1
|
Yang H, Sun T, Sun Z, Wang H, Liu D, Wu D, Qin T, Zhou M. Unravelling the role of ubiquitin-specific proteases in breast carcinoma: insights into tumour progression and immune microenvironment modulation. World J Surg Oncol 2025; 23:60. [PMID: 39979972 PMCID: PMC11841324 DOI: 10.1186/s12957-025-03667-8] [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: 10/18/2024] [Accepted: 01/19/2025] [Indexed: 02/22/2025] Open
Abstract
Breast cancer is a prevalent malignancy worldwide, and its treatment has increasingly shifted towards precision medicine, with immunotherapy emerging as a key therapeutic strategy. Deubiquitination, an essential epigenetic modification, is regulated by deubiquitinating enzymes (DUBs) and plays a critical role in immune function and tumor progression. Ubiquitin-specific proteases (USPs), a prominent subgroup of DUBs, are involved in regulating immune cell functions, antigen processing, and T cell development in the context of breast cancer. Certain USPs also modulate the differentiation of immune cells, such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs), within the breast cancer immune microenvironment. Furthermore, several USPs influence the expression of PD-L1, thus affecting the efficacy of immune checkpoint inhibitors. The overexpression of USPs may promote immune evasion, contributing to the development of treatment resistance. This review elucidates the role of USPs in modulating the immune microenvironment and immune responses in breast cancer. Additionally, it discusses effective strategies for combining USP inhibitors with other therapeutic agents to enhance treatment outcomes. Therefore, targeting USPs presents the potential to enhance the efficacy of immunotherapy and overcome drug resistance, offering a more effective treatment strategy for breast cancer patients.
Collapse
Affiliation(s)
- Huiyuan Yang
- Qingdao Municipal Hospital Affiliated with Qingdao University, Qingdao, Shandong Province, 266011, China
| | - Tingting Sun
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, Shandong Province, 266011, China
| | - Zhenni Sun
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, Shandong Province, 266011, China
| | - Haining Wang
- Department of Oncology, No. 971 Hospital of the People's Liberation Army Navy, Qingdao, 266001, China
| | - Dongjie Liu
- Department of Second Recuperation, Dalian Rehabilitation Recuperation Center of Joint Logistics Support Force of PLA, Dalian, 116013, China
| | - Dapeng Wu
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, Shandong Province, 266011, China.
| | - Tao Qin
- Qingdao Municipal Hospital Affiliated with Qingdao University, Qingdao, Shandong Province, 266011, China.
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, Shandong Province, 266011, China.
| | - Mi Zhou
- Qingdao Municipal Hospital Affiliated with Qingdao University, Qingdao, Shandong Province, 266011, China.
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, Shandong Province, 266011, China.
| |
Collapse
|
2
|
Liang L, Yue C, Li W, Tang J, He Q, Zeng F, Cao J, Liu S, Chen Y, Li X, Zhou Y. CD38 symmetric dimethyl site R58 promotes malignant tumor cell immune escape by regulating the cAMP-GSK3β-PD-L1 axis. Heliyon 2024; 10:e37958. [PMID: 39386836 PMCID: PMC11462232 DOI: 10.1016/j.heliyon.2024.e37958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024] Open
Abstract
In recent years, immunotherapy has emerged as an effective approach for treating tumors, with programmed cell death ligand 1 (PD-L1)/programmed cell death protein-1 (PD-1) immune checkpoint blockade (ICB) being a promising strategy. However, suboptimal therapeutic efficacy limits its clinical benefit. Understanding the regulation mechanism of PD-L1 expression is crucial for improving anti-PD-L1/PD-1 therapy and developing more effective tumor immunotherapy. Previous studies have revealed that resistance to PD-L1/PD-1 blockade therapy arises from the upregulation of CD38 on tumor cells induced by ATRA and IFN-β, which mediates the inhibition of CD8+ T cell function through adenosine receptor signaling, thereby promoting immune evasion.Yet, the precise role of CD38 in regulating PD-L1 on malignant tumor cells and its impact on CD8+ T cells through PD-L1 remain unclear. Here, we demonstrate that CD38 is highly expressed in malignant tumors (lung cancer, nasopharyngeal carcinoma, cervical cancer) and upregulates PD-L1 protein expression, impairing CD8+ T cell function. Mechanistically, CD38 phosphorylates GSK3β via the adenosine-activated cAMP-PKA signaling pathway, leading to GSK3β inactivation and enhanced PD-L1 stability and expression, facilitating tumor immune escape. Furthermore, we identify PRMT5 as a novel CD38-interacting molecule that symmetrically dimethylates CD38 arginine position 58, augmenting PD-L1 stability and expression through the ADO-cAMP-GSK3β signaling axis. This inhibits CD8+ T cell-mediated tumor cell killing, enabling tumor cells to evade immune surveillance. Our findings suggest that targeting the CD38 R58 site offers a new avenue for enhancing anti-PD-L1/PD-1 therapy efficacy in tumor treatment.
Collapse
Affiliation(s)
- Lin Liang
- Breast Cancer Center, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Chunxue Yue
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Wentao Li
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Jingqiong Tang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Qian He
- Department of Radiation Oncology, Hunan Cancer Hospital & the Afliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Feng Zeng
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Jiaying Cao
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Siyi Liu
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Yan Chen
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| | - Xin Li
- Breast Cancer Center, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
3
|
Zhang Y, Zeng L, Wang M, Yang Z, Zhang H, Gao L, Zhang R, Liu J, Shan W, Chang Y, Liu L, Zhao Q, Li Y, Liu J. RIG-I promotes immune evasion of colon cancer by modulating PD-L1 ubiquitination. J Immunother Cancer 2023; 11:e007313. [PMID: 37758653 PMCID: PMC10537859 DOI: 10.1136/jitc-2023-007313] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Colon cancer is one of the most prevalent cancers and exhibits high mortality worldwide. Despite the certain success in the immunotherapy of many tumor types, the limited response of colon cancer to immunotherapy remains a difficult problem. Retinoic acid-inducible gene-I (RIG-I) is a crucial component in innate antiviral immunity, but its role in antitumor immunity remains unclear. Here, in this report, we found that silencing RIG-I decreased resistance to tumor cells killed by T cells and attenuated colon tumor growth in immunocompetent mice. Meanwhile, overexpressing RIG-I promoted tumor progression, and high expression of RIG-I sensitized cells to anti-programmed cell death protein-1 (PD-1) therapy in vivo. Interestingly, we found that RIG-I influenced programmed cell death ligand 1 (PD-L1) expression to promote colon cancer immune evasion without relying on type I interferon stimulation. Mechanistically, RIG-I could compete with Speckle Type POZ protein (SPOP) to bind PD-L1, leading to attenuation of the polyubiquitination and proteasomal degradation of PD-L1. Collectively, our work reveals new insights into the contribution of RIG-I to driving immune evasion by maintaining the stability of PD-L1 through post-translational modification and provides a promising biomarker of the efficacy of immunotherapy in colon cancer.
Collapse
Affiliation(s)
- Yangyang Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lingxiu Zeng
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Meng Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhenwei Yang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hailin Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Liping Gao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ranran Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jialong Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenqing Shan
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ying Chang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lan Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yong Li
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Jing Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Hubei Clinical Center & Key Lab of Intestinal & Colorectal Diseases, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
4
|
Lu LG, Zhou ZL, Wang XY, Liu BY, Lu JY, Liu S, Zhang GB, Zhan MX, Chen Y. PD-L1 blockade liberates intrinsic antitumourigenic properties of glycolytic macrophages in hepatocellular carcinoma. Gut 2022; 71:2551-2560. [PMID: 35173040 PMCID: PMC9664131 DOI: 10.1136/gutjnl-2021-326350] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/06/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Patients with increased PD-L1+ host cells in tumours are more potent to benefit from antiprogrammed death-1/programmed death ligand-1 (PD-L1) treatment, but the underlying mechanism is still unclear. We aim to elucidate the nature, regulation and functional relevance of PD-L1+ host cells in hepatocellular carcinoma (HCC). DESIGN A total of untreated 184 HCC patients was enrolled randomly. C57BL/6 mice are given injection of Hepa1-6 cells to form autologous hepatoma. ELISpot, flow cytometry and real-time PCR are applied to analyse the phenotypic characteristics of PD-L1+ cells isolated directly from HCC specimens paired with blood samples or generated from ex vivo and in vitro culture systems. Immunofluorescence and immunohistochemistry are performed to detect the presence of immune cells on paraffin-embedded and formalin-fixed samples. The underlying regulatory mechanisms of metabolic switching are assessed by both in vitro and in vivo studies. RESULTS We demonstrate that PD-L1+ host macrophages, which constructively represent the major cellular source of PD-L1 in HCC tumours, display an HLA-DRhighCD86high glycolytic phenotype, significantly produce antitumourigenic IL-12p70 and are polarised by intrinsic glycolytic metabolism. Mechanistically, a key glycolytic enzyme PKM2 triggered by hepatoma cell derived fibronectin 1, via a HIF-1α-dependent manner, concurrently controls the antitumourigenic properties and inflammation-mediated PD-L1 expression in glycolytic macrophages. Importantly, although increased PKM2+ glycolytic macrophages predict poor prognosis of patients, blocking PD-L1 on these cells eliminates PD-L1-dominant immunosuppression and liberates intrinsic antitumourigenic properties. CONCLUSIONS Selectively modulating the 'context' of glycolytic macrophages in HCC tumours might restore their antitumourigenic properties and provide a precise strategy for anticancer therapy.
Collapse
Affiliation(s)
- Li-Gong Lu
- Interventional Radiology Center, Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong, China
| | - Zhi-Ling Zhou
- Interventional Radiology Center, Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong, China
| | - Xu-Yan Wang
- Interventional Radiology Center, Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong, China
| | - Bo-Yuan Liu
- Department of Immunology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Gusu School, Nanjing Medical University, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, China
| | - Jin-Ying Lu
- Department of Immunology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Gusu School, Nanjing Medical University, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, China
| | - Shuai Liu
- Department of Immunology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Gusu School, Nanjing Medical University, Nanjing, Jiangsu, China,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, China
| | - Guang-Bo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Mei-Xiao Zhan
- Interventional Radiology Center, Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong, China
| | - Yun Chen
- Department of Immunology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Gusu School, Nanjing Medical University, Nanjing, Jiangsu, China .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Shen Y, Zhang B, Wei X, Guan X, Zhang W. CXCL8 is a prognostic biomarker and correlated with TNBC brain metastasis and immune infiltration. Int Immunopharmacol 2021; 103:108454. [PMID: 34929481 DOI: 10.1016/j.intimp.2021.108454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/28/2021] [Accepted: 12/07/2021] [Indexed: 12/31/2022]
Abstract
Patients with TNBC are associated with an increased risk of developing brain metastasis and shortest median survival post-brain metastasis-diagnosis. However, the regulatory mechanism of TNBC brain metastasis has not been addressed. Here, by a series of integrated analyses of differential gene expression profile from brain metastases and primary triple negative breast cancer, we identified 15 differentially expressed genes in both TNBC brain metastasis tissue samples and TNBC brain metastasis cell line. After analyzing the prognostic value of those 15 differentially expressed genes, we found that CXCL8 was the only gene associated with multiple prognostic indicators in both all-breast cancer and TNBC populations. Functional and pathway enrichment analyses demonstrated that CXCL8 was associated with humoral immune response and immune cell infiltration. CXCL8 expression had a positive correlation with three immune-related scores (ImmuneScore, ESTIMATEScore and StromalScore), and multiple types of immune cell infiltration, including macrophages, neutrophils and Th1 cells. Besides, we also verified the prometastatic effect of CXCL8, by treating MDA-MB-231 and Hs578t cells with different concentrations of recombinant human CXCL8. Taken together, our results suggest that CXCL8 can be used as a prognostic biomarker and is associated with TNBC brain metastasis and immune infiltration. Our findings provide a new perspective on TNBC brain metastasis and illustrate great potential to develop new CXCL8-targeted therapy for clinical TNBC patients.
Collapse
Affiliation(s)
- Yunzhu Shen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Baoguo Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaowei Wei
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Xiaoxiang Guan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China.
| | - Wenwen Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
6
|
Rasihashemi SZ, Rezazadeh Gavgani E, Majidazar R, Seraji P, Oladghaffari M, Kazemi T, Lotfinejad P. Tumor-derived exosomal PD-L1 in progression of cancer and immunotherapy. J Cell Physiol 2021; 237:1648-1660. [PMID: 34825383 DOI: 10.1002/jcp.30645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/19/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022]
Abstract
Cancer is a gravely important health issue all over the world and has been spreading fast. In recent years immune checkpoint treatment options have been used extensively as a primary line of treatment for different cancer types. PD-1 and its ligand, PD-L1, are members of the immune-checkpoints superfamily. Anti-PD-L1 and anti-PD-1 antibodies have shown efficacy against different cancer types, but fewer than 30% of patients have shown robust therapeutic responses and, therefore, it is hypothesized that exosomal PD-L1 is the mechanism to blame for failure in primary immune checkpoint therapy. The identical membrane topology of exosomal PD-L1 with tumor cell membrane-type provides the possibility to mimic immunosuppressive effects of tumor cell membrane PD-L1. In this review, it is discussed whether exosomal PD-L1 binds to antibodies and hence resistance to immunotherapy will be developed, and targeting exosome biogenesis inhibition can provide a new strategy to overcome tumor resistance to anti-PD-L1 therapy. Diagnostic and prognostic values of exosomal PD-L1 in different cancer types are discussed. Multiple clinical studies conclude that the level of tumor-derived exosomes (TEXs) as a biomarker for diagnosis could distinguish cancer patients from healthy controls. Elevated exosomal PD-L1 levels may be predictive of advanced disease stages, cancer metastasis, lower response to anti-PD-1/PD-L1 therapy, lower overall survival rates, and poor tumor prognosis. These novel findings of TEXs serve as promising therapeutic targets for early diagnosis and prevention of cancer progression.
Collapse
Affiliation(s)
- Seyed Z Rasihashemi
- Department of Cardiothoracic Surgery, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Majidazar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parya Seraji
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mobina Oladghaffari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Lotfinejad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Ghanam W, Bebars SMM. Cluster of Differentiation 274 Antigen Immunohistochemical Expression in Tumor and Peri-tumor Cells of Hodgkin and Non-Hodgkin Lymphoma and Clinicopathological Relation (Single-center Study). Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Cluster of differentiation 274 (CD274) antigen has been investigated in tumors to evaluate its regulation and effect as a predictive of targeted therapy. Its expression and effect in lymphoma have raised interest recently. However, results were mixed and showed wide variations.
AIM: This study aims to explore and compare CD274 antigen immunohistochemical expression in tumor and peri-tumor cells of classic Hodgkin lymphoma (HL) and diffuse large B cells non-HL (NHL) and its relation with clinicopathological criteria.
METHODS: This work was carried out on 78 cases of lymph node excision biopsy (48 HL and 30 NHL). Prepared sections were applied for immunohistochemistry using CD274 monoclonal rabbit anti-human (programmed cell death protein 1 [PD-L1] ZR3-ASR, a Sigma Aldrich company). Assessment of CD274 antigen in tumor cells was considered positive if detected in >10% (membranous staining with cytoplasmic accentuation). Peri-tumor cells were scored as: 0, no positive cells/high-power field (HPF); 1, <10 positive cells/HPF; 2, 10–30 positive cells/HPF; 3, >30 positive cells/HPF.
RESULTS: CD274 antigen was expressed in 53.8% of total lymphoma cases with significantly more expression of CD274 antigen in HL than NHL (66.7% vs. 33.3%). Classic HL showed significantly higher expression of CD274 antigen in tumor and peri-tumor cells and significant association with elevated erythrocyte sedimentation rate and lactate dehydrogenase and male gender.
INTERPRETATION AND CONCLUSION: There is a more frequent and significant expression of CD274 antigen in classic HL than NHL cases in tumor and peri-tumor cells and a significant association with bad prognostic criteria in classic HL. High expression of CD274 antigen in classic HL proposes its potential use as a marker, especially for prognostic indication.
Collapse
|
8
|
Abstract
The development of tumors requires an initiator event, usually exposure to DNA damaging agents that cause genetic alterations such as gene mutations or chromosomal abnormalities, leading to deregulated cell proliferation. Although the mere stochastic accumulation of further mutations may cause tumor progression, it is now clear that an inflammatory microenvironment has a major tumor-promoting influence on initiated cells, in particular when a chronic inflammatory reaction already existed before the initiated tumor cell was formed. Moreover, inflammatory cells become mobilized in response to signals emanating from tumor cells. In both cases, the microenvironment provides signals that initiated tumor cells perceive by membrane receptors and transduce via downstream kinase cascades to modulate multiple cellular processes and respond with changes in cell gene expression, metabolism, and morphology. Cytokines, chemokines, and growth factors are examples of major signals secreted by immune cells, fibroblast, and endothelial cells and mediate an intricate cell-cell crosstalk in an inflammatory microenvironment, which contributes to increased cancer cell survival, phenotypic plasticity and adaptation to surrounding tissue conditions. Eventually, consequent changes in extracellular matrix stiffness and architecture, coupled with additional genetic alterations, further fortify the malignant progression of tumor cells, priming them for invasion and metastasis. Here, we provide an overview of the current knowledge on the composition of the inflammatory tumor microenvironment, with an emphasis on the major signals and signal-transducing events mediating different aspects of stromal cell-tumor cell communication that ultimately lead to malignant progression.
Collapse
|
9
|
Liang B, Hu X, Ding Y, Liu M. Tumor-derived exosomes in the PD-1/PD-L1 axis: Significant regulators as well as promising clinical targets. J Cell Physiol 2020; 236:4138-4151. [PMID: 33275291 DOI: 10.1002/jcp.30197] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022]
Abstract
Programmed cell death-1 (PD-1) is a negative coreceptor mainly expressed on the surface of activated T cells. The binding of PD-1 to its ligand PD-L1 significantly induces non-reactivity of T cells to maintain the balance of autoimmunity and immune tolerance. It is reported that tumor cells highly express PD-L1 to restrict cellular immune response, which is one of the most important mechanisms for tumor to mediate immune escape. Cancer immunotherapy targeting PD-1/PD-L1 has achieved remarkable success so far. Tumor-derived exosomes (TEXs) are lipid bilayer vesicles released by tumor cells in an endosome-dependent manner, mediating communication between tumor cells and adjacent cells in the tumor microenvironment. Through signals transmitted by TEXs, tumor can alter the biological characteristics of these cells to promote tumor growth and metastasis. Recent studies have demonstrated that TEXs not only carry tumor-derived PD-L1, but are also closely related to PD-1/PD-L1 expression on target cells. The primary focus of this review will be on how TEXs regulate the PD-1/PD-L1 axis to promote tumor progression, and the promising clinical applications targeting TEXs and exosomal PD-L1.
Collapse
Affiliation(s)
- Benhui Liang
- Department of Cell Biology, Central South University, Changsha, China
- Xiangya Hospital, Central South University, Changsha, China
| | - Ximin Hu
- Department of Cell Biology, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yinghe Ding
- Department of Cell Biology, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Mujun Liu
- Department of Cell Biology, Central South University, Changsha, China
| |
Collapse
|