtained from the Tumour Immune Single-Cell Hub database. Subsequently, a diagnostic signature, a tumour subtyping system and an ECPI were constructed using data from The Cancer Genome Atlas and Gene Expression Omnibus. Associations between the ECPI and the tumour microenvironment, drug sensitivity and biofunctions were assessed. The hub genes in the ECPI were identified as drug candidates by molecular docking. Subtype identification indicated that high EC levels were associated with a worse prognosis and immunosuppressive effect. The diagnostic signature and ECPI were used to effectively diagnose BC and accurately assess the prognosis of BC and drug sensitivity among patients. Three hub genes in the ECPI were extracted, and the three genes had the closest affinity for doxorubicin and curcumin. There was a close relationship between EC and BC. EC-related genes can help clinicians diagnose BC, predict the prognosis of BC and select effective drugs.
Collapse
Affiliation(s)
- Yang Fu
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Shanshan Sun
- Department of PharmacyThe People's Hospital of Liaoning ProvinceShenyangLiaoningChina
| | - Du Shi
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jianbin Bi
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
33
|
Kabil MF, Badary OA, Bier F, Mousa SA, El-Sherbiny IM. A comprehensive review on lipid nanocarrier systems for cancer treatment: fabrication, future prospects and clinical trials. J Liposome Res 2024; 34:135-177. [PMID: 37144339 DOI: 10.1080/08982104.2023.2204372] [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: 05/03/2022] [Accepted: 04/02/2023] [Indexed: 05/06/2023]
Abstract
Over the last few decades, cancer has been considered a clinical challenge, being among the leading causes of mortality all over the world. Although many treatment approaches have been developed for cancer, chemotherapy is still the most utilized in the clinical setting. However, the available chemotherapeutics-based treatments have several caveats including their lack of specificity, adverse effects as well as cancer relapse and metastasis which mainly explains the low survival rate of patients. Lipid nanoparticles (LNPs) have been utilized as promising nanocarrier systems for chemotherapeutics to overcome the challenges of the currently applied therapeutic strategies for cancer treatment. Loading chemotherapeutic agent(s) into LNPs improves drug delivery at different aspects including specific targeting of tumours, and enhancing the bioavailability of drugs at the tumour site through selective release of their payload, thus reducing their undesired side effects on healthy cells. This review article delineates an overview of the clinical challenges in many cancer treatments as well as depicts the role of LNPs in achieving optimal therapeutic outcomes. Moreover, the review contains a comprehensive description of the many LNPs categories used as nanocarriers in cancer treatment to date, as well as the potential of LNPs for future applications in other areas of medicine and research.
Collapse
Affiliation(s)
- Mohamed Fawzi Kabil
- Nanomedicine Research Labs, Center for Materials Science (CMS), Zewail City of Science and Technology, Giza, Egypt
| | - Osama A Badary
- Clinical Pharmacy Department, Faculty of Pharmacy, The British University in Egypt, El-Shorouk City, Egypt
| | - Frank Bier
- AG Molekulare Bioanalytik und Bioelektronik, Institut für Biochemie und Biologie, Universität Potsdam Karl-Liebknecht-Straße 24/25, Potsdam (OT Golm), Germany
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Science (CMS), Zewail City of Science and Technology, Giza, Egypt
| |
Collapse
|
34
|
Ni Z, Cong S, Li H, Liu J, Zhang Q, Wei C, Pan G, He H, Liu W, Mao A. Integration of scRNA and bulk RNA-sequence to construct the 5-gene molecular prognostic model based on the heterogeneity of thyroid carcinoma endothelial cell. Acta Biochim Biophys Sin (Shanghai) 2024; 56:255-269. [PMID: 38186223 PMCID: PMC10984871 DOI: 10.3724/abbs.2023254] [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: 03/30/2023] [Accepted: 09/22/2023] [Indexed: 01/09/2024] Open
Abstract
Thyroid cancer (TC) is a kind of cancer with high heterogeneity, which leads to significant difference in prognosis. The prognostic molecular processes are not well understood. Cancer cells and tumor microenvironment (TME) cells jointly determine the heterogeneity. However, quite a little attention was paid to cells in the TME in the past years. In this study, we not only reveal that endothelial cells (ECs) are strongly associated with the progress of papillary thyroid cancer (PTC) using single-cell RNA-seq (scRNA-seq) data downloaded from Gene Expression Omnibus (GEO) and WGCNA, but also screen 5 crucial genes of ECs: CLDN5, ABCG2, NOTCH4, PLAT, and TMEM47. Furthermore, the 5-gene molecular prognostic model is constructed, which can predict how well a patient will do on PD-L1 blockade immunotherapy for TC and evaluate prognosis. Quantitative real-time polymerase chain reaction (qRT-PCR) analysis demonstrates that PLAT is decreased in TC and the increase of PLAT can restrain the migratory capacity of TC cells. Meanwhile, in TC cells, PLAT suppresses VEGFa/VEGFR2-mediated human umbilical vascular endothelial cell (HUVEC) proliferation and tube formation. Totally, we construct the 5-gene molecular prognostic model from the perspective of EC and provide a new idea for immunotherapy of TC.
Collapse
Affiliation(s)
- Zhaoxian Ni
- Department of General SurgeryMinhang HospitalFudan UniversityShanghai201199China
- Department of Head and Neck SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Shan Cong
- Department of Laparoscopic Surgerythe First Affiliated Hospital of Dalian Medical UniversityDalian116000China
| | - Hongchang Li
- Department of General SurgeryMinhang HospitalFudan UniversityShanghai201199China
| | - Jiazhe Liu
- Department of General SurgeryMinhang HospitalFudan UniversityShanghai201199China
| | - Qing Zhang
- Department of General SurgeryMinhang HospitalFudan UniversityShanghai201199China
| | - Chuanchao Wei
- Department of General SurgeryMinhang HospitalFudan UniversityShanghai201199China
| | - Gaofeng Pan
- Department of General SurgeryMinhang HospitalFudan UniversityShanghai201199China
| | - Hui He
- Department of Head and Neck SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Department of Laparoscopic Surgerythe First Affiliated Hospital of Dalian Medical UniversityDalian116000China
| | - Weiyan Liu
- Department of General SurgeryMinhang HospitalFudan UniversityShanghai201199China
| | - Anwei Mao
- Department of General SurgeryMinhang HospitalFudan UniversityShanghai201199China
| |
Collapse
|
35
|
Xie P, Tan SY, Li HF, Tang HD, Zhou JH. Transcriptomic landscape of endothelial cells: Key tumor microenvironment components indicating variable clinical outcomes in pancreatic ductal adenocarcinoma. ENVIRONMENTAL TOXICOLOGY 2024; 39:572-582. [PMID: 37449672 DOI: 10.1002/tox.23881] [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: 05/27/2023] [Revised: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023]
Abstract
Endothelial cells (ECs) present in the tumor microenvironment (TME) exhibit significant diversity that may impact the efficacy of anti-tumor treatments. Thus, our study sought to elucidate the various clusters of ECs present in pancreatic ductal adenocarcinoma (PDAC) and explore their possible interactions and influence on clinical outcomes. We obtained single-cell transcriptome data from 24 PDAC tumors and 11 normal pancreases, minimizing any batch effects between samples. Next, we compared the relative abundance of various ECs clusters across distinct sample types. Pseudo-time analysis was employed to investigate the differentiation origin of ECs. A variety of bioinformatics methods were used to investigate potential communication between ECs and malignant cells, as well as assess metabolic changes, pathway alterations, and immune-related markers expression within distinct EC clusters. Lastly, we investigated the impact of particular ECs clusters on patient prognosis in bulk transcriptome data. Our study identified seven distinct clusters of ECs, denoted as CA4+ ECs, MMP2+ ECs, SPP1+ ECs, MT1F+ ECs, CCL5+ ECs, RGS5+ ECs, and TYROBP+ ECs. Pseudo-time analysis suggested that the loss of CA4+ ECs and MT1F+ ECs may promote malignant progression. Cell communication elucidated that MT1F+ ECs exhibited the strongest outgoing interaction strength, whereas RGS5+ ECs displayed the strongest incoming interaction strength. Furthermore, TYROBP+ ECs exhibited greater metabolic activity, and notably, CCL5+ ECs displayed increased expression of immune-related molecules. Lastly, across cohorts of bulk transcriptome levels, CA4+ ECs, MT1F+ ECs, and RGS5+ ECs consistently demonstrated prognostic indicative effects. PDAC patients exhibit the presence of seven distinct EC clusters, each demonstrating significant metabolic and immunological heterogeneity. Targeted therapeutic approaches directed toward CA4+ ECs and MT1F+ ECs may prove advantageous in addressing challenges associated with PDAC treatment. Additionally, variations in the relative abundance of CA4+ ECs, MT1F+ ECs, and RGS5+ ECs were indicated as predictive of patient prognosis.
Collapse
Affiliation(s)
- Peng Xie
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
| | - Si-Yuan Tan
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
| | - Hai-Feng Li
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
| | - Hao-Dong Tang
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
| | - Jia-Hua Zhou
- Department of Surgery, School of Medicine, Southeast University, Nanjing, China
- Department of Hepato-Pancreatico-Biliary Surgery, Zhongda Hospital Southeast University, Nanjing, China
| |
Collapse
|
36
|
Wendong Y, Jiali J, Qiaomei F, Yayun W, Xianze X, Zheng S, Wei H. Biomechanical forces and force-triggered drug delivery in tumor neovascularization. Biomed Pharmacother 2024; 171:116117. [PMID: 38171243 DOI: 10.1016/j.biopha.2023.116117] [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: 11/02/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024] Open
Abstract
Tumor angiogenesis is one of the typical hallmarks of tumor occurrence and development, and tumor neovascularization also exhibits distinct characteristics from normal blood vessels. As the number of cells and matrix inside the tumor increases, the biomechanical force is enhanced, specifically manifested as solid stress, fluid stress, stiffness, and topology. This mechanical microenvironment also provides shelter for tumors and intensifies angiogenesis, providing oxygen and nutritional support for tumor progression. During tumor development, the biomechanical microenvironment also emerges, which in turn feeds back to regulate the tumor progression, including tumor angiogenesis, and biochemical and biomechanical signals can regulate tumor angiogenesis. Blood vessels possess inherent sensitivity to mechanical stimuli, but compared to the extensive research on biochemical signal regulation, the study of the regulation of tumor neovascularization by biomechanical signals remains relatively scarce. Biomechanical forces can affect the phenotypic characteristics and mechanical signaling pathways of tumor blood vessels, directly regulating angiogenesis. Meanwhile, they can indirectly regulate tumor angiogenesis by causing an imbalance in angiogenesis signals and affecting stromal cell function. Understanding the regulatory mechanism of biomechanical forces in tumor angiogenesis is beneficial for better identifying and even taming the mechanical forces involved in angiogenesis, providing new therapeutic targets for tumor vascular normalization. Therefore, we summarized the composition of biomechanical forces and their direct or indirect regulation of tumor neovascularization. In addition, this review discussed the use of biomechanical forces in combination with anti-angiogenic therapies for the treatment of tumors, and biomechanical forces triggered delivery systems.
Collapse
Affiliation(s)
- Yao Wendong
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Jiang Jiali
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Fan Qiaomei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Weng Yayun
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Xie Xianze
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Shi Zheng
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| | - Huang Wei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| |
Collapse
|
37
|
Yu L, Hong Y, Maishi N, Matsuda AY, Hida Y, Hasebe A, Kitagawa Y, Hida K. Oral bacterium Streptococcus mutans promotes tumor metastasis through thrombosis formation. Cancer Sci 2024; 115:648-659. [PMID: 38096871 PMCID: PMC10859626 DOI: 10.1111/cas.16010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/10/2023] [Accepted: 10/25/2023] [Indexed: 02/13/2024] Open
Abstract
Thrombosis is a well-known cardiovascular disease (CVD) complication that has caused death in many patients with cancer. Oral bacteria have been reported to contribute to systemic diseases, including CVDs, and tumor metastasis. However, whether oral bacteria-induced thrombosis induces tumor metastasis remains poorly understood. In this study, the cariogenic oral bacterium Streptococcus mutans was used to examine thrombosis in vitro and in vivo. Investigation of tumor metastasis to the lungs was undertaken by intravenous S. mutans implantation using a murine breast cancer metastasis model. The results indicated that platelet activation, aggregation, and coagulation were significantly altered in S. mutans-stimulated endothelial cells (ECs), with elevated neutrophil migration, thereby inducing thrombosis formation. Streptococcus mutans stimulation significantly enhances platelet and tumor cell adhesion to the inflamed ECs. Furthermore, S. mutans-induced pulmonary thrombosis promotes breast cancer cell metastasis to the lungs in vivo, which can be reduced by using aspirin, an antiplatelet drug. Our findings indicate that oral bacteria promote tumor metastasis through thrombosis formation. Oral health management is important to prevent CVDs, tumor metastasis, and their associated death.
Collapse
Affiliation(s)
- Li Yu
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Yuying Hong
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
- Oral Diagnosis and Medicine, Faculty of Dental Medicine and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Nako Maishi
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Aya Yanagawa Matsuda
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Yasuhiro Hida
- Advanced Robotic and Endoscopic Surgery, School of MedicineFujita Health UniversityToyoakeJapan
| | - Akira Hasebe
- Oral Molecular Microbiology, Faculty of Dental Medicine and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Yoshimasa Kitagawa
- Oral Diagnosis and Medicine, Faculty of Dental Medicine and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Kyoko Hida
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| |
Collapse
|
38
|
Liu H, Tang L, Li Y, Xie W, Zhang L, Tang H, Xiao T, Yang H, Gu W, Wang H, Chen P. Nasopharyngeal carcinoma: current views on the tumor microenvironment's impact on drug resistance and clinical outcomes. Mol Cancer 2024; 23:20. [PMID: 38254110 PMCID: PMC10802008 DOI: 10.1186/s12943-023-01928-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
The incidence of nasopharyngeal carcinoma (NPC) exhibits significant variations across different ethnic groups and geographical regions, with Southeast Asia and North Africa being endemic areas. Of note, Epstein-Barr virus (EBV) infection is closely associated with almost all of the undifferentiated NPC cases. Over the past three decades, radiation therapy and chemotherapy have formed the cornerstone of NPC treatment. However, recent advancements in immunotherapy have introduced a range of promising approaches for managing NPC. In light of these developments, it has become evident that a deeper understanding of the tumor microenvironment (TME) is crucial. The TME serves a dual function, acting as a promoter of tumorigenesis while also orchestrating immunosuppression, thereby facilitating cancer progression and enabling immune evasion. Consequently, a comprehensive comprehension of the TME and its intricate involvement in the initiation, progression, and metastasis of NPC is imperative for the development of effective anticancer drugs. Moreover, given the complexity of TME and the inter-patient heterogeneity, personalized treatment should be designed to maximize therapeutic efficacy and circumvent drug resistance. This review aims to provide an in-depth exploration of the TME within the context of EBV-induced NPC, with a particular emphasis on its pivotal role in regulating intercellular communication and shaping treatment responses. Additionally, the review offers a concise summary of drug resistance mechanisms and potential strategies for their reversal, specifically in relation to chemoradiation therapy, targeted therapy, and immunotherapy. Furthermore, recent advances in clinical trials pertaining to NPC are also discussed.
Collapse
Affiliation(s)
- Huai Liu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Ling Tang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yanxian Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Wenji Xie
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Ling Zhang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Tengfei Xiao
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Hongmin Yang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Wangning Gu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Hui Wang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
| | - Pan Chen
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
39
|
Xie T, Fu DJ, Li KJ, Guo JD, Xiao ZM, Li Z, Zhao SC. Identification of a basement membrane gene signature for predicting prognosis and estimating the tumor immune microenvironment in prostate cancer. Aging (Albany NY) 2024; 16:1581-1604. [PMID: 38240702 PMCID: PMC10866409 DOI: 10.18632/aging.205445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/01/2023] [Indexed: 02/06/2024]
Abstract
Basement membrane plays an important role in tumor invasion and metastasis, which is closely related to prognosis. However, the prognostic value and biology of basement membrane genes (BMGs) in prostate cancer (PCa) remain unknown. In the TCGA training set, we used differentially expressed gene analysis, protein-protein interaction networks, univariate and multivariate Cox regression, and least absolute shrinkage and selection operator regression to construct a basement membrane-related risk model (BMRM) and validated its effectiveness in the MSKCC validation set. Furthermore, the accurate nomogram was constructed to improve clinical applicability. Patients with PCa were divided into high-risk and low-risk groups according to the optimal cut-off value of the basement membrane-related risk score (BMRS). It was found that BMRS was significantly associated with RFS, T-stage, Gleason score, and tumor microenvironmental characteristics in PCa patients. Further analysis showed that the model grouping was closely related to tumor immune microenvironment characteristics, immune checkpoint inhibitors, and chemotherapeutic drug sensitivity. In this study, we developed a new BMGs-based prognostic model to determine the prognostic value of BMGs in PCa. Finally, we confirmed that THBS2, a key gene in BMRM, may be an important link in the occurrence and progression of PCa. This study provides a novel perspective to assess the prognosis of PCa patients and provides clues for the selection of future personalized treatment regimens.
Collapse
Affiliation(s)
- Tao Xie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510500, China
| | - Du-Jiang Fu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kang-Jing Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jia-Ding Guo
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510500, China
| | - Zhao-Ming Xiao
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510500, China
| | - Zhijie Li
- Department of Geriatric Medicine, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Shan-Chao Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510500, China
| |
Collapse
|
40
|
Su Z, Li W, Lei Z, Hu L, Wang S, Guo L. Regulation of Angiogenesis by Non-Coding RNAs in Cancer. Biomolecules 2024; 14:60. [PMID: 38254660 PMCID: PMC10813527 DOI: 10.3390/biom14010060] [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: 10/31/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Non-coding RNAs, including microRNAs, long non-coding RNAs, and circular RNAs, have been identified as crucial regulators of various biological processes through epigenetic regulation, transcriptional regulation, and post-transcriptional regulation. Growing evidence suggests that dysregulation and activation of non-coding RNAs are closely associated with tumor angiogenesis, a process essential for tumor growth and metastasis and a major contributor to cancer-related mortality. Therefore, understanding the molecular mechanisms underlying tumor angiogenesis is of utmost importance. Numerous studies have documented the involvement of different types of non-coding RNAs in the regulation of angiogenesis. This review provides an overview of how non-coding RNAs regulate tumor angiogenesis. Additionally, we discuss emerging strategies that exploit non-coding RNAs for anti-angiogenic therapy in cancer treatment. Ultimately, this review underscores the crucial role played by non-coding RNAs in tumor angiogenesis and highlights their potential as therapeutic targets for anti-angiogenic interventions against cancer.
Collapse
Affiliation(s)
- Zhiyue Su
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wenshu Li
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Zhe Lei
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Shengjie Wang
- Department of Basic Medicine, Kangda College, Nanjing Medical University, Lianyungang 222000, China
| | - Lingchuan Guo
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
41
|
Yang C, Cheng X, Gao S, Pan Q. Integrating bulk and single-cell data to predict the prognosis and identify the immune landscape in HNSCC. J Cell Mol Med 2024; 28:e18009. [PMID: 37882107 PMCID: PMC10805493 DOI: 10.1111/jcmm.18009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/20/2023] [Accepted: 10/13/2023] [Indexed: 10/27/2023] Open
Abstract
The complex interplay between tumour cells and the tumour microenvironment (TME) underscores the necessity for gaining comprehensive insights into disease progression. This study centres on elucidating the elusive the elusive role of endothelial cells within the TME of head and neck squamous cell carcinoma (HNSCC). Despite their crucial involvement in angiogenesis and vascular function, the mechanistic diversity of endothelial cells among HNSCC patients remains largely uncharted. Leveraging advanced single-cell RNA sequencing (scRNA-Seq) technology and the Scissor algorithm, we aimed to bridge this knowledge gap and illuminate the intricate interplay between endothelial cells and patient prognosis within the context of HNSCC. Here, endothelial cells were categorized into Scissorhigh and Scissorlow subtypes. We identified Scissor+ endothelial cells exhibiting pro-tumorigenic profiles and constructed a prognostic risk model for HNSCC. Additionally, four biomarkers also were identified by analysing the gene expression profiles of patients with HNSCC and a prognostic risk prediction model was constructed based on these genes. Furthermore, the correlations between endothelial cells and prognosis of patients with HNSCC were analysed by integrating bulk and single-cell sequencing data, revealing a close association between SHSS and the overall survival (OS) of HNSCC patients with malignant endothelial cells. Finally, we validated the prognostic model by RT-qPCR and IHC analysis. These findings enhance our comprehension of TME heterogeneity at the single-cell level and provide a prognostic model for HNSCC.
Collapse
Affiliation(s)
- Chunlong Yang
- Clinical Research CenterAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Xiaoning Cheng
- Zhanjiang Central HospitalGuangdong Medical UniversityZhanjiangChina
| | - Shenglan Gao
- Clinical Research CenterAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Qingjun Pan
- Clinical Research CenterAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| |
Collapse
|
42
|
Yang Y, Wang Q, Zhan F. Unraveling the Action Mechanism of Tubeimoside-1 against Tumor Microvessels via Network Pharmacology and Experimental Validation. J Cancer 2024; 15:955-965. [PMID: 38230220 PMCID: PMC10788730 DOI: 10.7150/jca.90391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/27/2023] [Indexed: 01/18/2024] Open
Abstract
Objective: Tubeimoside-1 (TBMS1) is a plant-derived triterpenoid saponin that exhibits pharmacological properties and anti-tumor effects, but the anti-tumor microvessels of action of TBMS1 remains to be completely elucidated. This study aims to verify the effect of TBMS1 on tumor microvessels and its underlying mechanism. Methods: A SKOV3 xenografted mouse model were constructed to evaluate the anti-tumor microvessels of TBMS1 in vivo, followed by function assays to verify the effects of TBMS1 on the proliferation, cell cycle, migration, and tubule formation of vascular endothelial cells in vitro. Next, based on network pharmacology, the drug/disease-target protein-protein interaction (PPI) networks, biological functions and gene enrichment analyses were performed to predict the underlying mechanism. Finally, molecules and pathways associated with tumor trans-endothelial migration were identified. Results: TBMS1 treatment effectively reduced tumor microvessel density in ovarian cancer model and inhibited the proliferation, cell cycle, migration, and induced apoptosis of vascular endothelial cells in vitro. Network pharmacological data suggested that tumor cell adhesion and trans-endothelial migration may participate in antiangiogenic effects of TBMS1. By endothelial adhesion and permeability assay, we identified that tumor adhesion and the permeability of endothelial monolayers were reduced by TBMS1. Furthermore, adhesion protein (VCAM-1and ICAM-1) and tight junction (TJ) proteins (VE-cadhsion, ZO-1 and claudin-5) were found to be regulated. Finally, Akt, Erk1/2, Stat3 and NF-κB signaling were decreased by TBMS1 treatment. Conclusion: To sum up, our findings strongly suggest that clinical application of TBSM1 may serve as a vasoactive drug treatment to suppress tumor progression.
Collapse
Affiliation(s)
- YinRong Yang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University (Qingdao), Qingdao, Shandong 266035, China
| | - Qian Wang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University (Qingdao), Qingdao, Shandong 266035, China
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong 250012, China
| | - FengXia Zhan
- Department of Clinical Laboratory, Shandong University School Hospital, Jinan, Shandong, 250012, China
| |
Collapse
|
43
|
Yamamoto M, Omori T, Shinno N, Hara H, Mukai Y, Sugase T, Takeoka T, Kanemura T, Mikamori M, Hasegawa S, Akita H, Haraguchi N, Nishimura J, Wada H, Matsuda C, Yasui M, Miyata H, Ohue M. Prognostic Value of a Novel Index Combining the Prognostic Nutritional Index and D-Dimer Levels for Gastric Cancer after Gastrectomy. Oncology 2023; 103:1-10. [PMID: 38160660 PMCID: PMC11731837 DOI: 10.1159/000533150] [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: 03/27/2023] [Accepted: 07/09/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION The prognostic nutritional index (PNI) and D-dimer level are two useful measures for gastric cancer prognosis. As they each comprise different factors, it is possible to employ a more useful combined indicator. This study therefore aimed to establish a PNI-D score - which combines the PNI and D-dimer level - and validate its usefulness as a prognostic marker. METHODS We collected data from 1,218 patients with gastric cancer who had undergone radical gastrectomy (R0) between January 2004 and December 2015. Patients were divided into three PNI-D score groups based on the following criteria: score 2, low-PNI (≤46) and high D-dimer levels (>1.0 µg/mL); score 1, either low-PNI or high D-dimer levels; and score 0, no abnormality. We defined the PNI-D score as low (score 0 or 1) and high (score 2), respectively. RESULTS The PNI-D score was significantly associated with overall, recurrence-free, and disease-specific survival (all log-rank p < 0.0001). The 5-year overall survival rates of patients with PNI-D scores of low and high were 88.1% and 64.7%, respectively; their 5-year recurrence-free survival rates were 86.7% and 61.3%, respectively; and their 5-year disease-specific survival rates were 99.3% and 76.5%, respectively. Cox multivariate analysis revealed that a high-PNI-D score was an independent, statistically significant prognostic factor for poor overall (p = 0.01) survival in patients with gastric cancer. CONCLUSIONS The PNI-D is an independent prognostic factor for patients with gastric cancer.
Collapse
Affiliation(s)
- Masaaki Yamamoto
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Takeshi Omori
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Naoki Shinno
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Hisashi Hara
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Yosuke Mukai
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Takahito Sugase
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Tomohira Takeoka
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Takashi Kanemura
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Manabu Mikamori
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Shinichiro Hasegawa
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Hirofumi Akita
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Naotsugu Haraguchi
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Junichi Nishimura
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroshi Wada
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Chu Matsuda
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Masayoshi Yasui
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroshi Miyata
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Masayuki Ohue
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
44
|
Ciummo SL, Sorrentino C, Fieni C, Di Carlo E. Interleukin-30 subverts prostate cancer-endothelium crosstalk by fostering angiogenesis and activating immunoregulatory and oncogenic signaling pathways. J Exp Clin Cancer Res 2023; 42:336. [PMID: 38087324 PMCID: PMC10714661 DOI: 10.1186/s13046-023-02902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Cancer-endothelial interplay is crucial for tumor behavior, yet the molecular mechanisms involved are largely unknown. Interleukin(IL)-30, which is expressed as a membrane-anchored cytokine by human prostate cancer (PC) cells, promotes PC vascularization and progression, but the underlying mechanisms have yet to be fully explored. METHODS PC-endothelial cell (EC) interactions were investigated, after coculture, by flow cytometry, transcriptional profiling, western blot, and ELISA assays. Proteome profiler phospho-kinase array unveiled the molecular pathways involved. The role of tumor-derived IL30 on the endothelium's capacity to generate autocrine circuits and vascular budding was determined following IL30 overexpression, by gene transfection, or its deletion by CRISPR/Cas9 genome editing. Clinical value of the experimental findings was determined through immunopathological study of experimental and patient-derived PC samples, and bioinformatics of gene expression profiles from PC patients. RESULTS Contact with PC cells favors EC proliferation and production of angiogenic and angiocrine factors, which are boosted by PC expression of IL30, that feeds autocrine loops, mediated by IGF1, EDN1, ANG and CXCL10, and promotes vascular budding and inflammation, via phosphorylation of multiple signaling proteins, such as Src, Yes, STAT3, STAT6, RSK1/2, c-Jun, AKT and, primarily CREB, GSK-3α/β, HSP60 and p53. Deletion of the IL30 gene in PC cells inhibits endothelial expression of IGF1, EDN1, ANG and CXCL10 and substantially impairs tumor angiogenesis. In its interaction with IL30-overexpressing PC cells the endothelium boosts their expression of a wide range of immunity regulatory genes, including CCL28, CCL4, CCL5, CCR2, CCR7, CXCR4, IL10, IL13, IL17A, FASLG, IDO1, KITLG, TNFA, TNFSF10 and PDCD1, and cancer driver genes, including BCL2, CCND2, EGR3, IL6, VEGFA, KLK3, PTGS1, LGALS4, GNRH1 and SHBG. Immunopathological analyses of PC xenografts and in silico investigation of 1116 PC cases, from the Prostate Cancer Transcriptome Atlas, confirmed the correlation between the expression of IL30 and that of both pro-inflammatory genes, NOS2, TNFA, CXCR5 and IL12B, and cancer driver genes, LGALS4, GNRH1 and SHBG, which was validated in a cohort of 80 PC patients. CONCLUSIONS IL30 regulates the crosstalk between PC and EC and reshapes their transcriptional profiles, triggering angiogenic, immunoregulatory and oncogenic gene expression programs. These findings highlight the angiostatic and oncostatic efficacy of targeting IL30 to fight PC.
Collapse
Affiliation(s)
- Stefania Livia Ciummo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University" of Chieti-Pescara, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via L. Polacchi 11, 66100, Chieti, Italy
| | - Carlo Sorrentino
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University" of Chieti-Pescara, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via L. Polacchi 11, 66100, Chieti, Italy
| | - Cristiano Fieni
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University" of Chieti-Pescara, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via L. Polacchi 11, 66100, Chieti, Italy
| | - Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University" of Chieti-Pescara, Chieti, Italy.
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Via L. Polacchi 11, 66100, Chieti, Italy.
| |
Collapse
|
45
|
Zefferino R, Conese M. A Vaccine against Cancer: Can There Be a Possible Strategy to Face the Challenge? Possible Targets and Paradoxical Effects. Vaccines (Basel) 2023; 11:1701. [PMID: 38006033 PMCID: PMC10674257 DOI: 10.3390/vaccines11111701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/07/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
Is it possible to have an available vaccine that eradicates cancer? Starting from this question, this article tries to verify the state of the art, proposing a different approach to the issue. The variety of cancers and different and often unknown causes of cancer impede, except in some cited cases, the creation of a classical vaccine directed at the causative agent. The efforts of the scientific community are oriented toward stimulating the immune systems of patients, thereby preventing immune evasion, and heightening chemotherapeutic agents effects against cancer. However, the results are not decisive, because without any warning signs, metastasis often occurs. The purpose of this paper is to elaborate on a vaccine that must be administered to a patient in order to prevent metastasis; metastasis is an event that leads to death, and thus, preventing it could transform cancer into a chronic disease. We underline the fact that the field has not been studied in depth, and that the complexity of metastatic processes should not be underestimated. Then, with the aim of identifying the target of a cancer vaccine, we draw attention to the presence of the paradoxical actions of different mechanisms, pathways, molecules, and immune and non-immune cells characteristic of the tumor microenvironment at the primary site and pre-metastatic niche in order to exclude possible vaccine candidates that have opposite effects/behaviors; after a meticulous evaluation, we propose possible targets to develop a metastasis-targeting vaccine. We conclude that a change in the current concept of a cancer vaccine is needed, and the efforts of the scientific community should be redirected toward a metastasis-targeting vaccine, with the increasing hope of eradicating cancer.
Collapse
Affiliation(s)
- Roberto Zefferino
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| |
Collapse
|
46
|
Davodabadi F, Sajjadi SF, Sarhadi M, Mirghasemi S, Nadali Hezaveh M, Khosravi S, Kamali Andani M, Cordani M, Basiri M, Ghavami S. Cancer chemotherapy resistance: Mechanisms and recent breakthrough in targeted drug delivery. Eur J Pharmacol 2023; 958:176013. [PMID: 37633322 DOI: 10.1016/j.ejphar.2023.176013] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Conventional chemotherapy, one of the most widely used cancer treatment methods, has serious side effects, and usually results in cancer treatment failure. Drug resistance is one of the primary reasons for this failure. The most significant drawbacks of systemic chemotherapy are rapid clearance from the circulation, the drug's low concentration in the tumor site, and considerable adverse effects outside the tumor. Several ways have been developed to boost neoplasm treatment efficacy and overcome medication resistance. In recent years, targeted drug delivery has become an essential therapeutic application. As more mechanisms of tumor treatment resistance are discovered, nanoparticles (NPs) are designed to target these pathways. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation. Nano-drugs have been increasingly employed in medicine, incorporating therapeutic applications for more precise and effective tumor diagnosis, therapy, and targeting. Many benefits of NP-based drug delivery systems in cancer treatment have been proven, including good pharmacokinetics, tumor cell-specific targeting, decreased side effects, and lessened drug resistance. As more mechanisms of tumor treatment resistance are discovered, NPs are designed to target these pathways. At the moment, this innovative technology has the potential to bring fresh insights into cancer therapy. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shaghayegh Mirghasemi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Nadali Hezaveh
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Samin Khosravi
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Kamali Andani
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Saeid Ghavami
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555. Katowice, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada.
| |
Collapse
|
47
|
Qie Y, Sun X, Yang Y, Yan T. Emerging functions and applications of exosomes in oral squamous cell carcinoma. J Oral Pathol Med 2023; 52:886-894. [PMID: 37701945 DOI: 10.1111/jop.13479] [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: 07/30/2023] [Revised: 08/09/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023]
Abstract
Oral squamous cell carcinoma is the most common phenotype in pathology, which accounts for 80% of all oral cancers. The therapeutic methods of oral squamous cell carcinoma include surgical excision, chemotherapy, and radiotherapy. Whereas, the high recurrence rate and poor prognosis lead to a 5-year survival rate less than 50%. In order to explore more therapeutic strategies of oral squamous cell carcinoma, the relevant risk factors, mechanisms, and diagnostics are widely detected. The various exosome-mediated biological effects on the development of oral squamous cell carcinoma have drawn lots of attention. Exosomes, a kind of extracellular vesicles secreted from host cells and transferred to other cells, show great potential in the regulations of tumorigenesis, progression, and metastasis on oral squamous cell carcinoma. Moreover, some studies reported that the exosomes could interact with tumor microenvironment and be applied to diagnosis or therapy of oral squamous cell carcinoma. In this work, we will summarize the frontier studies of exosomes in the tumor growth, tumor-associated angiogenesis, invasion, and metastasis of oral squamous cell carcinoma, and then probe the current biological functions and applications of exosomes and exosome-derived materials for the therapeutic strategies of oral squamous cell carcinoma, which would help us to update the understanding of exosomes in oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Yingchun Qie
- Stomatology Department, Zibo First Hospital, Zibo, Shandong Province, China
| | - Xia Sun
- Stomatology Department, Yidu Central Hospital of Weifang, Qingzhou, Shandong Province, China
| | - Yongqiang Yang
- Stomatology Department, Yidu Central Hospital of Weifang, Qingzhou, Shandong Province, China
| | - Tao Yan
- Intensive Care Unit, Zibo First Hospital, Zibo, Shandong Province, China
| |
Collapse
|
48
|
Wang J, Liu Z, Lin L, Wu Z, Gao X, Cai X, Chang L, Xia X, Zhang H, Chen G. Collagen-related gene expression level predicts the prognosis and immune therapy response. Gastric Cancer 2023; 26:891-903. [PMID: 37543986 DOI: 10.1007/s10120-023-01416-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Gastric cancer patients responded differently to the same treatment strategy and had various prognoses for the lack of biomarkers to guide the therapy choice. METHODS RNA data of a local gastric cancer cohort with 103 patients were processed and used to explore potential treatment guiding factors. Cluster analysis was performed by non-negative matrix factorization. The expression level of collagen-related genes was evaluated by ssGSEA named collagen score (CS). Data from TCGA, ACRG, and an immune therapy cohort were utilized to explore prognosis and efficacy. Prognostic predictive power of CS was assessed using the nomogram. RESULTS In our study, local RNA data were processed by cluster analysis, and it was found that cluster 2 contained a worse tumor infiltration status. The GSEA result showed that collagen-related pathways were differentially activated in two clusters. In TCGA and ACRG cohorts, the CS can be used as an independent prognostic factor (TCGA OS: p = 0.018, HR = 3.5; ACRG OS: p = 0.014, HR = 4.88). An immunotherapy cohort showed that the patients with higher CS had a significantly worse ORR (p = 0.0025). The high CS group contained several cell death pathways down-regulated and contained the worse tumor microenvironment. The nomogram demonstrated the survival prediction capability of collagen score. CONCLUSION CS was verified as an independent prognostic factor and potentially reflected the therapeutic effect of immunotherapy. The CS could provide a new way to evaluate the clinical prognosis and response information helping develop the collagen-targeted treatment.
Collapse
Affiliation(s)
- Jianchao Wang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Zhentian Liu
- Department of Translational Medicine, Geneplus-Beijing Institute, Beijing, 102205, China
| | - Liyan Lin
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Zhida Wu
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Xuan Gao
- Geneplus-Shenzhen Clinical Laboratory, Shenzhen, 518122, China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiqian Cai
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Lianpeng Chang
- Department of Translational Medicine, Geneplus-Beijing Institute, Beijing, 102205, China
| | - Xuefeng Xia
- Department of Translational Medicine, Geneplus-Beijing Institute, Beijing, 102205, China
| | - Hejun Zhang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Gang Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China.
| |
Collapse
|
49
|
Zheng S, Liao J, Sun M, Liu R, Lv J. Extracellular shuttling miR-21 contributes to esophageal cancers and human umbilical vein endothelial cell communication in the tumor microenvironment and promotes tumor angiogenesis by targeting phosphatase and tensinhomolog. Thorac Cancer 2023; 14:3119-3132. [PMID: 37726969 PMCID: PMC10626251 DOI: 10.1111/1759-7714.15103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Cell-cell communication by carcinoma-derived exosomes can influence the tumor microenvironment (TME) and regulate cancer progression. Based on the overexpression of microRNA-21-5p (miR-21) in plasma from patients diagnosed with esophageal squamous cell carcinoma (ESCC) and exosomes from ESCC cell lines identified earlier, this study aimed to explore the influence of exosomal miR-21 within the TME. METHOD ScRNA-Seq and Bulk RNA-Seq were integrated to elucidate the communication between cancer and endothelial cells. The functionality and mechanisms by which exo-miR-21 derived from carcinoma regulate endothelial cell-mediated angiogenesis were assessed using a cocultivation model of EC9706 cells and recipient human umbilical vein endothelial cells (HUVECs), through blood vessel formation experiments, luciferase reporter assays, RT-qPCR, and western blot analysis. RESULT A total of 3842 endothelial cells were extracted from the scRNA-seq data of ESCC samples and reclustered into five cell subtype. Cell-cell communication analysis revealed cancer cells presented a strong interaction with angiogenesis-like endothelial cells in secreted signaling. MiR-21 was unregulated in ESCC and the carcinoma-derived exo-miR-21 was significantly raised in HUVECs. The exo-miR-21 promoted the proliferation and migration of HUVECs while also enhancing, closed mesh count, and junction number in HUVECs. Mechanistically, dual-luciferase reporter assay revealed that PTEN was the target of miR-21. Meanwhile, p-Akt was significantly increased and suppressed by inhibition of miR-21 and PI3K inhibitor LY294002. CONCLUSION Exo-miR-21-mediated communication between endothelial and cancer cells plays a pivotal role in promoting the angiogenesis of ESCC. Therefore, controlling exo-miR-21 could serve as a novel therapeutic strategy for ESCC by targeting angiogenesis.
Collapse
Affiliation(s)
- Shanbo Zheng
- Department of Thoracic Surgery and State Key Laboratory of Genetic EngineeringFudan University Shanghai Cancer CenterShanghaiPeople's Republic of China
- Institute of Thoracic OncologyFudan UniversityShanghaiPeople's Republic of China
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiPeople's Republic of China
| | - Juan Liao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public HealthSoutheast UniversityNanjingPeople's Republic of China
- Department of Science and Education, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouPeople's Republic of China
| | - Mingjun Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public HealthSoutheast UniversityNanjingPeople's Republic of China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public HealthSoutheast UniversityNanjingPeople's Republic of China
| | - Junjie Lv
- Department of Thoracic Surgery and State Key Laboratory of Genetic EngineeringFudan University Shanghai Cancer CenterShanghaiPeople's Republic of China
- Institute of Thoracic OncologyFudan UniversityShanghaiPeople's Republic of China
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiPeople's Republic of China
| |
Collapse
|
50
|
Ling CC, Sun T, Chen F, Wu H, Tao W, Xie X, Ji D, Gao G, Chen J, Ling Y, Zhang Y. Precise tumor delineation in clinical tissues using a novel acidic tumor microenvironment activatable near-infrared fluorescent contrast agent. Anal Chim Acta 2023; 1279:341815. [PMID: 37827620 DOI: 10.1016/j.aca.2023.341815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/04/2023] [Accepted: 09/09/2023] [Indexed: 10/14/2023]
Abstract
Tumor selective near-infrared (NIR) fluorescent contrast agents has the potential to greatly enhance the efficiency and precision of tumor surgery by enabling real-time tumor margin identification for tumor resection guided by imaging. However, the development of these agents is still challenging. In this study, based on the acidic tumor microenvironment (TME), we designed and synthesized a novel pH-sensitive NIR fluorescent contrast agent OBD from β-carboline. The fluorescence quantum yield of OBD exhibited a notable increase at pH 3.6, approximately 12-fold higher compared to its value at pH 7.4. After cellular uptake, OBD lighted up the cancer cells with high specificity and accumulated in the mitochondria. Spraying OBD emitted selective fluorescence in xenograft tumor tissues with tumor-to-normal tissue ratios (TNR) as high as 11.18, implying successful image-guided surgery. Furthermore, OBD was also shown to track metastasis in spray mode. After simple topical spray, OBD rapidly and precisely visualized the tumor margins of clinical colon and liver tissues with TNR over 4.2. Therefore, the small-molecule fluorescent contrast agent OBD has promising clinical applications in tumor identification during surgery.
Collapse
Affiliation(s)
- Chang-Chun Ling
- Department of General Surgery and Vascular Surgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China.
| | - Tiantian Sun
- Department of General Surgery and Vascular Surgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China; School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Fang Chen
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Hongmei Wu
- Department of General Surgery and Vascular Surgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China; School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Weizhi Tao
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Xudong Xie
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Dongliang Ji
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Ge Gao
- School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001 Nantong, Jiangsu, PR China
| | - Jun Chen
- Department of Hepatobiliary Surgery, Nantong Third People's Hospital and the Third Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China
| | - Yong Ling
- Department of General Surgery and Vascular Surgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China.
| | - Yanan Zhang
- Department of General Surgery and Vascular Surgery, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu, PR China; School of Pharmacy, Nantong Key Laboratory of Small Molecular Drug Innovation, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, 226001 Nantong, Jiangsu, PR China.
| |
Collapse
|