1
|
Sun J, Shi X, Wang M, He M, Yang W, Song L. NCAM1-SHIP2 axis upon recognizing microbes inhibits the expressions of inflammatory factors through P38-H3K4me and P38-NF-κB pathways in oyster. Cell Commun Signal 2025; 23:102. [PMID: 39979940 PMCID: PMC11841013 DOI: 10.1186/s12964-025-02087-1] [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: 11/09/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
Neural cell adhesion molecule 1 (NCAM1/CD56) as a well-known surface marker for natural killer (NK) cells plays important roles in cell migration, adhesion, and inflammation. In the present study, NCAM1 homolog containingthree immunoglobulin domains, one fibronectin type 3 domain, a transmembrane region and a cytoplasmic tail with two intracellular immunoreceptor tyrosine-based inhibitory motifs (ITIMs) was identified from the Pacific oyster, Crassostrea gigas (defined as CgNCAM1). The mRNA transcripts of CgNCAM1 were highly expressed in haemocytes. The mRNA expressions of CgNCAM1 in haemocytes increased significantly after Vibrio splendidus stimulation. The positive green signals of CgNCAM1 and SH2-containing inositol 5-phosphatase (CgSHIP2) could translocate onto the haemocyte membrane after V. splendidus stimulation. The recombinant extracellular domains of CgNCAM1 exhibited binding activity towards various pathogen-associated molecular patterns (PAMPs) and microbes. Upon binding to its ligands, CgNCAM1 recruited CgSHIP2 to transduce inhibitor signals to reduce the phosphorylation of CgP38. The inhibition of CgP38 reduced the methylation of histone H3K4 and nuclear translocation of NF-κB, which eventually inhibited the mRNA expressions of inflammatory factors (CgIL17-2/3/6 and CgTNF-2) to suppress inflammation. These results suggested that CgNCAM1 could function as an immune checkpoint to sense different PAMPs and microbes and reduce the inflammation through inhibiting P38-epigenetic and P38-NF-κB pathways in oysters.
Collapse
Affiliation(s)
- Jiejie Sun
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China.
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
- Dalian Ocean University, 52 Heishijiao Street, Dalian, 116023, China.
| | - Xiangqi Shi
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Mengjia Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Muchun He
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Wenwen Yang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China.
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China.
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
- Dalian Ocean University, 52 Heishijiao Street, Dalian, 116023, China.
| |
Collapse
|
2
|
Ke J, Huang S, He Z, Lei S, Lin S, Duan M. TIGIT Regulates T Cell Inflammation in Airway Inflammatory Diseases. Inflammation 2025; 48:15-24. [PMID: 38780694 DOI: 10.1007/s10753-024-02045-y] [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: 03/16/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
TIGIT, a co-inhibitory receptor found on T cells and NK cells, transmits inhibitory signals upon binding to its ligand. This interaction suppresses the activation of various signaling pathways, leading to functional exhaustion of cells, ultimately dampening excessive inflammatory responses or facilitating immune evasion in tumors. Dysregulated TIGIT expression has been noted in T cells across different inflammatory conditions, exhibiting varying effects based on T cell subsets. TIGIT predominantly restrains the effector function of pro-inflammatory T cells, upholds the suppressive function of regulatory T cells, and influences Tfh maturation. Mechanistically, the IL27-induced transcription factors c-Maf and Blimp-1 are believed to be key regulators of TIGIT expression in T cells. Notably, TIGIT expression in T cells is implicated in lung diseases, particularly airway inflammatory conditions such as lung cancer, obstructive pulmonary disease, interstitial lung disease, sarcoidosis, and COVID-19. This review emphasizes the significance of TIGIT in the context of T cell immunity and airway inflammatory diseases.
Collapse
Affiliation(s)
- Junyi Ke
- Guangxi Medical University, Nanning, China
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shu Huang
- Wuming Hospital of Guangxi Medical University, Nanning, China
| | | | - Siyu Lei
- Wuming Hospital of Guangxi Medical University, Nanning, China
| | - Shiya Lin
- Guangxi Medical University, Nanning, China
| | - Minchao Duan
- Wuming Hospital of Guangxi Medical University, Nanning, China.
- Department of Respiratory Medicine, Wuming Hospital of Guangxi Medical University, No.26 Yongning Road, Wuming District, Nanning, 530100, China.
| |
Collapse
|
3
|
Sun J, Tian Y, Yang C. Target therapy of TIGIT; a novel approach of immunotherapy for the treatment of colorectal cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:231-241. [PMID: 39158733 DOI: 10.1007/s00210-024-03346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024]
Abstract
The T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT), a newly discovered checkpoint, is characterized by its elevated expression on CD4 + T cells, CD8 + T cells, natural killer (NK) cells, regulatory T cells (Tregs), and tumor-infiltrating lymphocytes (TILs). Research to date has been shown that TIGIT has been linked to exhaustion of NK cell both and T cells in numerous cancers. CD155, being the specific ligand of TIGIT in humans, emerges as a key target for immunotherapy owing to its crucial interaction with TIGIT. Furthermore, numerous studies have demonstrated that the combination of TIGIT with other immune checkpoint inhibitors (ICIs) and/or traditional treatments elicits a potent antitumor response in colorectal cancer (CRC). This review provides an overview of the structure, function, and signaling pathways associated with TIGIT across multiple immune system cell types. Additionally, focusing on the role of TIGIT in the progression of CRC, this study reviewed various studies exploring TIGIT-based immunotherapy in CRC.
Collapse
Affiliation(s)
- Jing Sun
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Yan Tian
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Changqing Yang
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China.
| |
Collapse
|
4
|
Dadgar-Zankbar L, Elahi Z, Shariati A, Khaledi A, Razavi S, Khoshbayan A. Exploring the role of Fusobacterium nucleatum in colorectal cancer: implications for tumor proliferation and chemoresistance. Cell Commun Signal 2024; 22:547. [PMID: 39548531 PMCID: PMC11566256 DOI: 10.1186/s12964-024-01909-y] [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: 08/15/2024] [Accepted: 10/24/2024] [Indexed: 11/18/2024] Open
Abstract
Fusobacterium nucleatum (Fn) has been extensively studied for its connection to colorectal cancer (CRC) and its potential role in chemotherapy resistance. Studies indicate that Fn is commonly found in CRC tissues and is associated with unfavorable prognosis and treatment failure. It has been shown that Fn promotes chemoresistance by affecting autophagy, a cellular process that helps cells survive under stressful conditions. Additionally, Fn targets specific signaling pathways that activate particular microRNAs and modulate the response to chemotherapy. Understanding the current molecular mechanisms and investigating the importance of Fn-inducing chemoresistance could provide valuable insights for developing novel therapies. This review surveys the role of Fn in tumor proliferation, metastasis, and chemoresistance in CRC, focusing on its effects on the tumor microenvironment, gene expression, and resistance to conventional chemotherapy drugs. It also discusses the therapeutic implications of targeting Fn in CRC treatment and highlights the need for further research.
Collapse
Affiliation(s)
- Leila Dadgar-Zankbar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Elahi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Vice Chancellery of Education and Research, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Aref Shariati
- Infectious Diseases Research Center (IDRC), Arak University of Medical Sciences, Arak, Iran
| | - Azad Khaledi
- Infectious Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
- Department of Microbiology and Immunology, School of Medicine, Kashan University of Medical Sciences, P.O. Box: 87155.111, Kashan, 87154, Iran
| | - Shabnam Razavi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Amin Khoshbayan
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Wu JW, Liu Y, Dai XJ, Liu HM, Zheng YC, Liu HM. CD155 as an emerging target in tumor immunotherapy. Int Immunopharmacol 2024; 131:111896. [PMID: 38518596 DOI: 10.1016/j.intimp.2024.111896] [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: 02/19/2024] [Revised: 03/08/2024] [Accepted: 03/16/2024] [Indexed: 03/24/2024]
Abstract
CD155 is an immunoglobulin-like protein overexpressed in almost all the tumor cells, which not only promotes proliferation, adhesion, invasion, and migration of tumor cells, but also regulates immune responses by interacting with TIGIT, CD226 or CD96 receptors expressed on several immune cells, thereby modulating the functionality of these cellular subsets. As a novel immune checkpoint, the inhibition of CD155/TIGIT, either as a standalone treatment or in conjunction with other immune checkpoint inhibitors, has demonstrated efficacy in managing advanced solid malignancies. In this review, we summarize the intricate relationship between on tumor surface CD155 and its receptors, with further discussion on how they regulate the occurrence of tumor immune escape. In addition, novel therapeutic strategies and clinical trials targeting CD155 and its receptors are summarized, providing a strong rationale and way forward for the development of next-generation immunotherapies.
Collapse
Affiliation(s)
- Jiang-Wan Wu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ying Liu
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Xing-Jie Dai
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Hui-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
6
|
Ye H, Yu W, Li Y, Bao X, Ni Y, Chen X, Sun Y, Chen A, Zhou W, Li J. AIM2 fosters lung adenocarcinoma immune escape by modulating PD-L1 expression in tumor-associated macrophages via JAK/STAT3. Hum Vaccin Immunother 2023; 19:2269790. [PMID: 37877820 PMCID: PMC10601527 DOI: 10.1080/21645515.2023.2269790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 10/08/2023] [Indexed: 10/26/2023] Open
Abstract
This work was devised to discuss the effect of AIM2 on the immunosuppression of LUAD tumors, as well as its molecular mechanism. An allograft mouse model was built. Mouse macrophages were isolated and collected. The infiltration level of Mø and expression of M1 Mø, M2 Mø markers, and PD-L1 were assayed by IHC and flow cytometry. Expression levels of M1 Mø and M2 Mø marker genes and PD-L1 were detected by qPCR. The expression of proteins linked with JAK/STAT3 was tested by western blot. CD8+T cells and NK cells were activated in vitro and co-cultured with mouse macrophages, and their cytotoxicity was detected by LDH method. The proportion of CD206+PD-L1+ cells and the activation and proliferation of CD8+T cells were assayed by flow cytometry. Multicolor immunofluorescence was utilized to assay the co-localization of proteins. AIM2 demonstrated a high expression in LUAD, exhibiting a conspicuous positive correlation with the expression of the M2 Mø markers as well as PD-L1. Expression of M1 markers was upregulated after knockdown of AIM2, while M2 markers expression and PD-L1 were downregulated, and the colocalization of proteins linked with PD-L1 and M2 Mø was decreased. The infiltration and cytotoxicity of CD8+T cells and NK cells increased after silencing AIM2. After the knockdown of AIM2, which was enriched in the JAK/STAT3 pathway, the phosphorylation levels of JAK1, JAK2, and STAT3 were reduced, the immune infiltration level of CD8+T cells increased, and the co-localization level of PD-L1 and PD-1 dropped. The activity and proliferation level of CD8+T cells were increased with the reduced PD-1 expression. AIM2 fosters M2 Mø polarization and PD-L1 expression via the JAK/STAT3 pathway. Moreover, AIM2 promotes the immune escape of LUAD via the PD-1/PD-L1 axis. Our work may blaze a trail for the clinical treatment of LUAD.
Collapse
Affiliation(s)
- Hua Ye
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wenwen Yu
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yunlei Li
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaoqiong Bao
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yangyang Ni
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiangxiang Chen
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yangjie Sun
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ali Chen
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Weilong Zhou
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jifa Li
- Department of Respiratory and Critical Care Medicine of Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Wu D, Tian S, Zhu W. Modulating multidrug resistance to drug-based antitumor therapies through NF-κB signaling pathway: mechanisms and perspectives. Expert Opin Ther Targets 2023; 27:503-515. [PMID: 37314372 DOI: 10.1080/14728222.2023.2225767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/12/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Despite the advances made in cancer treatment in the past decades, therapeutic efficacy is still quite challenging, partially due to the emergence of multidrug resistance (MDR). It is crucial to decipher the underlying mechanisms of resistance in order to develop new therapeutic strategies for cancer patients. Previous studies have shown that activation of nuclear factor-κB (NF-κB) plays key roles in various cellular processes including proliferation, anti-apoptosis, metastasis, invasion, and chemoresistance. AREAS COVERED In this review, we conduct an integrated analysis of the evidence suggesting the vital roles of the NF-κB signaling pathway in MDR during chemotherapy, immunotherapy, endocrine, and targeted therapy. A literature search was performed on NF-κB and drug resistance in PubMed up to February 2023. EXPERT OPINION This review summarizes that the NF-κB signaling pathway exhibits a crucial role in enhancing drug resistance in chemotherapy, immunotherapy, endocrine, and targeted therapy. The application of combination therapy with existing antineoplastic drugs and a safe NF-κB inhibitor could become a promising strategy in cancer treatment. A better understanding of the pathway and mechanisms of drug resistance may help exploit safer and more effective NF-κB-targeting agents for clinical use in the future.
Collapse
Affiliation(s)
- Dapeng Wu
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Sai Tian
- Department of Pediatric Clinic, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| | - Wenjing Zhu
- Clinical Research Center, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
- Department of Respiratory and Critical Care Medicine, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, China
| |
Collapse
|
8
|
Lin Y, Wilk U, Pöhmerer J, Hörterer E, Höhn M, Luo X, Mai H, Wagner E, Lächelt U. Folate Receptor-Mediated Delivery of Cas9 RNP for Enhanced Immune Checkpoint Disruption in Cancer Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205318. [PMID: 36399647 DOI: 10.1002/smll.202205318] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/17/2022] [Indexed: 06/16/2023]
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system offers great opportunities for the treatment of numerous diseases by precise modification of the genome. The functional unit of the system is represented by Cas9/sgRNA ribonucleoproteins (RNP), which mediate sequence-specific cleavage of DNA. For therapeutic applications, efficient and cell-specific transport into target cells is essential. Here, Cas9 RNP nanocarriers are described, which are based on lipid-modified oligoamino amides and folic acid (FolA)-PEG to realize receptor-mediated uptake and gene editing in cancer cells. In vitro studies confirm strongly enhanced potency of receptor-mediated delivery, and the nanocarriers enable efficient knockout of GFP and two immune checkpoint genes, PD-L1 and PVR, at low nanomolar concentrations. Compared with non-targeted nanoparticles, FolA-modified nanocarriers achieve substantially higher gene editing including dual PD-L1/PVR gene disruption after injection into CT26 tumors in vivo. In the syngeneic mouse model, dual disruption of PD-L1 and PVR leads to CD8+ T cell recruitment and distinct CT26 tumor growth inhibition, clearly superior to the individual knockouts alone. The reported Cas9 RNP nanocarriers represent a versatile platform for potent and receptor-specific gene editing. In addition, the study demonstrates a promising strategy for cancer immunotherapy by permanent and combined immune checkpoint disruption.
Collapse
Affiliation(s)
- Yi Lin
- Department of Pharmacy and Center for NanoScience (CeNS), LMU Munich, 81377, Munich, Germany
| | - Ulrich Wilk
- Department of Pharmacy and Center for NanoScience (CeNS), LMU Munich, 81377, Munich, Germany
| | - Jana Pöhmerer
- Department of Pharmacy and Center for NanoScience (CeNS), LMU Munich, 81377, Munich, Germany
| | - Elisa Hörterer
- Department of Pharmacy and Center for NanoScience (CeNS), LMU Munich, 81377, Munich, Germany
| | - Miriam Höhn
- Department of Pharmacy and Center for NanoScience (CeNS), LMU Munich, 81377, Munich, Germany
| | - Xianjin Luo
- Department of Pharmacy and Center for NanoScience (CeNS), LMU Munich, 81377, Munich, Germany
| | - Hongcheng Mai
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Ernst Wagner
- Department of Pharmacy and Center for NanoScience (CeNS), LMU Munich, 81377, Munich, Germany
| | - Ulrich Lächelt
- Department of Pharmacy and Center for NanoScience (CeNS), LMU Munich, 81377, Munich, Germany
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, 1090, Austria
| |
Collapse
|
9
|
Ren Y, Miao JM, Wang YY, Fan Z, Kong XB, Yang L, Cheng G. Oncolytic viruses combined with immune checkpoint therapy for colorectal cancer is a promising treatment option. Front Immunol 2022; 13:961796. [PMID: 35911673 PMCID: PMC9334725 DOI: 10.3389/fimmu.2022.961796] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 06/27/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy is one of the promising strategies in the treatment of oncology. Immune checkpoint inhibitors, as a type of immunotherapy, have no significant efficacy in the clinical treatment of patients with pMMR/MSS/MSI-L mCRC alone. Therefore, there is an urgent need to find combination therapies that can improve the response rate of immune checkpoint inhibitors. Oncolytic viruses are a new class of cancer drugs that, in addition to directly lysing tumor cells, can facilitate the action of immune checkpoint inhibitors by modulating the tumor microenvironment and transforming “cold” tumors into “hot” ones. The combination of oncolytic viruses and immune checkpoint inhibitors is currently being used in several primary and clinical studies to treat tumors with exciting results. The combination of genetically modified “armed” OV with ICIs is expected to be one of the treatment options for pMMR/MSS/MSI-L mCRC. In this paper, we will analyze the current status of oncolytic viruses and ICIs available for the treatment of CRC. The feasibility of OV in combination with ICI for CRC will be discussed in terms of the mechanism of action of OV in treating tumors.
Collapse
Affiliation(s)
- Yi Ren
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jia-Meng Miao
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuan-Yuan Wang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zheng Fan
- Department of Critical Medicine, The First Affiliated Hospital of Suzhou University, Suzhou, China
| | - Xian-Bin Kong
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Xian-Bin Kong, ; Long Yang, long ; Gong Cheng,
| | - Long Yang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Xian-Bin Kong, ; Long Yang, long ; Gong Cheng,
| | - Gong Cheng
- Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
- *Correspondence: Xian-Bin Kong, ; Long Yang, long ; Gong Cheng,
| |
Collapse
|
10
|
Annese T, Tamma R, Ribatti D. Update in TIGIT Immune-Checkpoint Role in Cancer. Front Oncol 2022; 12:871085. [PMID: 35656508 PMCID: PMC9152184 DOI: 10.3389/fonc.2022.871085] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
The in-depth characterization of cross-talk between tumor cells and T cells in solid and hematological malignancies will have to be considered to develop new therapeutical strategies concerning the reactivation and maintenance of patient-specific antitumor responses within the patient tumor microenvironment. Activation of immune cells depends on a delicate balance between activating and inhibitory signals mediated by different receptors. T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) is an inhibitory receptor expressed by regulatory T cells (Tregs), activated T cells, and natural killer (NK) cells. TIGIT pathway regulates T cell-mediated tumor recognition in vivo and in vitro and represents an exciting target for checkpoint blockade immunotherapy. TIGIT blockade as monotherapy or in combination with other inhibitor receptors or drugs is emerging in clinical trials in patients with cancer. The purpose of this review is to update the role of TIGIT in cancer progression, looking at TIGIT pathways that are often upregulated in immune cells and at possible therapeutic strategies to avoid tumor aggressiveness, drug resistance, and treatment side effects. However, in the first part, we overviewed the role of immune checkpoints in immunoediting, the TIGIT structure and ligands, and summarized the key immune cells that express TIGIT.
Collapse
Affiliation(s)
- Tiziana Annese
- Department of Medicine and Surgery, Libera Università del Mediterraneo (LUM) Giuseppe Degennaro University, Bari, Italy.,Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| |
Collapse
|
11
|
Wang D, Gu Y, Yan X, Huo C, Wang G, Zhao Y, Teng M, Li Y. Role of CD155/TIGIT in Digestive Cancers: Promising Cancer Target for Immunotherapy. Front Oncol 2022; 12:844260. [PMID: 35433470 PMCID: PMC9005749 DOI: 10.3389/fonc.2022.844260] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/04/2022] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment restricts the function and survival of various immune cells by up-regulating inhibitory immune checkpoints, and participates in the immune escape of tumors. The development of immunotherapies targeting immune checkpoints, such as programmed cell death receptor 1 antibody and anti-cytotoxic T lymphocyte-associated antigen 4 antibody, has provided many options for cancer treatment. The efficacy of other immune checkpoint inhibitors is also under development and research. Among them, T cell immunoreceptor with Ig and ITIM domains (TIGIT) has shown excellent clinical application prospects. Correspondingly, poliovirus receptor (PVR, CD155), one of the main ligands of TIGIT, is mainly expressed in various human malignant tumors and myeloid cells. CD155 interacts with TIGIT on natural killer cells and T cells, mediating inhibitory immunomodulatory regulation. This study summarized the mechanism of CD155/TIGIT in regulating immune cells and its role in the occurrence and development of digestive system tumors, aiming to provide a new perspective for immunotherapy of digestive cancers.
Collapse
Affiliation(s)
- Daijun Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Yanmei Gu
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Xin Yan
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Chengdong Huo
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Guan Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Yang Zhao
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Muzhou Teng
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yumin Li
- The Second Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
12
|
Bai J, Ding B, Li H. Targeting TNFR2 in Cancer: All Roads Lead to Rome. Front Immunol 2022; 13:844931. [PMID: 35251045 PMCID: PMC8891135 DOI: 10.3389/fimmu.2022.844931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/25/2022] [Indexed: 12/14/2022] Open
Abstract
TNF receptor 2 (TNFR2) has become one of the best potential immune checkpoints that might be targeted, mainly because of its vital role in tumor microenvironments (TMEs). Overexpression of TNFR2 in some tumor cells and essential function in immunosuppressive cells, especially regulatory T cells (Tregs), makes blocking TNFR2 an excellent strategy in cancer treatment; however, there is evidence showing that activating TNFR2 can also inhibit tumor progression in vivo. In this review, we will discuss drugs that block and activate TNFR2 under clinical trials or preclinical developments up till now. Meanwhile, we summarize and explore the possible mechanisms related to them.
Collapse
Affiliation(s)
- Jingchao Bai
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Bowen Ding
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of Breast Oncoplastic Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hui Li
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
13
|
Wu Q, Zhang Z, Ji M, Yan T, Jiang Y, Chen Y, Chang J, Zhang J, Tang D, Zhu D, Wei Y. The Establishment and Experimental Verification of an lncRNA-Derived CD8+ T Cell Infiltration ceRNA Network in Colorectal Cancer. Clin Med Insights Oncol 2022; 16:11795549221092218. [PMID: 35479766 PMCID: PMC9036385 DOI: 10.1177/11795549221092218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/17/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Long noncoding RNAs (LncRNA) lead a vital role in colorectal cancer (CRC) development. The infiltrating CD8+ T cell is the main target of immunotherapy. Our study aimed to figure out the potential mechanism of lncRNAs regulating the function of CD8+ T cells in CRC. METHODS We collected bulk RNA-seq, miRNA-seq, and single-cell RNA-seq (scRNA-seq) data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database. The cibersort algorithm and correlation analysis were used to estimate the abundance of CD8+ T cells and screened out the most relevant lncRNAs. We used scRNA-seq data to identify the main cell lncRNA expressed. Furthermore, one competing endogenous RNA (ceRNA) network focusing on the potential mechanism of lncRNA-derived CD8+ T cell infiltration was constructed. We established a co-culture system to assess the immunosuppressive function of the lncRNA. And we evaluated the effects of the lncRNA on CD8+ T cell cytotoxicity by flow cytometry, qPCR, and clone formation assay. RESULTS Three CD8+ T cell infiltration-related lncRNAs were identified, and LINC00657 was expressed mainly in tumor cells, negatively associated with CD8+ T cell infiltration. Hsa-miRNA-1224-3p and hsa-miRNA-338-5p and SCD, ETS2, UBE2H, and YY1 were identified to construct the ceRNA network. Immunosuppression-related tumor marker CD155 was proved to be positively correlated with LINC00657 and mRNAs in the ceRNA network. In addition, we proved that LINC00657 could impair the cytotoxicity of CD8+ T cells, and its expression was positively associated with CD155 in vitro. CONCLUSIONS We successfully constructed an lncRNA-derived CD8+ T cell infiltration ceRNA network in CRC. LINC00657 may play a leading role in the CRC immune escape and could be a novel immunotherapy target.
Collapse
Affiliation(s)
- Qi Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Technology, Shanghai, China
- Cancer Center, Zhongshan Hospital, Shanghai, China
| | - Zhiyuan Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Technology, Shanghai, China
- Cancer Center, Zhongshan Hospital, Shanghai, China
| | - Meiling Ji
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Technology, Shanghai, China
- Cancer Center, Zhongshan Hospital, Shanghai, China
| | - Tao Yan
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yudong Jiang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Technology, Shanghai, China
- Cancer Center, Zhongshan Hospital, Shanghai, China
| | - Yijiao Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Technology, Shanghai, China
- Cancer Center, Zhongshan Hospital, Shanghai, China
| | - Jiang Chang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Technology, Shanghai, China
- Cancer Center, Zhongshan Hospital, Shanghai, China
| | - Jicheng Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Technology, Shanghai, China
- Cancer Center, Zhongshan Hospital, Shanghai, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People’s Hospital, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Dexiang Zhu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Technology, Shanghai, China
- Cancer Center, Zhongshan Hospital, Shanghai, China
| | - Ye Wei
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Technology, Shanghai, China
- Cancer Center, Zhongshan Hospital, Shanghai, China
- Ye Wei, Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, 200030, Shanghai, China.
| |
Collapse
|