1
|
Marangio C, Milito ND, Putro E, Carnevale A, Capuano C, Zingoni A, Cippitelli M, Santoni A, Paolini R, Molfetta R. NKG2D triggering hampers DNAM-1-mediated signaling in human NK cells. Front Immunol 2025; 16:1575059. [PMID: 40421025 PMCID: PMC12104298 DOI: 10.3389/fimmu.2025.1575059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/17/2025] [Indexed: 05/28/2025] Open
Abstract
Introduction Natural Killer (NK) cells are cytotoxic innate lymphocytes able to detect transformed cells through the balanced action of inhibitory and activating receptors. NKG2D is one of the main activating receptors involved in tumor surveillance thanks to its ability to recognize stress-induced ligands. Of note, the prolonged exposure to NKG2D ligands promotes receptor down-modulation that results in defective activation of NKG2D and other unrelated activating receptors, including DNAM-1 that is also involved in tumor clearance. However, further investigations are necessary to characterize how the NKG2D/DNAM-1 interplay affects NK cell anti-tumor function. Methods Primary cultured human NK cells were stimulated with the natural ligand MICA or an anti-NKG2D agonist antibody. The expression of activating and inhibitory receptors as well as DNAM-1-triggered signaling events and cytotoxicity were evaluated by flow cytometry. DNAM-1-mediated granule polarization was evaluated by confocal microscopy. Results We showed that NKG2D crosslinking mediated by the natural ligand MICA or an agonist antibody had different consequences on primary cultured human NK cells. Indeed, MICA stimulation increases the expression of the checkpoint receptor TIGIT that is able to counteract DNAM-1 activation. Stimulation with the agonist antibody, without altering TIGIT expression, directly inhibits DNAM-1-mediated signal transduction and cytotoxic function with a mechanism that required NKG2D endocytosis. Discussion Our findings contribute to shed light on the functional consequences of NKG2D engagement, demonstrating that a direct impact on DNAM-1-mediated signal transduction occurs independently from the modality of NKG2D crosslinking. Understanding the molecular mechanisms responsible for suppression of NK cell activation may help the development of therapeutic anti-cancer strategies aimed to prevent NK cell dysfunction or to reinvigorate an impaired cytotoxic activity.
Collapse
Affiliation(s)
- Caterina Marangio
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Nadia Domenica Milito
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Erisa Putro
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessia Carnevale
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Cristina Capuano
- Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Alessandra Zingoni
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marco Cippitelli
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Angela Santoni
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Neuromed, Pozzilli, Isernia, Italy
| | - Rossella Paolini
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Rosa Molfetta
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
2
|
Xu X, Luo S, Liu J, Zhang E, Liang H, Duan L. Structural Basis of SARS-CoV-2 Nsp13-Derived Peptide-Mediated NK Cell Activation. Biomacromolecules 2025. [PMID: 40331402 DOI: 10.1021/acs.biomac.5c00168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
As pivotal effectors of antiviral immunity, natural killer (NK) cells are crucial for controlling the spread of COVID-19. The nonstructural protein 13 of SARS-CoV-2 can encode a viral peptide (Nsp13232-240) preventing human leukocyte antigen E (HLA-E) from recognizing inhibitory receptor NKG2A, thereby activating NK cells. The underlying molecular mechanisms of Nsp13232-240 remain unclear. Therefore, we compared the interaction discrepancy between the self-peptide and Nsp13232-240, theoretically predicting its source. Results indicate that electrostatic interaction energy provides the main source of binding, and its attenuation greatly promotes binding affinity differences. Nsp13232-240 disrupts the hydrogen bond network between CD94 and HLA-E, impacting the binding of hot-spot residues, including Q112CD94 and E161HLA-E. Moreover, Nsp13232-240 breaks the salt bridges formed by K217NKG2A and K199NKG2A with HLA-E. Conformational changes induced by Nsp13232-240 lead to diminished atomic contacts and an unstable binding pattern. These findings provide novel insights into the immunomodulatory role of Nsp13232-240 and may inform future NK cell-mediated strategies targeting SARS-CoV-2.
Collapse
Affiliation(s)
- Xiaole Xu
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China
| | - Song Luo
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China
| | - Jinxin Liu
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China
| | - Enhao Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China
| | - Houde Liang
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China
| | - Lili Duan
- School of Physics and Electronics, Shandong Normal University, Jinan, 250014, China
| |
Collapse
|
3
|
Chen WW, Kong YH, Zhang LY. Denosumab combined with immunotherapy, radiotherapy, and granulocyte-macrophage colony-stimulating factor for the treatment of metastatic nasopharyngeal carcinoma: A case report. World J Clin Oncol 2025; 16:95642. [PMID: 39995562 PMCID: PMC11686553 DOI: 10.5306/wjco.v16.i2.95642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/28/2024] [Accepted: 10/22/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Bone is a major site of metastasis in nasopharyngeal carcinoma (NPC). Recently, nuclear factor kappa-beta ligand (RANKL) inhibitors have garnered attention for their ability to inhibit osteoclast formation and bone resorption, as well as their potential to modulate immune functions and thereby enhance the efficacy of programmed cell death protein 1 (PD-1) inhibitor therapy. CASE SUMMARY We present a case of a patient with NPC who developed sternal stalk metastasis and multiple bone metastases with soft tissue invasion following radical chemoradiotherapy and targeted therapy. Prior to chemotherapy, the patient experienced severe bone marrow suppression and opted out of further chemotherapy sessions. However, the patient received combination therapy, including RANKL inhibitors (denosumab) alongside PD-1, radiotherapy, and granulocyte-macrophage colony-stimulating factor (PRaG) therapy (NCT05435768), and achieved 16 months of progression-free survival and more than 35 months of overall survival, without encountering any grade 2 or higher treatment-related adverse events. CONCLUSION Denosumab combined with PRaG therapy could be a new therapeutic approach for the second-line treatment in patients with bone metastases.
Collapse
Affiliation(s)
- Wei-Wu Chen
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| | - Yue-Hong Kong
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| | - Li-Yuan Zhang
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu Province, China
| |
Collapse
|
4
|
Chang TD, Chen YJ, Luo JL, Zhang C, Chen SY, Lin ZQ, Zhang PD, Shen YX, Tang TX, Li H, Dong LM, Tang ZH, Chen D, Wang YM. Adaptation of Natural Killer Cells to Hypoxia: A Review of the Transcriptional, Translational, and Metabolic Processes. Immunotargets Ther 2025; 14:99-121. [PMID: 39990274 PMCID: PMC11846490 DOI: 10.2147/itt.s492334] [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: 08/21/2024] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
As important innate immune cells, natural killer (NK) cells play an essential role in resisting pathogen invasion and eliminating transformed cells. However, the hypoxic microenvironment caused by disease conditions is an important physicochemical factor that impairs NK cell function. With the increasing prominence of NK cells in immunotherapy, there has been a surge of interest in developing biological means through which NK cells may overcome the inhibition caused by hypoxia in disease conditions. Although the effects of hypoxic conditions in shaping the functions of NK cells have been increasingly recognized and investigated, reviews have been scantly. A comprehensive understanding of how NK cells adapt to hypoxia can provide valuable insights into how the functional capacity of NK cells may be restored. This review focuses on the functional alterations of NK cells in response to hypoxia. It delineates the mechanisms by which NK cells adapt to hypoxia at the transcriptional, metabolic, translational levels. Furthermore, given the complexity of the hypoxic microenvironment, we also elucidated the effects of key hypoxic metabolites on NK cells. Finally, this review discusses the current clinical therapies derived from targeting hypoxic NK cells. The study of NK cell adaptation to hypoxia has yielded new insights into immunotherapy. These insights may lead to development of novel strategies to improve the treatment of infectious diseases and cancer.
Collapse
Affiliation(s)
- Te-Ding Chang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yu-Jie Chen
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jia-Liu Luo
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Cong Zhang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Shun-Yao Chen
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhi-Qiang Lin
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Pei-Dong Zhang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - You-Xie Shen
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ting-Xuan Tang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Hui Li
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Li-Ming Dong
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhao-Hui Tang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Deng Chen
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yu-Man Wang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
5
|
Zhao Y, Zhu W, Dong S, Zhang H, Zhou W. Glucose Metabolism Reprogramming of Immune Cells in the Microenvironment of Pancreatic and Hepatobiliary Cancers. J Gastroenterol Hepatol 2025; 40:355-366. [PMID: 39780341 DOI: 10.1111/jgh.16873] [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: 08/20/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND AND AIM Pancreatic and hepatobiliary cancers are increasing in prevalence and contribute significantly to cancer-related mortality worldwide. Emerging therapeutic approaches, particularly immunotherapy, are gaining attention for their potential to harness the patient's immune system to combat these tumors. Understanding the role of immune cells in the tumor microenvironment (TME) and their metabolic reprogramming is key to developing more effective treatment strategies. This review aims to explore the relationship between immune cell function and glucose metabolism in the TME of pancreatic and hepatobiliary cancers. METHODS This review synthesizes current research on the metabolic adaptations of immune cells, specifically focusing on glucose metabolism within the TME of pancreatic and hepatobiliary cancers. We examine the mechanisms by which immune cells influence tumor progression through metabolic reprogramming and how these interactions can be targeted for therapeutic purposes. RESULTS Immune cells in the TME undergo significant metabolic changes, with glucose metabolism playing a central role in modulating immune responses. These metabolic shifts not only affect immune cell function but also influence tumor behavior and progression. The unique metabolic features of immune cells in pancreatic and hepatobiliary cancers provide new opportunities for targeting immune responses to combat these malignancies more effectively. CONCLUSION Understanding the complex relationship between immune cell glucose metabolism and tumor progression in the TME of pancreatic and hepatobiliary cancers offers promising therapeutic strategies. By modulating immune responses through targeted metabolic interventions, it may be possible to improve the efficacy of immunotherapies and better combat these aggressive cancers.
Collapse
Affiliation(s)
- Yongqing Zhao
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Weixiong Zhu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Shi Dong
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Hui Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Wence Zhou
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
| |
Collapse
|
6
|
Teng H, Huang H, Lin C, Twu Y, Yang W, Lin W, Lan H, Lin Y, Hwang W. CT45A1-mediated MLC2 (MYL9) phosphorylation promotes natural killer cell resistance and outer cell fate in a cell-in-cell structure, potentiating the progression of microsatellite instability-high colorectal cancer. Mol Oncol 2025; 19:430-451. [PMID: 39322998 PMCID: PMC11793002 DOI: 10.1002/1878-0261.13736] [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: 11/17/2023] [Revised: 07/15/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024] Open
Abstract
Patients with microsatellite instability-high (MSI-H) colorectal cancer (CRC) have high tumor mutation burden and tumor immunogenicity, exhibiting a higher response rate to immunotherapy and better survival. However, a portion of MSI-H CRC patients still experience adverse disease outcomes. We aimed to identify the tumor-autonomous regulators determining these heterogeneous clinical outcomes. The Cancer Genome Atlas (TCGA) dataset was used to identify regulators in MSI-H CRC patients with unfavorable outcomes. Stable CRC tumor clones expressing targeted regulators were established to evaluate migratory and stemness properties, immune cell vulnerability, and cell-in-cell (CIC) structure formation. RNA-sequencing (RNA-seq) was used to identify enriched biological pathways in stable CRC tumor clones. Clinicopathological characterization of formalin-fixed paraffin-embedded (FFPE) MSI-H CRC specimens was performed to explore the underlying mechanisms involved. We showed that cancer/testis antigen family 45 member A1 (CT45A1) expression was upregulated in MSI-H CRC patients with poor survival outcomes. CT45A1-expressing microsatellite stable (MSS) CRC cells showed enhanced migratory ability. However, CT45A1-expressing MSI-H CRC cells, but not MSS CRC cells, showed higher resistance to natural killer (NK) cell cytotoxicity and served as outer cells in homotypic CIC structures, preventing exogenous or therapeutic antibody access to inner CRC cells. Inactivating RHO-ROCK/MLCK-MLC2 signaling with small-molecule inhibitors or short-hairpin RNAs (shRNAs) targeting myosin light chain kinase (MYLK) abolished NK cell resistance and reduced the outer cell fate of CT45A1-expressing MSI-H CRC cells. In MSI-H CRC patients, CT45A1-positive tumors exhibited increased MLC2 phosphorylation, increased outer cell fate, and decreased survival. We demonstrated that CT45A1 potentiates the advanced progression of MSI-H CRC, and targeting MLC2 phosphorylation may enhance immunotherapy efficacy in CT45A1-positive MSI-H CRC patients.
Collapse
Affiliation(s)
- Hao‐Wei Teng
- Division of Medical Oncology, Department of OncologyTaipei Veterans General HospitalTaiwan
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Hsiang‐Yueh Huang
- Department of Biotechnology and Laboratory Science in MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Chun‐Chi Lin
- Division of Colon and Rectum Surgery, Department of SurgeryTaipei Veterans General HospitalTaiwan
- Department of Surgery, Faculty of Medicine, School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yuh‐Ching Twu
- Department of Biotechnology and Laboratory Science in MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Wen‐Hao Yang
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular MedicineChina Medical UniversityTaichungTaiwan
| | - Wen‐Chun Lin
- Department of Biotechnology and Laboratory Science in MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Hsin‐Yi Lan
- Department of Biotechnology and Laboratory Science in MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yen‐Yu Lin
- Department of Pathology, Fu Jen Catholic University HospitalFu Jen Catholic UniversityNew Taipei CityTaiwan
- School of Medicine, College of MedicineFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Wei‐Lun Hwang
- Department of Biotechnology and Laboratory Science in MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Cancer and Immunology Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
7
|
Linghu D, Zhu Z, Zhang D, Luo Y, Ma J, Li T, Sun Z, Xie Z, Sun J, Cao C. Diethylhexyl phthalate induces immune dysregulation and is an environmental immune disruptor. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136244. [PMID: 39442302 DOI: 10.1016/j.jhazmat.2024.136244] [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: 08/13/2024] [Revised: 10/12/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
Diethylhexyl phthalate (DEHP) is the most abundant phthalate compound in the environment, and has been linked with multiple human diseases. The immune system is closely associated with the occurrence and progression of various diseases. However, minimal research has addressed the impact of DEHP on the immune system. In this study, single-cell RNA sequencing was performed using spleen tissue of mice to comprehensively determine alterations of the immune system in response to DEHP. The results showed that DEHP exposure reduced the absolute number of peripheral white blood cells (WBCs), including lymphocytes, monocytes, eosinophils, basophils, and neutrophils in mice. In addition, scRNA-seq analyses showed that inflammatory signaling and the expression of heat shock proteins (HSPs) were reduced in all peripheral immune cell populations. Furthermore, we established a mice cecal ligation and puncture (CLP) model, and showed that DEHP exacerbated sepsis-induced immunosuppression and organ damage. These results suggest that DEHP is an environmental immune disruptor that undermines the immune system, exacerbating acute infections and organ damage. Our findings offer a novel perspective on the hazards of DEHP to human health.
Collapse
Affiliation(s)
- Dongli Linghu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Zhenru Zhu
- Pingshan Hospital, Southern Medical University, Shenzhen, Guangdong, PR China; Pingshan District Peoples' Hospital of Shenzhen, Shenzhen, Guangdong, PR China
| | - Dongyan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Yongyi Luo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Jing Ma
- Information Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Tao Li
- Medical Department, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Zhichao Sun
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Zheng Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Jingyuan Sun
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| | - Chuanhui Cao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangzhou, Guangdong, PR China.
| |
Collapse
|
8
|
Greppi M, De Franco F, Obino V, Rebaudi F, Goda R, Frumento D, Vita G, Baronti C, Melaiu O, Bozzo M, Candiani S, Vellone VG, Papaccio F, Pesce S, Marcenaro E. NK cell receptors in anti-tumor and healthy tissue protection: Mechanisms and therapeutic advances. Immunol Lett 2024; 270:106932. [PMID: 39303993 DOI: 10.1016/j.imlet.2024.106932] [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: 08/21/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Natural Killer (NK) cells are integral to the innate immune system, renowned for their ability to target and eliminate cancer cells without the need for antigen presentation, sparing normal tissues. These cells are crucial in cancer immunosurveillance due to their diverse array of activating and inhibitory receptors that modulate their cytotoxic activity. However, the tumor microenvironment can suppress NK cell function through various mechanisms. Over recent decades, research has focused on overcoming these tumor escape mechanisms. Initially, efforts concentrated on enhancing T cell activity, leading to impressive results with immunotherapeutic approaches aimed at boosting T cell responses. Nevertheless, a substantial number of patients do not benefit from these treatments and continue to seek effective alternatives. In this context, NK cells present a promising avenue for developing new treatments, given their potent cytotoxic capabilities, safety profile, and activity against T cell-resistant tumors, such as those lacking HLA-I expression. Recent advancements in immunotherapy include strategies to restore and amplify NK cell activity through immune checkpoint inhibitors, cytokines, adoptive NK cell therapy, and CAR-NK cell technology. This review provides a comprehensive overview of NK cell receptors, the tumor escape mechanisms that hinder NK cell function, and the evolving field of NK cell-based cancer immunotherapy, highlighting ongoing efforts to develop more effective and targeted cancer treatment strategies.
Collapse
Affiliation(s)
- Marco Greppi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Fabiana De Franco
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Valentina Obino
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Federico Rebaudi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Rayan Goda
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Davide Frumento
- Department of Education Sciences, University of Rome Tre, Rome, Italy
| | - Giorgio Vita
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy
| | - Camilla Baronti
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Matteo Bozzo
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| | - Simona Candiani
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Valerio G Vellone
- Department of Integrated Surgical and Diagnostic Sciences (DISC), University of Genoa, Genoa, Italy; Pathology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Federica Papaccio
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy.
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Emanuela Marcenaro
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
9
|
Wang F, Li R, Xu JY, Bai X, Wang Y, Chen XR, Pan C, Chen S, Zhou K, Heng BC, Wu X, Guo W, Song Z, Jin SC, Zhou J, Zou XH, Ouyang HW, Liu H. Downregulating human leucocyte antigens on mesenchymal stromal cells by epigenetically repressing a β 2-microglobulin super-enhancer. Nat Biomed Eng 2024; 8:1682-1699. [PMID: 39433971 DOI: 10.1038/s41551-024-01264-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 09/13/2024] [Indexed: 10/23/2024]
Abstract
Immune rejection caused by mismatches in human leucocyte antigens (HLAs) remains a major obstacle to the success of allogeneic cell therapies. Current strategies for the generation of 'universal' immune-compatible cells, particularly the editing of HLA class I (HLA-I) genes or the modulation of proteins that inhibit natural killer cells, often result in genomic instability or cellular cytotoxicity. Here we show that a β2-microglobulin super-enhancer (B2M-SE) that is responsive to interferon-γ is a critical regulator of the expression of HLA-I on mesenchymal stromal cells (MSCs). Targeted epigenetic repression of B2M-SE in MSCs reduced the surface expression of HLA-I below the threshold required to activate allogenic T cells while maintaining levels sufficient to evade cytotoxicity mediated by natural killer cells. In a humanized mouse model, the epigenetically edited MSCs demonstrated improved survival by evading the immune system, allowing them to exert enhanced therapeutic effects on LPS-induced acute lung injury. Targeted epigenetic repression of B2M-SE may facilitate the development of off-the-shelf cell sources for allogeneic cell therapy.
Collapse
Affiliation(s)
- Fei Wang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ran Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University School of Medicine, Hangzhou, China
| | - Jing Yi Xu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxia Bai
- The Women's Hospital, Zhejiang University School of Medicine and Key Laboratory of Women's Reproduction Health of Zhejiang Province, Hangzhou, China
| | - Ying Wang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xu Ri Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Pan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University School of Medicine, Hangzhou, China
| | - Shen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ke Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Boon Chin Heng
- Central Laboratories, Peking University School of Stomatology, Beijing, China
| | - Xuewei Wu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, China
| | - Wei Guo
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, China
| | - Zhe Song
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Cheng Jin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Zhou
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Hui Zou
- Central laboratory, The First Affiliated Hospital School of Medicine, Zhejiang University, Hangzhou, China.
| | - Hong Wei Ouyang
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, China.
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| | - Hua Liu
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
10
|
Attrill MH, Shinko D, Alexiou V, Kartawinata M, Wedderburn LR, Pesenacker AM. The immune landscape of the inflamed joint defined by spectral flow cytometry. Clin Exp Immunol 2024; 218:221-241. [PMID: 39101538 PMCID: PMC11557149 DOI: 10.1093/cei/uxae071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/10/2024] [Accepted: 08/02/2024] [Indexed: 08/06/2024] Open
Abstract
Cellular phenotype and function are altered in different microenvironments. For targeted therapies it is important to understand site-specific cellular adaptations. Juvenile idiopathic arthritis (JIA) is characterized by autoimmune joint inflammation, with frequent inadequate treatment responses. To comprehensively assess the inflammatory immune landscape, we designed a 37-parameter spectral flow cytometry panel delineating mononuclear cells from JIA synovial fluid (SF) of autoimmune inflamed joints, compared to JIA and healthy control blood. Synovial monocytes and NK cells (CD56bright) lack Fc-receptor CD16, suggesting antibody-mediated targeting may be ineffective. B cells and DCs, both in small frequencies in SF, undergo maturation with high 4-1BB, CD71, CD39 expression, supporting T-cell activation. SF effector and regulatory T cells were highly active with newly described co-receptor combinations that may alter function, and suggestion of metabolic reprogramming via CD71, TNFR2, and PD-1. Most SF effector phenotypes, as well as an identified CD4-Foxp3+ T-cell population, were restricted to the inflamed joint, yet specific SF-predominant CD4+ Foxp3+ Treg subpopulations were increased in blood of active but not inactive JIA, suggesting possible recirculation and loss of immunoregulation at distal sites. This first comprehensive dataset of the site-specific inflammatory landscape at protein level will inform functional studies and the development of targeted therapeutics to restore immunoregulatory balance and achieve remission in JIA.
Collapse
Affiliation(s)
- Meryl H Attrill
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- UCL Great Ormond Street Institute of Child Health, Infection, Immunity, and Inflammation Research and Teaching Department, University College London, London, UK
| | - Diana Shinko
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Vicky Alexiou
- UCL Great Ormond Street Institute of Child Health, Infection, Immunity, and Inflammation Research and Teaching Department, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Melissa Kartawinata
- UCL Great Ormond Street Institute of Child Health, Infection, Immunity, and Inflammation Research and Teaching Department, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
| | - Lucy R Wedderburn
- UCL Great Ormond Street Institute of Child Health, Infection, Immunity, and Inflammation Research and Teaching Department, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, UK
- NIHR Biomedical Research Centre at GOSH, London, UK
| | - Anne M Pesenacker
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
11
|
Dong B, Obermajer N, Tsuji T, Matsuzaki J, Bonura CM, Sander C, Withers H, Long MD, Chavel C, Olejniczak SH, Minderman H, Kirkwood JM, Edwards RP, Storkus WJ, Romero P, Kalinski P. NK Receptor Signaling Lowers TCR Activation Threshold, Enhancing Selective Recognition of Cancer Cells by TAA-Specific CTLs. Cancer Immunol Res 2024; 12:1421-1437. [PMID: 38949179 PMCID: PMC11706306 DOI: 10.1158/2326-6066.cir-24-0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/10/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
Cytotoxic CD8+ T lymphocyte (CTL) recognition of non-mutated tumor-associated antigens (TAA), present on cancer cells and also in healthy tissues, is an important element of cancer immunity, but the mechanism of its selectivity for cancer cells and opportunities for its enhancement remain elusive. In this study, we found that CTL expression of the NK receptors (NKR) DNAM1 and NKG2D was associated with the effector status of CD8+ tumor-infiltrating lymphocytes and long-term survival of patients with melanoma. Using MART1 and NY-ESO-1 as model TAAs, we demonstrated that DNAM1 and NKG2D regulate T-cell receptor (TCR) functional avidity and set the threshold for TCR activation of human TAA-specific CTLs. Superior co-stimulatory effects of DNAM1 over CD28 involved enhanced TCR signaling, CTL killer function, and polyfunctionality. Double transduction of human CTLs with TAA-specific TCR and NKRs resulted in strongly enhanced antigen sensitivity, without a reduction in antigen specificity and selectivity of killer function. In addition, the elevation of NKR ligand expression on cancer cells due to chemotherapy also increased CTL recognition of cancer cells expressing low levels of TAAs. Our data help explain the ability of self-antigens to mediate tumor rejection in the absence of autoimmunity and support the development of dual-targeting adoptive T-cell therapies that use NKRs to enhance the potency and selectivity of recognition of TAA-expressing cancer cells.
Collapse
MESH Headings
- Humans
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Melanoma/immunology
- Melanoma/metabolism
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- Lymphocyte Activation/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- Cell Line, Tumor
- MART-1 Antigen/immunology
- MART-1 Antigen/metabolism
- Cytotoxicity, Immunologic
- T Lineage-Specific Activation Antigen 1
Collapse
Affiliation(s)
- Bowen Dong
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
- Department of Medicine, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Nataša Obermajer
- Department of Surgery, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Takemasa Tsuji
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Junko Matsuzaki
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Cindy M. Bonura
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Cindy Sander
- Department of Medicine, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Henry Withers
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Mark D. Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Colin Chavel
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Scott H. Olejniczak
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Hans Minderman
- Department of Flow and Immune Analysis Shared Resource, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - John M. Kirkwood
- Department of Medicine, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Robert P. Edwards
- Department of OB-GYN, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Walter J. Storkus
- Department of Dermatology, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Pedro Romero
- University of Lausanne and Ludwig Institute for Cancer Research; Lausanne, Switzerland
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
- Department of Medicine, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
- Department of Surgery, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| |
Collapse
|
12
|
Wang F, Bashiri Dezfouli A, Multhoff G. The immunomodulatory effects of cannabidiol on Hsp70-activated NK cells and tumor target cells. Mol Immunol 2024; 174:1-10. [PMID: 39126837 DOI: 10.1016/j.molimm.2024.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 05/07/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Cannabidiol (CBD), the major non-psychoactive component of cannabis, exhibits anti-inflammatory properties, but less is known about the immunomodulatory potential of CBD on activated natural killer (NK) cells and/or their targets. Many tumor cells present heat shock protein 70 (Hsp70) on their cell surface in a tumor-specific manner and although a membrane Hsp70 (mHsp70) positive phenotype serves as a target for Hsp70-activated NK cells, a high mHsp70 expression is associated with tumor aggressiveness. This study investigated the immuno-modulatory potential of CBD on NK cells stimulated with TKD Hsp70 peptide and IL-2 (TKD+IL-2) and also on HCT116 p53wt and HCT116 p53-/- colorectal cancer cells exhibiting high and low basal levels of mHsp70 expression. RESULTS Apart from an increase in the density of NTB-A and a reduced expression of LAMP-1, the expression of all other activatory NK cell receptors including NKp30, NKG2D and CD69 which are significantly up-regulated after stimulation with TKD+IL-2 remained unaffected after a co-treatment with CBD. However, the release of major pro-inflammatory cytokines by NK cells such as interferon-γ (IFN-γ) and the effector molecule granzyme B (GrzB) was significantly reduced upon CBD treatment. With respect to the tumor target cells, CBD significantly reduced the elevated expression of mHsp70 but had no effect on the low basal mHsp70 expression. Expression of other NK cell ligands such as MICA and MICB remained unaffected, and the NK cell ligands ULBP and B7-H6 were not expressed on these target cells. Consistent with the reduced mHsp70 expression, treatment of both effector and target cells with CBD reduced the killing of high mHsp70 expressing tumor cells by TKD+IL-2+CBD pre-treated NK cells but had no effect on the killing of low mHsp70 expressing tumor cells. Concomitantly, CBD treatment reduced the TKD+IL-2 induced increased release of IFN-γ, IL-4, TNF-α and GrzB, but CBD had no effect on the release of IFN-α when NK cells were co-incubated with tumor target cells. CONCLUSION Cannabidiol (CBD) may potentially diminish the anti-tumor effectiveness of TKD+IL-2 activated natural killer (NK) cells.
Collapse
Affiliation(s)
- Fei Wang
- Institute of Biological and Medical Imaging (IBMI), Helmholtz Center Munich and Department of Oncology, The second affiliated Hospital of Zunyi Medical University, Zunyi, China; Radiation Immuno-Oncology Group, TranslaTUM - Central Institute for Translational Cancer Research and Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Munich, Germany
| | - Ali Bashiri Dezfouli
- Department of Otolaryngology, Head and Neck Surgery, Klinikum rechts der Isar, TUM School of Medicine and Health, Munich, Germany
| | - Gabriele Multhoff
- Radiation Immuno-Oncology Group, TranslaTUM - Central Institute for Translational Cancer Research and Department of Radiation Oncology, Klinikum rechts der Isar, TUM School of Medicine and Health, Munich, Germany.
| |
Collapse
|
13
|
Hockemeyer K, Sakellaropoulos T, Chen X, Ivashkiv O, Sirenko M, Zhou H, Gambi G, Battistello E, Avrampou K, Sun Z, Guillamot M, Chiriboga L, Jour G, Dolgalev I, Corrigan K, Bhatt K, Osman I, Tsirigos A, Kourtis N, Aifantis I. The stress response regulator HSF1 modulates natural killer cell anti-tumour immunity. Nat Cell Biol 2024; 26:1734-1744. [PMID: 39223375 DOI: 10.1038/s41556-024-01490-z] [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: 02/06/2023] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
Diverse cellular insults converge on activation of the heat shock factor 1 (HSF1), which regulates the proteotoxic stress response to maintain protein homoeostasis. HSF1 regulates numerous gene programmes beyond the proteotoxic stress response in a cell-type- and context-specific manner to promote malignancy. However, the role(s) of HSF1 in immune populations of the tumour microenvironment remain elusive. Here, we leverage an in vivo model of HSF1 activation and single-cell transcriptomic tumour profiling to show that augmented HSF1 activity in natural killer (NK) cells impairs cytotoxicity, cytokine production and subsequent anti-tumour immunity. Mechanistically, HSF1 directly binds and regulates the expression of key mediators of NK cell effector function. This work demonstrates that HSF1 regulates the immune response under the stress conditions of the tumour microenvironment. These findings have important implications for enhancing the efficacy of adoptive NK cell therapies and for designing combinatorial strategies including modulators of NK cell-mediated tumour killing.
Collapse
Affiliation(s)
- Kathryn Hockemeyer
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Theodore Sakellaropoulos
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
- Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, NY, USA
| | - Xufeng Chen
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Olha Ivashkiv
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Maria Sirenko
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Hua Zhou
- Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, NY, USA
| | - Giovanni Gambi
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Elena Battistello
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Kleopatra Avrampou
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Zhengxi Sun
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Maria Guillamot
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Luis Chiriboga
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - George Jour
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Igor Dolgalev
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
- Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, NY, USA
| | - Kate Corrigan
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Kamala Bhatt
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Iman Osman
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Urology, NYU Grossman School of Medicine, New York, NY, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Langone Medical Center, New York, NY, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Aristotelis Tsirigos
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
- Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, NY, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Nikos Kourtis
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA.
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA.
- Regeneron Pharmaceuticals, Tarrytown, NY, USA.
| | - Iannis Aifantis
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA.
- Laura & Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Li F, Shi Y, Ma M, Yang X, Chen X, Xie Y, Liu S. Xianling Lianxia formula improves the efficacy of trastuzumab by enhancing NK cell-mediated ADCC in HER2-positive BC. J Pharm Anal 2024; 14:100977. [PMID: 39493309 PMCID: PMC11531627 DOI: 10.1016/j.jpha.2024.100977] [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: 10/12/2023] [Revised: 02/08/2024] [Accepted: 04/08/2024] [Indexed: 11/05/2024] Open
Abstract
Trastuzumab has improved survival rates in human epidermal growth factor receptor 2 (HER2)-positive breast cancer (BC), but drug resistance leads to treatment failure. Natural killer (NK) cell-mediated antibody-dependent cell cytotoxicity (ADCC) represents an essential antitumor immune mechanism of trastuzumab. Traditional Chinese medicine (TCM) has been used for centuries to treat diseases because of its capacity to improve immune responses. Xianling Lianxia formula (XLLXF), based on the principle of "strengthening body and eliminating toxin", exhibits a synergistic effect in the trastuzumab treatment of patients with HER2-positive BC. Notably, this synergistic effect of XLLXF was executed by enhancing NK cells and ADCC, as demonstrated through in vitro co-culture of NK cells and BC cells and in vivo intervention experiments. Mechanistically, the augmented impact of XLLXF on NK cells is linked to a decrease in cytokine inducible Src homology 2 (SH2) containing protein (CISH) expression, which in turn activates the Janus kinase 1 (JAK1)/signal transducer and activator of transcription 5 (STAT5) pathway. Collectively, these findings suggested that XLLXF holds promise for enhancing NK cell function and sensitizing patients with HER2-positive BC to trastuzumab.
Collapse
Affiliation(s)
- Feifei Li
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Youyang Shi
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Mei Ma
- Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Xiaojuan Yang
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Ying Xie
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Sheng Liu
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| |
Collapse
|
15
|
Blanquart E, Ekren R, Rigaud B, Joubert MV, Baylot V, Daunes H, Cuisinier M, Villard M, Carrié N, Mazzotti C, Lucca LE, Perrot A, Corre J, Walzer T, Avet-Loiseau H, Axisa PP, Martinet L. NK cells with adhesion defects and reduced cytotoxic functions are associated with a poor prognosis in multiple myeloma. Blood 2024; 144:1271-1283. [PMID: 38875515 DOI: 10.1182/blood.2023023529] [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: 12/06/2023] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024] Open
Abstract
ABSTRACT The promising results obtained with immunotherapeutic approaches for multiple myeloma (MM) call for a better stratification of patients based on immune components. The most pressing being cytotoxic lymphocytes such as natural killer (NK) cells that are mandatory for MM surveillance and therapy. Here, we performed a single-cell RNA sequencing analysis of NK cells from 10 patients with MM and 10 age/sex-matched healthy donors that revealed important transcriptomic changes in the NK cell landscape affecting both the bone marrow (BM) and peripheral blood compartment. The frequency of mature cytotoxic CD56dim NK cell subsets was reduced in patients with MM at the advantage of late-stage NK cell subsets expressing NF-κB and interferon-I inflammatory signatures. These NK cell subsets accumulating in patients with MM were characterized by low CD16 and CD226 expression and poor cytotoxic functions. MM CD16/CD226Lo NK cells also had adhesion defects with reduced lymphocyte function-associated antigen 1 (LFA-1) integrin activation and actin polymerization that may account for their limited effector functions in vitro. Finally, analysis of BM-infiltrating NK cells in a retrospective cohort of 177 patients with MM from the Intergroupe Francophone du Myélome (IFM) 2009 trial demonstrated that a high frequency of NK cells and their low CD16 and CD226 expression were associated with a shorter overall survival. Thus, CD16/CD226Lo NK cells with reduced effector functions accumulate along MM development and negatively affect patients' clinical outcomes. Given the growing interest in harnessing NK cells to treat myeloma, this improved knowledge around MM-associated NK cell dysfunction will stimulate the development of more efficient immunotherapeutic drugs against MM.
Collapse
Affiliation(s)
- Eve Blanquart
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Rüçhan Ekren
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Bineta Rigaud
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Marie-Véronique Joubert
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Virginie Baylot
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Hélène Daunes
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Marine Cuisinier
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Marine Villard
- Centre International de Recherche en Infectiologie, Université Lyon, Université Claude Bernard Lyon 1 INSERM U1111, Centre National de la Recherche Scientifique, UMR5308, École normale supérieure de Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| | - Nadège Carrié
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Céline Mazzotti
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Liliana E Lucca
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Aurore Perrot
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Jill Corre
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie, Université Lyon, Université Claude Bernard Lyon 1 INSERM U1111, Centre National de la Recherche Scientifique, UMR5308, École normale supérieure de Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| | - Hervé Avet-Loiseau
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
| | - Pierre-Paul Axisa
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Ludovic Martinet
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
- Institut Universitaire du Cancer, Centre hospitalier universitaire de Toulouse, Toulouse, France
- Centre International de Recherche en Infectiologie, Université Lyon, Université Claude Bernard Lyon 1 INSERM U1111, Centre National de la Recherche Scientifique, UMR5308, École normale supérieure de Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
| |
Collapse
|
16
|
Huang Z, Yoo KH, Li D, Yu Q, Ye L, Wei W. Pan-cancer analysis of m1A writer gene RRP8: implications for immune infiltration and prognosis in human cancers. Discov Oncol 2024; 15:437. [PMID: 39266915 PMCID: PMC11393379 DOI: 10.1007/s12672-024-01299-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Ribosomal RNA Processing 8 (RRP8) is a gene associated with RNA modification and has been implicated in the development of several types of tumors in recent research. Nevertheless, the biological importance of RRP8 in pan-cancer has not yet been thoroughly and comprehensively investigated. METHODS In this study, we conducted an analysis of various public databases to investigate the biological functions of RRP8. Our analysis included examining its correlation with pan-cancer prognosis, heterogeneity, stemness, immune checkpoint genes, and immune cell infiltration. Furthermore, we utilized the GDSC and CTRP databases to assess the sensitivity of RRP8 to small molecule drugs. RESULTS Our findings indicate that RRP8 exhibits differential expression between tumor and normal samples, particularly impacting the prognosis of various cancers such as Adrenocortical carcinoma (ACC) and Kidney Chromophobe (KICH). The expression of RRP8 is intricately linked to tumor heterogeneity and stemness markers. Additionally, RRP8 shows a positive correlation with the presence of tumor-infiltrating cells, with TP53 being the predominant mutated gene in these malignancies. CONCLUSION Our findings suggest that RRP8 may serve as a potential prognostic marker and therapeutic target in a variety of cancer types.
Collapse
Affiliation(s)
- Zhihui Huang
- Operating Room, West China Hospital, Sichuan University, Chengdu, China
- West China School of Nursing, Sichuan University, Chengdu, China
- West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, Seoul, South Korea
| | - Duohui Li
- Department of Pharmacy Management, Anqing Municipal Hospital, Anqing, 246000, Anhui, China.
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315211, Zhejiang, China.
- Department of pathology, Ningbo Medical Centre Lihuili Hospital, Ningbo, China.
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China.
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
17
|
Aublin-Gex A, Jacolin F, Diaz O, Jacquemin C, Marçais A, Walzer T, Lotteau V, Vidalain PO, Perrin-Cocon L. Tethering of hexokinase 2 to mitochondria promotes resistance of liver cancer cells to natural killer cell cytotoxicity. Eur J Immunol 2024; 54:e2350954. [PMID: 38837415 DOI: 10.1002/eji.202350954] [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: 12/13/2023] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Hexokinases (HKs) control the first step of glucose catabolism. A switch of expression from liver HK (glucokinase, GCK) to the tumor isoenzyme HK2 is observed in hepatocellular carcinoma progression. Our prior work revealed that HK isoenzyme switch in hepatocytes not only regulates hepatic metabolic functions but also modulates innate immunity and sensitivity to Natural Killer (NK) cell cytotoxicity. This study investigates the impact of HK2 expression and its mitochondrial binding on the resistance of human liver cancer cells to NK-cell-induced cytolysis. We have shown that HK2 expression induces resistance to NK cell cytotoxicity in a process requiring mitochondrial binding of HK2. Neither HK2 nor GCK expression affects target cells' ability to activate NK cells. In contrast, mitochondrial binding of HK2 reduces effector caspase 3/7 activity both at baseline and upon NK-cell activation. Furthermore, HK2 tethering to mitochondria enhances their resistance to cytochrome c release triggered by tBID. These findings indicate that HK2 mitochondrial binding in liver cancer cells is an intrinsic resistance factor to cytolysis and an escape mechanism from immune surveillance.
Collapse
Affiliation(s)
- Anne Aublin-Gex
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Florentine Jacolin
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Olivier Diaz
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Clémence Jacquemin
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie, Team Lymphocyte activation and signaling, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie, Team Lymphocyte activation and signaling, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Vincent Lotteau
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Laure Perrin-Cocon
- CIRI, Centre International de Recherche en Infectiologie, Team Viral Infection, Metabolism and Immunity, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| |
Collapse
|
18
|
Sun X, Wu Y, Li H, Zhao A, Niu T. Harmonizing efficacy and safety: the potentials of CAR-NK in effectively addressing severe toxicities of CAR-T therapy in mantle cell lymphoma. Int J Surg 2024; 110:5871-5872. [PMID: 38801456 PMCID: PMC11392170 DOI: 10.1097/js9.0000000000001638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024]
Affiliation(s)
- Xu Sun
- Department of Hematology, West China Hospital, Sichuan University
| | - Yijun Wu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - He Li
- Department of Hematology, West China Hospital, Sichuan University
| | - Ailin Zhao
- Department of Hematology, West China Hospital, Sichuan University
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University
| |
Collapse
|
19
|
Zang J, Mei Y, Zhu S, Yin S, Feng N, Ci T, Lyu Y. Natural Killer-Based Therapy: A Prospective Thought for Cancer Treatment Related to Diversified Drug Delivery Pathways. Pharmaceutics 2024; 16:939. [PMID: 39065636 PMCID: PMC11279587 DOI: 10.3390/pharmaceutics16070939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Immunotherapy has been a research hotspot due to its low side effects, long-lasting efficacy, and wide anti-tumor spectrum. Recently, NK cell-based immunotherapy has gained broad attention for its unique immunological character of tumor identification and eradication and low risk of graft-versus-host disease and cytokine storm. With the cooperation of a drug delivery system (DDS), NK cells activate tumoricidal activity by adjusting the balance of the activating and inhibitory signals on their surface after drug-loaded DDS administration. Moreover, NK cells or NK-derived exosomes can also be applied as drug carriers for distinct modification to promote NK activation and exert anti-tumor effects. In this review, we first introduce the source and classification of NK cells and describe the common activating and inhibitory receptors on their surface. Then, we summarize the strategies for activating NK cells in vivo through various DDSs. Finally, the application prospects of NK cells in tumor immunotherapy are also discussed.
Collapse
Affiliation(s)
- Jing Zang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Z.); (N.F.)
| | - Yijun Mei
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China;
| | - Shiguo Zhu
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China;
| | - Shaoping Yin
- School of Pharmacy, Jiangsu Provincial Engineering Research Center of Traditional Chinese Medicine External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Nianping Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Z.); (N.F.)
| | - Tianyuan Ci
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Z.); (N.F.)
| | - Yaqi Lyu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (J.Z.); (N.F.)
| |
Collapse
|
20
|
Huang M, Liu Y, Yan Q, Peng M, Ge J, Mo Y, Wang Y, Wang F, Zeng Z, Li Y, Fan C, Xiong W. NK cells as powerful therapeutic tool in cancer immunotherapy. Cell Oncol (Dordr) 2024; 47:733-757. [PMID: 38170381 DOI: 10.1007/s13402-023-00909-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Natural killer (NK) cells have gained considerable attention and hold great potential for their application in tumor immunotherapy. This is mainly due to their MHC-unrestricted and pan-specific recognition capabilities, as well as their ability to rapidly respond to and eliminate target cells. To artificially generate therapeutic NK cells, various materials can be utilized, such as peripheral blood mononuclear cells (PBMCs), umbilical cord blood (UCB), induced pluripotent stem cells (iPSCs), and NK cell lines. Exploiting the therapeutic potential of NK cells to treat tumors through in vivo and in vitro therapeutic modalities has yielded positive therapeutic results. CONCLUSION This review provides a comprehensive description of NK cell therapeutic approaches for tumors and discusses the current problems associated with these therapeutic approaches and the prospects of NK cell therapy for tumors.
Collapse
Affiliation(s)
- Mao Huang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yixuan Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Qijia Yan
- Department of Pathology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Miao Peng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Junshang Ge
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yumin Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Fuyan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Medicine, Comprehensive Cancer Center, Baylor College of Medicine, Alkek Building, RM N720, Houston, TX, USA
| | - Chunmei Fan
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, 410013, Changsha, Hunan Province, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
21
|
Frederiksen HR, Glantz A, Vøls KK, Skov S, Tveden-Nyborg P, Freude K, Doehn U. CRISPR-Cas9 immune-evasive hESCs are rejected following transplantation into immunocompetent mice. Front Genome Ed 2024; 6:1403395. [PMID: 38863835 PMCID: PMC11165197 DOI: 10.3389/fgeed.2024.1403395] [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: 03/19/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024] Open
Abstract
Although current stem cell therapies exhibit promising potential, the extended process of employing autologous cells and the necessity for donor-host matching to avert the rejection of transplanted cells significantly limit the widespread applicability of these treatments. It would be highly advantageous to generate a pluripotent universal donor stem cell line that is immune-evasive and, therefore, not restricted by the individual's immune system, enabling unlimited application within cell replacement therapies. Before such immune-evasive stem cells can be moved forward to clinical trials, in vivo testing via transplantation experiments in immune-competent animals would be a favorable approach preceding preclinical testing. By using human stem cells in immune competent animals, results will be more translatable to a clinical setting, as no parts of the immune system have been altered, although in a xenogeneic setting. In this way, immune evasiveness, cell survival, and unwanted proliferative effects can be assessed before clinical trials in humans. The current study presents the generation and characterization of three human embryonic stem cell lines (hESCs) for xenogeneic transplantation in immune-competent mice. The major histocompatibility complexes I- and II-encoding genes, B2M and CIITA, have been deleted from the hESCs using CRISPR-Cas9-targeted gene replacement strategies and knockout. B2M was knocked out by the insertion of murine CD47. Human-secreted embryonic alkaline phosphatase (hSEAP) was inserted in a safe harbor site to track cells in vivo. The edited hESCs maintained their pluripotency, karyotypic normality, and stable expression of murine CD47 and hSEAP in vitro. In vivo transplantation of hESCs into immune-competent BALB/c mice was successfully monitored by measuring hSEAP in blood samples. Nevertheless, transplantation of immune-evasive hESCs resulted in complete rejection within 11 days, with clear immune infiltration of T-cells on day 8. Our results reveal that knockout of B2M and CIITA together with species-specific expression of CD47 are insufficient to prevent rejection in an immune-competent and xenogeneic context.
Collapse
Affiliation(s)
- Henriette Reventlow Frederiksen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Søren Skov
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pernille Tveden-Nyborg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Doehn
- Cell Therapy Research, Novo Nordisk A/S, Maaloev, Denmark
| |
Collapse
|
22
|
Sakano Y, Sakano K, Hurrell BP, Helou DG, Shafiei-Jahani P, Kazemi MH, Li X, Shen S, Hilser JR, Hartiala JA, Allayee H, Barbers R, Akbari O. Blocking CD226 regulates type 2 innate lymphoid cell effector function and alleviates airway hyperreactivity. J Allergy Clin Immunol 2024; 153:1406-1422.e6. [PMID: 38244725 DOI: 10.1016/j.jaci.2024.01.003] [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: 08/25/2023] [Revised: 12/19/2023] [Accepted: 01/03/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND Type 2 innate lymphoid cells (ILC2s) play a pivotal role in type 2 asthma. CD226 is a costimulatory molecule involved in various inflammatory diseases. OBJECTIVE We aimed to investigate CD226 expression and function within human and mouse ILC2s, and to assess the impact of targeting CD226 on ILC2-mediated airway hyperreactivity (AHR). METHODS We administered IL-33 intranasally to wild-type mice, followed by treatment with anti-CD226 antibody or isotype control. Pulmonary ILC2s were sorted for ex vivo analyses through RNA sequencing and flow cytometry. Next, we evaluated the effects of CD226 on AHR and lung inflammation in wild-type and Rag2-/- mice. Additionally, we compared peripheral ILC2s from healthy donors and asthmatic patients to ascertain the role of CD226 in human ILC2s. RESULTS Our findings demonstrated an inducible expression of CD226 in activated ILC2s, enhancing their cytokine secretion and effector functions. Mechanistically, CD226 alters intracellular metabolism and enhances PI3K/AKT and MAPK signal pathways. Blocking CD226 ameliorates ILC2-dependent AHR in IL-33 and Alternaria alternata-induced models. Interestingly, CD226 is expressed and inducible in human ILC2s, and its blocking reduces cytokine production. Finally, we showed that peripheral ILC2s in asthmatic patients exhibited elevated CD226 expression compared to healthy controls. CONCLUSION Our findings underscore the potential of CD226 as a novel therapeutic target in ILC2s, presenting a promising avenue for ameliorating AHR and allergic asthma.
Collapse
Affiliation(s)
- Yoshihiro Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Kei Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Mohammad H Kazemi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Xin Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Stephen Shen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - James R Hilser
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Jaana A Hartiala
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Hooman Allayee
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Richard Barbers
- Department of Clinical Medicine, Division of Pulmonary and Critical Care Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif.
| |
Collapse
|
23
|
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
|
24
|
Niso-Santano M, Fuentes JM, Galluzzi L. Immunological aspects of central neurodegeneration. Cell Discov 2024; 10:41. [PMID: 38594240 PMCID: PMC11004155 DOI: 10.1038/s41421-024-00666-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/02/2024] [Indexed: 04/11/2024] Open
Abstract
The etiology of various neurodegenerative disorders that mainly affect the central nervous system including (but not limited to) Alzheimer's disease, Parkinson's disease and Huntington's disease has classically been attributed to neuronal defects that culminate with the loss of specific neuronal populations. However, accumulating evidence suggests that numerous immune effector cells and the products thereof (including cytokines and other soluble mediators) have a major impact on the pathogenesis and/or severity of these and other neurodegenerative syndromes. These observations not only add to our understanding of neurodegenerative conditions but also imply that (at least in some cases) therapeutic strategies targeting immune cells or their products may mediate clinically relevant neuroprotective effects. Here, we critically discuss immunological mechanisms of central neurodegeneration and propose potential strategies to correct neurodegeneration-associated immunological dysfunction with therapeutic purposes.
Collapse
Affiliation(s)
- Mireia Niso-Santano
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), Madrid, Spain.
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain.
| | - José M Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
25
|
Xue H, Zhang Z, Li L, Zhu C, Fei K, Sha H, Wu Z, Lin X, Wang F, Zhou S, Deng X, Li Y, Chen B, Xiong Y, Chen K. Characterization of a novel anti-PVRIG antibody with Fc-competent function that exerts strong antitumor effects via NK activation in preclinical models. Cancer Immunol Immunother 2024; 73:81. [PMID: 38554184 PMCID: PMC10981589 DOI: 10.1007/s00262-024-03671-z] [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: 01/08/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024]
Abstract
Poliovirus receptor-related immunoglobulin domain-containing protein, or PVRIG, is a newly discovered immune checkpoint that has emerged as a promising target for cancer immunotherapy. It is primarily expressed on activated T and natural killer (NK) cells, and once engaged with its ligand, PVRL2, it induces inhibitory signaling in T cells, thereby promoting the functional exhaustion of tumor-infiltrating lymphocytes (TILs). Here, we characterized IBI352g4a, a novel humanized anti-PVRIG antibody with Fc-competent function, explored the mechanism of its antitumor activity in preclinical models, and systemically evaluated the contribution of FcrR engagement to PVRIG blockade-induced antitumor activity. IBI352g4a binds to the extracellular domain of human PVRIG with high affinity (Kd = 0.53 nM) and specificity, and fully blocks the interaction between PVRIG and its ligand PVRL2. Unlike other immune checkpoints, IBI352g4a significantly induced NK cell activation and degranulation, but had a minimal effect on T-cell activation in in vitro functional assays. IBI352g4a induced strong antitumor effect in several preclinic models, through in vivo mechanism analysis we found that both NK and T cells contribute to the antitumor effect, but NK cells play predominant roles. Specifically, a single dose of IBI352g4a induced significant NK cell activation in TILs, but T-cell activation was observed only after the second dose. Moreover, the Fc effector function is critical for both NK cell activation and treatment efficacy in vitro and in vivo. Our study, for the first time, demonstrates that both NK activation and FcrR engagement are required for antitumor efficacy induced by PVRIG blockade.
Collapse
Affiliation(s)
- Hongyu Xue
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Zhimin Zhang
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Li Li
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Chenjuan Zhu
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Keke Fei
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Huijun Sha
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Zhihai Wu
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Xiaomin Lin
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Feifei Wang
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Shuaixiang Zhou
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Xiya Deng
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Yiming Li
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China
| | - Bingliang Chen
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China.
| | - Yao Xiong
- Innovent Biologics (Suzhou) Co., Ltd., 168 Dongping Street, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, China.
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
26
|
Reggiani F, Talarico G, Gobbi G, Sauta E, Torricelli F, Manicardi V, Zanetti E, Orecchioni S, Falvo P, Piana S, Lococo F, Paci M, Bertolini F, Ciarrocchi A, Sancisi V. BET inhibitors drive Natural Killer activation in non-small cell lung cancer via BRD4 and SMAD3. Nat Commun 2024; 15:2567. [PMID: 38519469 PMCID: PMC10960013 DOI: 10.1038/s41467-024-46778-8] [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: 03/31/2023] [Accepted: 03/11/2024] [Indexed: 03/25/2024] Open
Abstract
Non-small-cell lung carcinoma (NSCLC) is the most common lung cancer and one of the pioneer tumors in which immunotherapy has radically changed patients' outcomes. However, several issues are emerging and their implementation is required to optimize immunotherapy-based protocols. In this work, we investigate the ability of the Bromodomain and Extra-Terminal protein inhibitors (BETi) to stimulate a proficient anti-tumor immune response toward NSCLC. By using in vitro, ex-vivo, and in vivo models, we demonstrate that these epigenetic drugs specifically enhance Natural Killer (NK) cell cytotoxicity. BETi down-regulate a large set of NK inhibitory receptors, including several immune checkpoints (ICs), that are direct targets of the transcriptional cooperation between the BET protein BRD4 and the transcription factor SMAD3. Overall, BETi orchestrate an epigenetic reprogramming that leads to increased recognition of tumor cells and the killing ability of NK cells. Our results unveil the opportunity to exploit and repurpose these drugs in combination with immunotherapy.
Collapse
Affiliation(s)
- Francesca Reggiani
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| | - Giovanna Talarico
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Giulia Gobbi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Elisabetta Sauta
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Federica Torricelli
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Veronica Manicardi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Eleonora Zanetti
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Biobank, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Stefania Orecchioni
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Paolo Falvo
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Simonetta Piana
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Biobank, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Filippo Lococo
- Università Cattolica del Sacro Cuore, Rome, Italy
- Department of General Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Massimiliano Paci
- Thoracic Surgery Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Alessia Ciarrocchi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Valentina Sancisi
- Translational Research Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| |
Collapse
|
27
|
Miao L, Lu C, Zhang B, Li H, Zhao X, Chen H, Liu Y, Cui X. Advances in metabolic reprogramming of NK cells in the tumor microenvironment on the impact of NK therapy. J Transl Med 2024; 22:229. [PMID: 38433193 PMCID: PMC10909296 DOI: 10.1186/s12967-024-05033-w] [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: 12/06/2023] [Accepted: 02/24/2024] [Indexed: 03/05/2024] Open
Abstract
Natural killer (NK) cells are unique from other immune cells in that they can rapidly kill multiple neighboring cells without the need for antigenic pre-sensitization once the cells display surface markers associated with oncogenic transformation. Given the dynamic role of NK cells in tumor surveillance, NK cell-based immunotherapy is rapidly becoming a "new force" in tumor immunotherapy. However, challenges remain in the use of NK cell immunotherapy in the treatment of solid tumors. Many metabolic features of the tumor microenvironment (TME) of solid tumors, including oxygen and nutrient (e.g., glucose, amino acids) deprivation, accumulation of specific metabolites (e.g., lactate, adenosine), and limited availability of signaling molecules that allow for metabolic reorganization, multifactorial shaping of the immune-suppressing TME impairs tumor-infiltrating NK cell function. This becomes a key barrier limiting the success of NK cell immunotherapy in solid tumors. Restoration of endogenous NK cells in the TME or overt transfer of functionally improved NK cells holds great promise in cancer therapy. In this paper, we summarize the metabolic biology of NK cells, discuss the effects of TME on NK cell metabolism and effector functions, and review emerging strategies for targeting metabolism-improved NK cell immunotherapy in the TME to circumvent these barriers to achieve superior efficacy of NK cell immunotherapy.
Collapse
Affiliation(s)
- Linxuan Miao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116000, People's Republic of China
| | - Chenglin Lu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Bin Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Huili Li
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116000, People's Republic of China
| | - Xu Zhao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116000, People's Republic of China
| | - Haoran Chen
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116000, People's Republic of China
| | - Ying Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China.
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China.
| | - Xiaonan Cui
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China.
| |
Collapse
|
28
|
Zhao J, Li L, Feng X, Fan X, Yin H, Lu Q. T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain as a promising immune checkpoint target for the treatment of SLE. Lupus 2024; 33:209-216. [PMID: 38291414 DOI: 10.1177/09612033241226536] [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] [Indexed: 02/01/2024]
Abstract
Immune checkpoints (ICs) play a pivotal role in orchestrating immune regulation, crucial for the maintenance of immune tolerance and prevention of autoimmune diseases. One noteworthy example among these immune regulators is T cell immunoglobulin (Ig) and immunoreceptor tyrosine-based inhibitory motif (ITIM) domain (TIGIT). The TIGIT pathway's inhibition or the absence of TIGIT has been linked to the hyperactivation and excessive proliferation of T cells, rendering individuals more susceptible to autoimmune diseases and exacerbating inflammatory responses. Conversely, the activation of TIGIT has exhibited promising outcomes in ameliorating autoimmune disorders, as observed in murine models of systemic lupus erythematosus (SLE). Consequently, a judicious exploration of the co-inhibitory axis appears warranted for the effective management of pathogenic immune responses in SLE. In light of compelling evidence, this review undertakes a comprehensive examination of TIGIT's characteristics within the context of autoimmunity, offering insights into its potential as a therapeutic target for SLE.
Collapse
Affiliation(s)
- Junpeng Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Liming Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiwei Feng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xinyu Fan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Huiqi Yin
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
29
|
Chung KP, Su JY, Wang YF, Budiarto BR, Yeh YC, Cheng JC, Keng LT, Chen YJ, Lu YT, Juan YH, Nakahira K, Ruan SY, Chien JY, Chang HT, Jerng JS, Huang YT, Chen SY, Yu CJ. Immunometabolic features of natural killer cells are associated with infection outcomes in critical illness. Front Immunol 2024; 15:1334882. [PMID: 38426112 PMCID: PMC10902670 DOI: 10.3389/fimmu.2024.1334882] [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: 11/08/2023] [Accepted: 01/10/2024] [Indexed: 03/02/2024] Open
Abstract
Immunosuppression increases the risk of nosocomial infection in patients with chronic critical illness. This exploratory study aimed to determine the immunometabolic signature associated with nosocomial infection during chronic critical illness. We prospectively recruited patients who were admitted to the respiratory care center and who had received mechanical ventilator support for more than 10 days in the intensive care unit. The study subjects were followed for the occurrence of nosocomial infection until 6 weeks after admission, hospital discharge, or death. The cytokine levels in the plasma samples were measured. Single-cell immunometabolic regulome profiling by mass cytometry, which analyzed 16 metabolic regulators in 21 immune subsets, was performed to identify immunometabolic features associated with the risk of nosocomial infection. During the study period, 37 patients were enrolled, and 16 patients (43.2%) developed nosocomial infection. Unsupervised immunologic clustering using multidimensional scaling and logistic regression analyses revealed that expression of nuclear respiratory factor 1 (NRF1) and carnitine palmitoyltransferase 1a (CPT1a), key regulators of mitochondrial biogenesis and fatty acid transport, respectively, in natural killer (NK) cells was significantly associated with nosocomial infection. Downregulated NRF1 and upregulated CPT1a were found in all subsets of NK cells from patients who developed a nosocomial infection. The risk of nosocomial infection is significantly correlated with the predictive score developed by selecting NK cell-specific features using an elastic net algorithm. Findings were further examined in an independent cohort of COVID-19-infected patients, and the results confirm that COVID-19-related mortality is significantly associated with mitochondria biogenesis and fatty acid oxidation pathways in NK cells. In conclusion, this study uncovers that NK cell-specific immunometabolic features are significantly associated with the occurrence and fatal outcomes of infection in critically ill population, and provides mechanistic insights into NK cell-specific immunity against microbial invasion in critical illness.
Collapse
Affiliation(s)
- Kuei-Pin Chung
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jia-Ying Su
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Bioinformatics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
| | - Yi-Fu Wang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Bugi Ratno Budiarto
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Yu-Chang Yeh
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jui-Chen Cheng
- Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Ta Keng
- Department of Internal Medicine, National Taiwan University Hospital, Hsinchu, Taiwan
| | - Yi-Jung Chen
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Ting Lu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Hsiu Juan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kiichi Nakahira
- Department of Pharmacology, Nara Medical University, Kashihara, Nara, Japan
| | - Sheng-Yuan Ruan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jung-Yien Chien
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hou-Tai Chang
- Department of Critical Care Medicine, Far Eastern Memorial Hospital, New Taipei, Taiwan
- Department of Industrial Engineering and Management, Yuan Ze University, Taoyuan, Taiwan
| | - Jih-Shuin Jerng
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yen-Tsung Huang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Shih-Yu Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chong-Jen Yu
- Department of Internal Medicine, National Taiwan University Hospital, Hsinchu, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
30
|
Deng R, Zhang L, Chen S, Li X, Xue B, Li H, Xu Y, Tian R, Liu Q, Wang L, Liu S, Yang D, Li P, Tang S, Zhu H. PZR suppresses innate immune response to RNA viral infection by inhibiting MAVS activation in interferon signaling mediated by RIG-I and MDA5. Antiviral Res 2024; 222:105797. [PMID: 38185222 DOI: 10.1016/j.antiviral.2024.105797] [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: 09/27/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/09/2024]
Abstract
RNA viral infections seriously endanger human health. Src homology 2 (SH2) domain-containing protein tyrosine phosphatase 2 (SHP2) suppresses innate immunity against influenza A virus, and pharmacological inhibition of SHP2 provokes hepatic innate immunity. SHP2 binds and catalyzes tyrosyl dephosphorylation of protein zero-related (PZR), but the regulatory effect of PZR on innate immune response to viral infection is unclear. In this study, the transcription and protein level of PZR in host cells were found to be decreased with RNA viral infection, and high level of PZR was uncovered to inhibit interferon (IFN) signaling mediated by RIG-I and MDA5. Through localizing in mitochondria, PZR targeted and interacted with MAVS (also known as IPS-1/VISA/Cardif), suppressing the aggregation and activation of MAVS. Specifically, Y263 residue in ITIM is critical for PZR to exert immunosuppression under RNA viral infection. Moreover, the recruited SHP2 by PZR that modified with tyrosine phosphorylation under RNA viral infection might inhibit phosphorylation activation of MAVS. In conclusion, PZR and SHP2 suppress innate immune response to RNA viral infection through inhibiting MAVS activation. This study reveals the regulatory mechanism of PZR-SHP2-MAVS signal axis on IFN signaling mediated by RIG-I and MDA5, which may provide new sight for developing antiviral drugs.
Collapse
Affiliation(s)
- Rilin Deng
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Lini Zhang
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Shengwen Chen
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Xinran Li
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Binbin Xue
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Pathogen Biology, School of Basic Medicine and Life Science, Department of Pathology and Hainan Province Clinical Medical Center of the First Affiliated Hospital, The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571199, Hainan, China
| | - Huiyi Li
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Pathogen Biology, School of Basic Medicine and Life Science, Department of Pathology and Hainan Province Clinical Medical Center of the First Affiliated Hospital, The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571199, Hainan, China
| | - Yan Xu
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Renyun Tian
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Qian Liu
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Luoling Wang
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Shun Liu
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Di Yang
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Penghui Li
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Songqing Tang
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China
| | - Haizhen Zhu
- Institute of Pathogen Biology and Immunology, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, 410082, Hunan, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Pathogen Biology, School of Basic Medicine and Life Science, Department of Pathology and Hainan Province Clinical Medical Center of the First Affiliated Hospital, The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571199, Hainan, China.
| |
Collapse
|
31
|
Li Y, Li W, Chen J, Qiu S, Liu Y, Xu L, Tian T, Li JP. Deciphering single-cell protein secretion and gene expressions by constructing cell-antibody conjugates. Bioorg Chem 2024; 143:106987. [PMID: 38039927 DOI: 10.1016/j.bioorg.2023.106987] [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: 10/12/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 12/03/2023]
Abstract
Secreted proteins play critical roles in regulating immune responses, exerting cytotoxic effects on tumor cells, promoting inflammatory processes, and influencing cellular metabolism. Deciphering the intricate relationship between the heterogeneity of secreted proteins and their transcriptional states is pivotal in the study of cellular heterogeneity. Here we proposed a cell-antibody conjugate-based sequencing methodology (Cellab-seq) for joint characterization of secreted proteins and transcriptome. Cellab-seq utilizes a chemoenzymatic strategy to construct cell-antibody conjugates, which enables the capture of secreted proteins and their signal transduction with the incorporation of barcode detection antibodies. We applied Cellab-seq to investigate how gene expression influences the activity of secreted proteins in NK cells. Altogether, this strategy facilitates a nuanced understanding of cellular dynamics under diverse physiological conditions, ultimately contributing to the prevention, diagnosis and treatment of diseases.
Collapse
Affiliation(s)
- Yachao Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Wannan Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Jiashang Chen
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Shuang Qiu
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Yilong Liu
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Lingjie Xu
- Vazyme Biotech, Red Maple Hi-tech Industry Park, Kechuang Road, Qixia District, Nanjing, Jiangsu 210023, China
| | - Tian Tian
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China.
| | - Jie P Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
32
|
Barshidi A, Ardeshiri K, Ebrahimi F, Alian F, Shekarchi AA, Hojjat-Farsangi M, Jadidi-Niaragh F. The role of exhausted natural killer cells in the immunopathogenesis and treatment of leukemia. Cell Commun Signal 2024; 22:59. [PMID: 38254135 PMCID: PMC10802000 DOI: 10.1186/s12964-023-01428-2] [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: 09/28/2023] [Accepted: 12/08/2023] [Indexed: 01/24/2024] Open
Abstract
The immune responses to cancer cells involve both innate and acquired immune cells. In the meantime, the most attention has been drawn to the adaptive immune cells, especially T cells, while, it is now well known that the innate immune cells, especially natural killer (NK) cells, play a vital role in defending against malignancies. While the immune cells are trying to eliminate malignant cells, cancer cells try to prevent the function of these cells and suppress immune responses. The suppression of NK cells in various cancers can lead to the induction of an exhausted phenotype in NK cells, which will impair their function. Recent studies have shown that the occurrence of this phenotype in various types of leukemic malignancies can affect the prognosis of the disease, and targeting these cells may be considered a new immunotherapy method in the treatment of leukemia. Therefore, a detailed study of exhausted NK cells in leukemic diseases can help both to understand the mechanisms of leukemia progression and to design new treatment methods by creating a deeper understanding of these cells. Here, we will comprehensively review the immunobiology of exhausted NK cells and their role in various leukemic malignancies. Video Abstract.
Collapse
Affiliation(s)
- Asal Barshidi
- Department of Biological Sciences, Faculty of Sciences, University of Kurdistan, Sanandaj, Iran
| | - Keivan Ardeshiri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Farbod Ebrahimi
- Nanoparticle Process Technology, Faculty of Engineering, University of Duisburg-Essen, Duisburg, Germany
| | - Fatemeh Alian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Liu PC, Li JB, Huang YP, Zhang M, Yu SJ, Wu R. Overexpression of regulatory T cells in patients with unexplained recurrent pregnancy loss: friend or foe? Front Med (Lausanne) 2024; 10:1244424. [PMID: 38239620 PMCID: PMC10794536 DOI: 10.3389/fmed.2023.1244424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/14/2023] [Indexed: 01/22/2024] Open
Abstract
Background This study aimed to investigate the role of regulatory T cells in patients with unexplained recurrent pregnancy loss (URPL). Methods We retrospectively analyzed 136 women who had experienced two or more miscarriages before 24 weeks of gestation for no obvious reason from May 2018 to October 2021. The basic clinical data of the patients and expression of lymphocyte subsets such as regulatory T cells (Tregs) and natural killer cells (NKs) by flow cytometry were collected to explore the risk factors of pregnancy outcome in URPL patients. Results A total of 136 URPL patients were enrolled in this study. Eventually, 50 patients attained clinical pregnancy. The median age was 31.8 ± 4.6 years in patients with clinical pregnancy. The univariate and multivariate logistic regression analyses indicated that Tregs was associated with the pregnancy outcomes of patients with URPL (odds ratio 0.63, 95% confidence interval 0.50-0.80). More importantly, a U-shaped association was found between Tregs and pregnancy outcome (p < 0.001), with either higher or lower Tregs levels adversely affecting pregnancy outcome. Conclusion Tregs levels that are either too high or too low can harm pregnancy outcomes. It was expected to be a very promising quantitative biomarker for predicting pregnancy outcomes in URPL patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Rui Wu
- Department of Rheumatology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
34
|
Zacharias ZR, Houtman JCD. OMIP-099: 31-color spectral flow cytometry panel to investigate the steady-state phenotype of human T cells. Cytometry A 2024; 105:10-15. [PMID: 37814476 PMCID: PMC10842108 DOI: 10.1002/cyto.a.24799] [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: 02/02/2023] [Revised: 08/07/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023]
Abstract
We have developed a 31-color panel to define the steady-state phenotype of T cells in human peripheral blood (Table 1). The panel presented here was optimized using cryopreserved peripheral blood mononuclear cells (PBMC). The markers included in this panel were chosen in order to characterize the steady-state phenotype of T cells and includes markers (CD45RA, CD45RO, CCR7, CD95) to distinguish the main subsets (e.g., naïve, TEM , TCM , TEMRA , TSCM etc.) of CD4, CD8, and γδ T cells. This panel also includes markers for the identification of differentiation status (CD27, CD28), activation/antigen experience status (CD11a, CD49d, CD38, HLA-DR, CD56, and CD39), co-inhibitory marker expression (PD-1, TIM-3), and CD4 T helper subsets (CXCR3, CXCR5, CCR4, CCR6, Foxp3, CD25, and CD127). This optimized panel provides a broad assessment of the steady-state phenotype of human T cells.
Collapse
Affiliation(s)
- Zeb R. Zacharias
- Human Immunology Core, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Jon C. D. Houtman
- Human Immunology Core, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
35
|
Rahimi A, Malakoutikhah Z, Rahimmanesh I, Ferns GA, Nedaeinia R, Ishaghi SMM, Dana N, Haghjooy Javanmard S. The nexus of natural killer cells and melanoma tumor microenvironment: crosstalk, chemotherapeutic potential, and innovative NK cell-based therapeutic strategies. Cancer Cell Int 2023; 23:312. [PMID: 38057843 DOI: 10.1186/s12935-023-03134-y] [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: 05/09/2023] [Accepted: 11/09/2023] [Indexed: 12/08/2023] Open
Abstract
The metastasis of melanoma cells to regional lymph nodes and distant sites is an important contributor to cancer-related morbidity and mortality among patients with melanoma. This intricate process entails dynamic interactions involving tumor cells, cellular constituents, and non-cellular elements within the microenvironment. Moreover, both microenvironmental and systemic factors regulate the metastatic progression. Central to immunosurveillance for tumor cells are natural killer (NK) cells, prominent effectors of the innate immune system with potent antitumor and antimetastatic capabilities. Recognizing their pivotal role, contemporary immunotherapeutic strategies are actively integrating NK cells to combat metastatic tumors. Thus, a meticulous exploration of the interplay between metastatic melanoma and NK cells along the metastatic cascade is important. Given the critical involvement of NK cells within the melanoma tumor microenvironment, this comprehensive review illuminates the intricate relationship between components of the melanoma tumor microenvironment and NK cells, delineating their multifaceted roles. By shedding light on these critical aspects, this review advocates for a deeper understanding of NK cell dynamics within the melanoma context, driving forward transformative strategies to combat this cancer.
Collapse
Affiliation(s)
- Azadeh Rahimi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Malakoutikhah
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Nasim Dana
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
36
|
Wang H, Sun D, Chen J, Li H, Chen L. Nectin-4 has emerged as a compelling target for breast cancer. Eur J Pharmacol 2023; 960:176129. [PMID: 38059449 DOI: 10.1016/j.ejphar.2023.176129] [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: 05/17/2023] [Revised: 10/08/2023] [Accepted: 10/18/2023] [Indexed: 12/08/2023]
Abstract
The incidence of breast cancer in women has increased year by year, becoming one of the most common malignant tumors in females worldwide. Most patients can be treated with surgery and endocrine drugs, but there are still some patients who lack effective treatment, such as triple-negative breast cancer (TNBC). Nectin-4, a protein encoded by poliovirus receptor-associated protein 4, is a Ca2+-independent immunoglobulin-like protein. It is mainly involved in the adhesion between cells. In recent years, studies have found that Nectin-4 is overexpressed in breast cancer and several other malignancies. Otherwise, several monoclonal antibodies and inhibitors targeting Nectin-4 have shown prosperous outcomes, so Nectin-4 has great potential to be a therapeutic target for breast cancer. The present review systematically describes the significance of Nectin-4 in each aspect of breast cancer, as well as the molecular mechanisms of these aspects mediated by Nectin-4. We further highlight ongoing or proposed therapeutic strategies for breast cancer specific to Nectin-4.
Collapse
Affiliation(s)
- Hui Wang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dejuan Sun
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jinxia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; Institute of Structural Pharmacology & TCM Chemical Biology, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
37
|
Janes ME, Kinlein A, Flajnik MF, Du Pasquier L, Ohta Y. Genomic view of the origins of cell-mediated immunity. Immunogenetics 2023; 75:479-493. [PMID: 37735270 PMCID: PMC11019866 DOI: 10.1007/s00251-023-01319-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/10/2023] [Indexed: 09/23/2023]
Abstract
NKp30 is an activating natural killer cell receptor (NKR) with a single-exon variable (VJ)-type immunoglobulin superfamily (IgSF) domain. Such VJ-IgSF domains predate the emergence of the antigen receptors (immunoglobulin and T cell receptor), which possess the same domain but undergo gene rearrangement. NCR3, the gene encoding NKp30, is present in jawed vertebrates from sharks to mammals; thus, unlike most NKR that are highly divergent among vertebrate taxa, NKp30 is uniquely conserved. We previously hypothesized that an ancestral NCR3 gene was encoded in the proto-major histocompatibility complex (MHC), the region where many immune-related genes have accumulated. Herein, we searched in silico databases to identify NCR3 paralogues and examined their genomic locations. We found a paralogue, NCR3H, in many vertebrates but was lost in mammals. Additionally, we identified a set of voltage-gated sodium channel beta (SCNB) genes as NCR3-distantly-related genes. Like NCR3, both NCR3H and SCNB proteins contain a single VJ-IgSF domain followed by a transmembrane region. These genes map to MHC paralogous regions, originally described in an invertebrate, along with genes encoding cell adhesion molecules involved in NK cell recognition networks. Other genes having no obvious relationship to immunity also map to these paralogous regions. These gene complexes were traced to several invertebrates, suggesting that the foundation of these cellular networks emerged before the genome-wide duplications in early gnathostome history. Here, we propose that this ancestral region was involved in cell-mediated immunity prior to the emergence of adaptive immunity and that NCR3 piggybacked onto this primordial complex, heralding the emergence of vertebrate NK cell/T cells.
Collapse
Affiliation(s)
- Morgan E Janes
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD, 21201, USA
| | - Allison Kinlein
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD, 21201, USA
| | - Martin F Flajnik
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD, 21201, USA
| | - Louis Du Pasquier
- Department of Environmental Sciences, Zoology, University of Basel, Vesalgasse 1, 4051, Basel, Switzerland
| | - Yuko Ohta
- Department of Microbiology and Immunology, University of Maryland, Baltimore, MD, 21201, USA.
| |
Collapse
|
38
|
Nersesian S, Carter EB, Lee SN, Westhaver LP, Boudreau JE. Killer instincts: natural killer cells as multifactorial cancer immunotherapy. Front Immunol 2023; 14:1269614. [PMID: 38090565 PMCID: PMC10715270 DOI: 10.3389/fimmu.2023.1269614] [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: 07/30/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Natural killer (NK) cells integrate heterogeneous signals for activation and inhibition using germline-encoded receptors. These receptors are stochastically co-expressed, and their concurrent engagement and signaling can adjust the sensitivity of individual cells to putative targets. Against cancers, which mutate and evolve under therapeutic and immunologic pressure, the diversity for recognition provided by NK cells may be key to comprehensive cancer control. NK cells are already being trialled as adoptive cell therapy and targets for immunotherapeutic agents. However, strategies to leverage their naturally occurring diversity and agility have not yet been developed. In this review, we discuss the receptors and signaling pathways through which signals for activation or inhibition are generated in NK cells, focusing on their roles in cancer and potential as targets for immunotherapies. Finally, we consider the impacts of receptor co-expression and the potential to engage multiple pathways of NK cell reactivity to maximize the scope and strength of antitumor activities.
Collapse
Affiliation(s)
- Sarah Nersesian
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Emily B. Carter
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Stacey N. Lee
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | | | - Jeanette E. Boudreau
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
39
|
Chen H, Zhang Y, Shen Y, Jiang L, Zhang G, Zhang X, Xu Y, Fu F. Deficiency of N-linked glycosylation impairs immune function of B7-H6. Front Immunol 2023; 14:1255667. [PMID: 38035117 PMCID: PMC10684670 DOI: 10.3389/fimmu.2023.1255667] [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: 07/09/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
B7-H6 is a novel immune checkpoint molecule that triggers NK cell cytotoxicity, but the role of N-glycosylation in B7-H6 is poorly understood. We here identified the existence of N-glycosylation of B7-H6 in different cell lines and exogenous expression cells by PNGase F digestion and tunicamycin blockage. Subsequently, we demonstrated that B7-H6 contains 6 functional N-linked glycosylation sites by single site mutation and electrophoresis. Phylogenetical and structural analysis revealed that N43 and N208 glycan are conserved in jawed vertebrates and may thus contribute more to the biological functions. We further demonstrated that N43 and N208 glycosylation are essential for B7-H6 to trigger NK cell activation. Mechanistically, we found that N43 and N208 glycan contributed to the stability and membrane expression of B7-H6 protein. Lack of N208 glycosylation led to membrane B7-H6 shedding, while N43 mutation resulted in impaired B7-H6/NKp30 binding affinity. Together, our findings highlight the significance of N-linked glycosylation in B7-H6 biological functions and suggest potential targets for modulating NK cell-mediated immunity.
Collapse
Affiliation(s)
- Hanqing Chen
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Hematology, the First affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Zhang
- Department of Respiratory and Critical Medicine, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Yu Shen
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Jiang
- Suzhou Red Cross Blood Center, Suzhou, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Xu
- Department of Hematology, the First affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fengqing Fu
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
40
|
Ko E, Yoon T, Lee Y, Kim J, Park YB. ADSC secretome constrains NK cell activity by attenuating IL-2-mediated JAK-STAT and AKT signaling pathway via upregulation of CIS and DUSP4. Stem Cell Res Ther 2023; 14:329. [PMID: 37964351 PMCID: PMC10648656 DOI: 10.1186/s13287-023-03516-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: 05/01/2023] [Accepted: 09/25/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have immunomodulatory properties and therapeutic effects on autoimmune diseases through their secreted factors, referred to as the secretome. However, the specific key factors of the MSC secretome and their mechanisms of action in immune cells have not been fully determined. Most in vitro experiments are being performed using immune cells, but experiments using natural killer (NK) cells have been neglected, and a few studies using NK cells have shown discrepancies in results. NK cells are crucial elements of the immune system, and adjustment of their activity is essential for controlling various pathological conditions. The aim of this study was to elucidate the role of the adipose tissue-derived stem cell (ADSC) secretome on NK cell activity. METHODS To obtain the ADSC secretome, we cultured ADSCs in medium and concentrated the culture medium using tangential flow filtration (TFF) capsules. We assessed NK cell viability and proliferation using CCK-8 and CFSE assays, respectively. We analyzed the effects of the ADSC secretome on NK cell activity and pathway-related proteins using a combination of flow cytometry, ELISA, cytotoxicity assay, CD107a assay, western blotting, and quantitative real-time PCR. To identify the composition of the ADSC secretome, we performed LC-MS/MS profiling and bioinformatics analysis. To elucidate the molecular mechanisms involved, we used mRNA sequencing to profile the transcriptional expression of human blood NK cells. RESULTS The ADSC secretome was found to restrict IL-2-mediated effector function of NK cells while maintaining proliferative potency. This effect was achieved through the upregulation of the inhibitory receptor CD96, as well as downregulation of activating receptors and IL-2 receptor subunits IL-2Rα and IL-2Rγ. These changes were associated with attenuated JAK-STAT and AKT pathways in NK cells, which were achieved through the upregulation of cytokine-inducible SH2-containing protein (CIS, encoded by Cish) and dual specificity protein phosphatase 4 (DUSP4). Furthermore, proteomic analysis revealed twelve novel candidates associated with the immunomodulatory effects of MSCs. CONCLUSIONS Our findings reveal a detailed cellular outcome and regulatory mechanism of NK cell activity by the ADSC secretome and suggest a therapeutic tool for treating NK-mediated inflammatory and autoimmune diseases using the MSC secretome.
Collapse
Affiliation(s)
- Eunhee Ko
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Taejun Yoon
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yoojin Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jongsun Kim
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Microbiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
41
|
Bi W, Kraft A, Engelskircher S, Mischke J, Witte M, Klawonn F, van Ham M, Cornberg M, Wedemeyer H, Hengst J, Jänsch L. Proteomics reveals a global phenotypic shift of NK cells in HCV patients treated with direct-acting antivirals. Eur J Immunol 2023; 53:e2250291. [PMID: 37515498 DOI: 10.1002/eji.202250291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 07/31/2023]
Abstract
Chronic hepatitis C virus (HCV) infections compromise natural killer (NK)-cell immunity. Direct-acting antivirals (DAA) effectively eliminate HCV, but the long-term effects on NK cells in cured patients are debated. We conducted a proteomic study on CD56+ NK cells of chronic HCV-infected patients before and 1 year after DAA therapy. Donor-variation was observed in NK-cell proteomes of HCV-infected patients, with 46 dysregulated proteins restored after DAA therapy. However, 30% of the CD56+ NK-cell proteome remained altered 1 year post-therapy, indicating a phenotypic shift with low donor-variation. NK cells from virus-negative cured patients exhibited global regulation of RNA-processing and pathways related to "stimuli response", "chemokine signaling", and "cytotoxicity regulation". Proteomics identified downregulation of vesicle transport components (CD107a, COPI/II complexes) and altered receptor expression profiles, indicating an inhibited NK-cell phenotype. Yet, activated NK cells from HCV patients before and after therapy effectively upregulated IFN-γ and recruited CD107a. Conversely, reduced surface expression levels of Tim-3 and 2B4 were observed before and after therapy. In conclusion, this study reveals long-term effects on the CD56+ NK-cell compartment in convalescent HCV patients 1 year after therapy, with limited abundance of vesicle transport complexes and surface receptors, associated with a responsive NK-cell phenotype.
Collapse
Affiliation(s)
- Wenjie Bi
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, P. R. China
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anke Kraft
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Sophie Engelskircher
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Jasmin Mischke
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Moana Witte
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Frank Klawonn
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Department of Computer Science, Ostfalia University, Wolfenbüttel, Germany
| | - Marco van Ham
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Heiner Wedemeyer
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover Medical School, Hannover, Germany
| | - Julia Hengst
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Lothar Jänsch
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
42
|
Hojjatipour T, Sharifzadeh Z, Maali A, Azad M. Chimeric antigen receptor-natural killer cells: a promising sword against insidious tumor cells. Hum Cell 2023; 36:1843-1864. [PMID: 37477869 DOI: 10.1007/s13577-023-00948-w] [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: 04/01/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023]
Abstract
Natural killer (NK) cells are a critical component of innate immunity, particularly in initial cancer recognition and inhibition of additional tumor growth or metastasis propagation. NK cells recognize transformed cells without prior sensitization via stimulatory receptors and rapidly eradicate them. However, the protective tumor microenvironment facilitates tumor escaping via induction of an exhaustion state in immune cells, including NK cells. Hence, genetic manipulation of NK cells for specific identification of tumor-associated antigens or a more robust response against tumor cells is a promising strategy for NK cells' tumoricidal augmentation. Regarding the remarkable achievement of engineered CAR-T cells in treating hematologic malignancies, there is evolving interest in CAR-NK cell recruitment in cancer immunotherapy. Innate functionality of NK cells, higher safety, superior in vivo maintenance, and the off-the-shelf potential move CAR-NK-based therapy superior to CAR-T cells treatment. In this review, we have comprehensively discussed the recent genetic manipulations of CAR-NK cell manufacturing regarding different domains of CAR constructs and their following delivery systems into diverse sources of NK cells. Then highlight the preclinical and clinical investigations of CAR-NK cells and examine the current challenges and prospects as an optimistic remedy in cancer immunotherapy.
Collapse
Affiliation(s)
- Tahereh Hojjatipour
- Department of Hematology and Blood Transfusion, Students Research Center, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciecnes, Qazvin, Iran
| | - Mehdi Azad
- Department of Medical Laboratory Sciences, School of Paramedicine, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, 3419759811, Iran.
| |
Collapse
|
43
|
Varisli L, Dancik GM, Tolan V, Vlahopoulos S. Critical Roles of SRC-3 in the Development and Progression of Breast Cancer, Rendering It a Prospective Clinical Target. Cancers (Basel) 2023; 15:5242. [PMID: 37958417 PMCID: PMC10648290 DOI: 10.3390/cancers15215242] [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: 10/06/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer (BCa) is the most frequently diagnosed malignant tumor in women and is also one of the leading causes of cancer-related death. Most breast tumors are hormone-dependent and estrogen signaling plays a critical role in promoting the survival and malignant behaviors of these cells. Estrogen signaling involves ligand-activated cytoplasmic estrogen receptors that translocate to the nucleus with various co-regulators, such as steroid receptor co-activator (SRC) family members, and bind to the promoters of target genes and regulate their expression. SRC-3 is a member of this family that interacts with, and enhances, the transcriptional activity of the ligand activated estrogen receptor. Although SRC-3 has important roles in normal homeostasis and developmental processes, it has been shown to be amplified and overexpressed in breast cancer and to promote malignancy. The malignancy-promoting potential of SRC-3 is diverse and involves both promoting malignant behavior of tumor cells and creating a tumor microenvironment that has an immunosuppressive phenotype. SRC-3 also inhibits the recruitment of tumor-infiltrating lymphocytes with effector function and promotes stemness. Furthermore, SRC-3 is also involved in the development of resistance to hormone therapy and immunotherapy during breast cancer treatment. The versatility of SRC-3 in promoting breast cancer malignancy in this way makes it a good target, and methodical targeting of SRC-3 probably will be important for the success of breast cancer treatment.
Collapse
Affiliation(s)
- Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey;
| | - Garrett M. Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA;
| | - Veysel Tolan
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey;
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece
| |
Collapse
|
44
|
Tang L, Li Q, Chen L, Li X, Gu S, He W, Pan Q, Wang L, Sun J, Yi X, Li Y. IL-21 collaborates with anti-TIGIT to restore NK cell function in chronic HBV infection. J Med Virol 2023; 95:e29142. [PMID: 37815034 DOI: 10.1002/jmv.29142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/29/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023]
Abstract
Available therapies for chronic hepatitis B virus (HBV) infection are not satisfying, and interleukin-21 (IL-21) and checkpoint inhibitors are potential therapeutic options. However, the mechanism underlying IL-21 and checkpoint inhibitors in treating chronic HBV infection is unclear. To explore whether IL-21 and checkpoint inhibitors promote HBV clearance by modulating the function of natural killer (NK) cells, we measured the phenotypes and functions of NK cells in chronic HBV-infected patients and healthy controls on mRNA and protein levels. We found that chronic HBV infection disturbed the transcriptome of NK cells, including decreased expression of KLRK1, TIGIT, GZMA, PRF1, and increased expression of CD69. We also observed altered phenotypes and functions of NK cells in chronic HBV-infected patients, characterized by decreased NKG2D expression, increased TIGIT expression and impaired interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α) production. Furthermore, these alterations cannot be restored by telbivudine treatment but can be partially restored by IL-21 and anti-TIGIT stimulation. IL-21 upregulated the expression of activating receptor CD16, CD69, and NKG2D on NK cells, enhanced IFN-γ production, cytolysis, and proliferation of NK cells, while anti-TIGIT promoted IFN-γ production in CD56dim subset exclusively in chronic HBV infected patients. Additionally, IL-21 was indispensable for anti-TIGIT in HBsAg clearance in mice bearing HBV. It enhanced IFN-γ production in splenic NK cells rather than intrahepatic NK cells, indicating a brand-new mechanism of IL-21 in HBV clearance when combined with anti-TIGIT. Overall, our findings contribute to the design of immunotherapy through enhancing the antiviral efficacy of NK cells in chronic HBV infection.
Collapse
Affiliation(s)
- Libo Tang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Quanrun Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Key Infectious Diseases Laboratory (Preparatory) of Yunnan Provincial Department of Education, Department of Infectious Diseases, School of Clinical Medicine, The First Affiliated Yunnan Provincial Clinical Medical Center (Branch) for Infectious Diseases, Hospital of Dali University, Dali University, Dali, Yunnan, China
| | - Liang Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Infectious Diseases, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Xiaowei Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuqin Gu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiying He
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingqing Pan
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liping Wang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianru Sun
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Yi
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongyin Li
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
45
|
Scopelliti F, Dimartino V, Cattani C, Cavani A. Functional TRPA1 Channels Regulate CD56 dimCD16 + NK Cell Cytotoxicity against Tumor Cells. Int J Mol Sci 2023; 24:14736. [PMID: 37834182 PMCID: PMC10572725 DOI: 10.3390/ijms241914736] [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: 09/04/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Transient receptor potential ankyrin 1 (TRPA1) channels are expressed on the surface of different cell types, including immune cells. However, TRPA1's role in the context of innate and adaptive immune responses has not been fully elucidated so far. In this study, we aimed at investigating the expression and function of TRPA1 channels on NK cells. Among NK cells, TRPA1 was highly expressed by the CD56dimCD16+ subpopulation, but not by CD56brightCD16- cells, as detected by FACS. TRPA1 activation with the potent ligand allyl isothiocyanate (AITC) induces intracellular calcium flux in CD56dimCD16+ cells, which was prevented by the TRPA1 antagonist HC-030031. AITC treatment increased the membrane around NKp44 and strongly decreased CD16 and CD8 expression, while CD158a, CD159a, NKG2d, NKp46 were substantially unaffected. Importantly, AITC increased the granzyme production and CD107 expression and increased NK cell-mediated cytotoxicity towards the K562 cell line and two different melanoma cell lines. In parallel, TRPA1 activation also plays regulatory roles by affecting the survival of NK cells to limit uncontrolled and prolonged NK cell-mediated cytotoxicity. Our results indicate that the activation of TRPA1 is an important regulatory signal for NK cells, and agonists of TRPA1 could be used to strengthen the tumor response of the immune system.
Collapse
Affiliation(s)
- Fernanda Scopelliti
- National Institute for Health, Migration and Poverty INMP/NIHMP, Via di S.Gallicano, 25, 00153 Rome, Italy (C.C.); (C.A.)
| | - Valentina Dimartino
- National Institute for Health, Migration and Poverty INMP/NIHMP, Via di S.Gallicano, 25, 00153 Rome, Italy (C.C.); (C.A.)
- National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Via Portuense 292, 00149 Rome, Italy
| | - Caterina Cattani
- National Institute for Health, Migration and Poverty INMP/NIHMP, Via di S.Gallicano, 25, 00153 Rome, Italy (C.C.); (C.A.)
| | - Andrea Cavani
- National Institute for Health, Migration and Poverty INMP/NIHMP, Via di S.Gallicano, 25, 00153 Rome, Italy (C.C.); (C.A.)
| |
Collapse
|
46
|
Singh S, Barik D, Arukha AP, Prasad S, Mohapatra I, Singh A, Singh G. Small Molecule Targeting Immune Cells: A Novel Approach for Cancer Treatment. Biomedicines 2023; 11:2621. [PMID: 37892995 PMCID: PMC10604364 DOI: 10.3390/biomedicines11102621] [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: 06/28/2023] [Revised: 09/05/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Conventional and cancer immunotherapies encompass diverse strategies to address various cancer types and stages. However, combining these approaches often encounters limitations such as non-specific targeting, resistance development, and high toxicity, leading to suboptimal outcomes in many cancers. The tumor microenvironment (TME) is orchestrated by intricate interactions between immune and non-immune cells dictating tumor progression. An innovative avenue in cancer therapy involves leveraging small molecules to influence a spectrum of resistant cell populations within the TME. Recent discoveries have unveiled a phenotypically diverse cohort of innate-like T (ILT) cells and tumor hybrid cells (HCs) exhibiting novel characteristics, including augmented proliferation, migration, resistance to exhaustion, evasion of immunosurveillance, reduced apoptosis, drug resistance, and heightened metastasis frequency. Leveraging small-molecule immunomodulators to target these immune players presents an exciting frontier in developing novel tumor immunotherapies. Moreover, combining small molecule modulators with immunotherapy can synergistically enhance the inhibitory impact on tumor progression by empowering the immune system to meticulously fine-tune responses within the TME, bolstering its capacity to recognize and eliminate cancer cells. This review outlines strategies involving small molecules that modify immune cells within the TME, potentially revolutionizing therapeutic interventions and enhancing the anti-tumor response.
Collapse
Affiliation(s)
- Shilpi Singh
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Debashis Barik
- Center for Computational Natural Science and Bioinformatics, International Institute of Information Technology, Hyderabad 500032, Telangana, India
| | | | | | - Iteeshree Mohapatra
- Department of Veterinary and Biomedical Sciences, University of Minnesota—Twin Cities, Saint Paul, MN 55108, USA
| | - Amar Singh
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Gatikrushna Singh
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
47
|
Qin Y, Mace EM, Barton JP. An inference model gives insights into innate immune adaptation and repertoire diversity. Proc Natl Acad Sci U S A 2023; 120:e2305859120. [PMID: 37695895 PMCID: PMC10515141 DOI: 10.1073/pnas.2305859120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/08/2023] [Indexed: 09/13/2023] Open
Abstract
The innate immune system is the body's first line of defense against infection. Natural killer (NK) cells, a vital part of the innate immune system, help to control infection and eliminate cancer. Studies have identified a vast array of receptors that NK cells use to discriminate between healthy and unhealthy cells. However, at present, it is difficult to explain how NK cells will respond to novel stimuli in different environments. In addition, the expression of different receptors on individual NK cells is highly stochastic, but the reason for these variegated expression patterns is unclear. Here, we studied the recognition of unhealthy target cells as an inference problem, where NK cells must distinguish between healthy targets with normal variability in ligand expression and ones that are clear "outliers." Our mathematical model fits well with experimental data, including NK cells' adaptation to changing environments and responses to different target cells. Furthermore, we find that stochastic, "sparse" receptor expression profiles are best able to detect a variety of possible threats, in agreement with experimental studies of the NK cell repertoire. While our study was specifically motivated by NK cells, our model is general and could also apply more broadly to explain principles of target recognition for other immune cell types.
Collapse
Affiliation(s)
- Yawei Qin
- Department of Physics and Astronomy, University of California, Riverside, CA92521
| | - Emily M. Mace
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY10032
| | - John P. Barton
- Department of Physics and Astronomy, University of California, Riverside, CA92521
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15260
| |
Collapse
|
48
|
Paolini R, Molfetta R. Dysregulation of DNAM-1-Mediated NK Cell Anti-Cancer Responses in the Tumor Microenvironment. Cancers (Basel) 2023; 15:4616. [PMID: 37760586 PMCID: PMC10527063 DOI: 10.3390/cancers15184616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
NK cells play a pivotal role in anti-cancer immune responses, thanks to the expression of a wide array of inhibitory and activating receptors that regulate their cytotoxicity against transformed cells while preserving healthy cells from lysis. However, NK cells exhibit severe dysfunction in the tumor microenvironment, mainly due to the reduction of activating receptors and the induction or increased expression of inhibitory checkpoint receptors. An activating receptor that plays a central role in tumor recognition is the DNAM-1 receptor. It recognizes PVR and Nectin2 adhesion molecules, which are frequently overexpressed on the surface of cancerous cells. These ligands are also able to trigger inhibitory signals via immune checkpoint receptors that are upregulated in the tumor microenvironment and can counteract DNAM-1 activation. Among them, TIGIT has recently gained significant attention, since its targeting results in improved anti-tumor immune responses. This review aims to summarize how the recognition of PVR and Nectin2 by paired co-stimulatory/inhibitory receptors regulates NK cell-mediated clearance of transformed cells. Therapeutic approaches with the potential to reverse DNAM-1 dysfunction in the tumor microenvironment will be also discussed.
Collapse
Affiliation(s)
| | - Rosa Molfetta
- Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
49
|
Wu X, Huang S, He W, Song M. Emerging insights into mechanisms of trastuzumab resistance in HER2-positive cancers. Int Immunopharmacol 2023; 122:110602. [PMID: 37437432 DOI: 10.1016/j.intimp.2023.110602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/19/2023] [Accepted: 07/02/2023] [Indexed: 07/14/2023]
Abstract
HER2 is an established therapeutic target in breast, gastric, and gastroesophageal junction carcinomas with HER2 overexpression or genomic alterations. The humanized monoclonal antibody trastuzumab targeting HER2 has substantially improved the clinical outcomes of HER2-positive patients, yet the inevitable intrinsic or acquired resistance to trastuzumab limits its clinical benefit, necessitating the elucidation of resistance mechanisms to develop alternate therapeutic strategies. This review presents an overview of trastuzumab resistance mechanisms involving signaling pathways, cellular metabolism, cell plasticity, and tumor microenvironment, particularly discussing the prospects of developing rational combinations to improve patient outcomes.
Collapse
Affiliation(s)
- Xiaoxue Wu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Shuting Huang
- School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weiling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361000, China.
| | - Mei Song
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
50
|
Guo F, Zhang Y, Bai L, Cui J. Natural killer cell therapy targeting cancer stem cells: Old wine in a new bottle. Cancer Lett 2023; 570:216328. [PMID: 37499742 DOI: 10.1016/j.canlet.2023.216328] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/14/2023] [Accepted: 07/22/2023] [Indexed: 07/29/2023]
Abstract
A small proportion of cancer cells that have stem cell-like properties are known as cancer stem cells (CSCs). They can be used to identify malignant tumor phenotypes and patients with poor prognosis. Targeting these cells has been shown to improve the effectiveness of cancer therapies. Owing to the nature of CSCs, they are resistant to conventional treatment methods such as radio- and chemotherapy. Therefore, more effective anti-CSC therapies are required. Immunotherapy, including natural killer (NK) and T cell therapy, has demonstrated the ability to eliminate CSCs. NK cells have demonstrated superior anti-CSC capabilities compared to T cells in recognizing low levels of major histocompatibility complex (MHC) class I expression. However, CSC escape also occurs during NK cell therapy. It is important to determine CSC-specific immune evasion mechanisms and find out potential solutions to optimize NK cell function. Therefore, this review discusses promising strategies that can improve the efficiency of NK cell therapy in treating CSCs, and aims to provide a reference for future research.
Collapse
Affiliation(s)
- Feifei Guo
- Cancer Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Yi Zhang
- Cancer Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Ling Bai
- Cancer Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.
| |
Collapse
|