1
|
Zhang SQ, Thomas F, Fang J, Austgen K, Cowan C, Welstead GG. Universal protection of allogeneic T-cell therapies from natural killer cells via CD300a agonism. Blood Adv 2025; 9:254-264. [PMID: 39368806 DOI: 10.1182/bloodadvances.2024013436] [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: 04/19/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/07/2024] Open
Abstract
ABSTRACT Immunogenicity limits the persistence of off-the-shelf allogeneic cell therapies and transplants. Although ablation of HLA removes most T cell and humoral alloreactivity, no solution has enabled universal protection against the resulting natural killer (NK) cell response. Here, we engineered trans-antigen signaling receptors (TASRs) as a new class of NK inhibitory ligands and discovered CD300a, a previously inaccessible receptor, as a functional target. CD300a TASR outperformed leading alternative strategies in focused screens, including CD47 and HLA-E, and was solely capable of universally protecting allogeneic T cells against a large human cohort (45/45 donors), spanning diverse demographics and NK cell phenotypes. A model allogeneic T-cell therapy coexpressing an anti-CD19 chimeric antigen receptor and CD300a TASR, produced using multiplexed nonviral integration, exhibited enhanced B-cell killing potency under allogeneic immune pressure. CD300 TASR represents a universal solution to NK alloreactivity, broadening the population that could be effectively treated by next-generation allogeneic cell therapies.
Collapse
Affiliation(s)
- Shu-Qi Zhang
- Department of Discovery Biology, Clade Therapeutics, Inc, Boston, MA
- Department of Molecular Immunology, Clade Therapeutics, Inc, Boston, MA
| | - Faith Thomas
- Department of Discovery Biology, Clade Therapeutics, Inc, Boston, MA
| | - Justin Fang
- Department of Discovery Biology, Clade Therapeutics, Inc, Boston, MA
- Department of Discovery Biology, Century Therapeutics, Inc, Boston, MA
- Department of Molecular Immunology, Clade Therapeutics, Inc, Boston, MA
| | - Kathryn Austgen
- Department of Molecular Immunology, Clade Therapeutics, Inc, Boston, MA
| | - Chad Cowan
- Clade Therapeutics, Inc, Boston, MA
- Century Therapeutics, Inc, Philadelphia, PA
| | - G Grant Welstead
- Department of Discovery Biology, Clade Therapeutics, Inc, Boston, MA
- Department of Discovery Biology, Century Therapeutics, Inc, Boston, MA
| |
Collapse
|
2
|
Toh J, Reitsma AJ, Tajima T, Younes SF, Ezeiruaku C, Jenkins KC, Peña JK, Zhao S, Wang X, Lee EYZ, Glass MC, Kalesinskas L, Ganesan A, Liang I, Pai JA, Harden JT, Vallania F, Vizcarra EA, Bhagat G, Craig FE, Swerdlow SH, Morscio J, Dierickx D, Tousseyn T, Satpathy AT, Krams SM, Natkunam Y, Khatri P, Martinez OM. Multi-modal analysis reveals tumor and immune features distinguishing EBV-positive and EBV-negative post-transplant lymphoproliferative disorders. Cell Rep Med 2024; 5:101851. [PMID: 39657667 PMCID: PMC11722118 DOI: 10.1016/j.xcrm.2024.101851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 03/09/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024]
Abstract
The oncogenic Epstein-Barr virus (EBV) can drive tumorigenesis with disrupted host immunity, causing malignancies including post-transplant lymphoproliferative disorders (PTLDs). PTLD can also arise in the absence of EBV, but the biological differences underlying EBV(+) and EBV(-) B cell PTLD and the associated host-EBV-tumor interactions remain poorly understood. Here, we reveal the core differences between EBV(+) and EBV(-) PTLD, characterized by increased expression of genes related to immune processes or DNA interactions, respectively, and the augmented ability of EBV(+) PTLD B cells to modulate the tumor microenvironment through elaboration of monocyte-attracting cytokines/chemokines. We create a reference resource of proteins distinguishing EBV(+) B lymphoma cells from EBV(-) B lymphoma including the immunomodulatory molecules CD300a and CD24, respectively. Moreover, we show that CD300a is essential for maximal survival of EBV(+) PTLD B lymphoma cells. Our comprehensive multi-modal analyses uncover the biological underpinnings of PTLD and offer opportunities for precision therapies.
Collapse
Affiliation(s)
- Jiaying Toh
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA; PhD Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Andrea J Reitsma
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Tetsuya Tajima
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Sheren F Younes
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chimere Ezeiruaku
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Kayla C Jenkins
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Josselyn K Peña
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA; PhD Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shuchun Zhao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xi Wang
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Esmond Y Z Lee
- PhD Program in Stem Cell and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Marla C Glass
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Laurynas Kalesinskas
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA; PhD Program in Biomedical Informatics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ananthakrishnan Ganesan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA; Institute for Computational and Mathematical Engineering, School of Engineering, Stanford University, Stanford, CA, USA
| | - Irene Liang
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Joy A Pai
- PhD Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - James T Harden
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA; PhD Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Francesco Vallania
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Edward A Vizcarra
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| | - Govind Bhagat
- Department of Pathology, Columbia University, New York, NY, USA
| | - Fiona E Craig
- Laboratory of Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Steven H Swerdlow
- Division of Hematopathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Julie Morscio
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium
| | - Daan Dierickx
- Department of Hematology, University Hospitals Leuven, and the Laboratory for Experimental Hematology, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Thomas Tousseyn
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium; Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sheri M Krams
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA; Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA, USA; Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Olivia M Martinez
- Department of Surgery, Division of Abdominal Transplantation, Stanford University School of Medicine, Stanford, CA, USA; Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
3
|
Alsalloum A, Alrhmoun S, Perik-Zavosdkaia O, Fisher M, Volynets M, Lopatnikova J, Perik-Zavodskii R, Shevchenko J, Philippova J, Solovieva O, Zavjalov E, Kurilin V, Shiku H, Silkov A, Sennikov S. Decoding NY-ESO-1 TCR T cells: transcriptomic insights reveal dual mechanisms of tumor targeting in a melanoma murine xenograft model. Front Immunol 2024; 15:1507218. [PMID: 39660132 PMCID: PMC11628372 DOI: 10.3389/fimmu.2024.1507218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
The development of T cell receptor-engineered T cells (TCR-T) targeting intracellular antigens is a promising strategy for treating solid tumors; however, the mechanisms underlying their effectiveness remain poorly understood. In this study, we employed advanced techniques to investigate the functional state of T cells engineered with retroviral vectors to express a TCR specific for the NY-ESO-1 157-165 peptide in the HLA-A*02:01 context. Flow cytometry revealed a predominance of naïve T cells. Gene expression profiling using NanoString technology revealed upregulation of genes encoding chemokine receptors CCR2 and CCR5, indicating enhanced migration towards tumor sites. In the SK-Mel-37 xenograft model, these transduced T cells achieved complete tumor eradication. Furthermore, single-cell RNA sequencing (scRNA-seq) conducted 14 days post-TCR T cell infusion provided a comprehensive analysis of the in vivo adaptation of these cells, identifying a distinct subset of CD8+ effector T cells with an NK cell-like gene expression profile. Our findings indicate that NY-ESO-1 TCR-transduced T cells have the potential to mediate dual antitumor effects through both antigen-independent NK-like and antigen-specific CTL-like responses. This study underscores the potential of NY-ESO-1 TCR-T cells as potent tumor-eradicating agents, highlighting the importance of harnessing their versatile functional capabilities to refine and enhance therapeutic strategies.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Transcriptome
- Immunotherapy, Adoptive/methods
- Cell Line, Tumor
- Melanoma/therapy
- Melanoma/immunology
- Melanoma/genetics
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Xenograft Model Antitumor Assays
- HLA-A2 Antigen/genetics
- HLA-A2 Antigen/immunology
- CD8-Positive T-Lymphocytes/immunology
- Membrane Proteins/genetics
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Gene Expression Profiling
- Neoplasm Proteins
- Peptide Fragments
Collapse
Affiliation(s)
- Alaa Alsalloum
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Saleh Alrhmoun
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Olga Perik-Zavosdkaia
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Marina Fisher
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Marina Volynets
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Julia Lopatnikova
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Roman Perik-Zavodskii
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Julia Shevchenko
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Julia Philippova
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Olga Solovieva
- Center for Collective Use SPF-vivarium ICG SB RAS, Ministry of Science and High Education of Russian Federation, Novosibirsk, Russia
| | - Evgenii Zavjalov
- Center for Collective Use SPF-vivarium ICG SB RAS, Ministry of Science and High Education of Russian Federation, Novosibirsk, Russia
| | - Vasily Kurilin
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Hiroshi Shiku
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Alexander Silkov
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Sergey Sennikov
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Department of Immunology, V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
4
|
Liu Y, Zhao Y, Song H, Li Y, Liu Z, Ye Z, Zhao J, Wu Y, Tang J, Yao M. Metabolic reprogramming in tumor immune microenvironment: Impact on immune cell function and therapeutic implications. Cancer Lett 2024; 597:217076. [PMID: 38906524 DOI: 10.1016/j.canlet.2024.217076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/23/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Understanding of the metabolic reprogramming has revolutionized our insights into tumor progression and potential treatment. This review concentrates on the aberrant metabolic pathways in cancer cells within the tumor microenvironment (TME). Cancer cells differ from normal cells in their metabolic processing of glucose, amino acids, and lipids in order to adapt to heightened biosynthetic and energy needs. These metabolic shifts, which crucially alter lactic acid, amino acid and lipid metabolism, affect not only tumor cell proliferation but also TME dynamics. This review also explores the reprogramming of various immune cells in the TME. From a therapeutic standpoint, targeting these metabolic alterations represents a novel cancer treatment strategy. This review also discusses approaches targeting the regulation of metabolism of different nutrients in tumor cells and influencing the tumor microenvironment to enhance the immune response. In summary, this review summarizes metabolic reprogramming in cancer and its potential as a target for innovative therapeutic strategies, offering fresh perspectives on cancer treatment.
Collapse
Affiliation(s)
- Yuqiang Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yu Zhao
- Department of Thoracic Surgery, Sheng Jing Hospital, China Medical University, Shenyang, Liaoning, 110000, China
| | - Huisheng Song
- Affiliated Qingyuan Hospital, Guangzhou Medica University, Qingyuan People's Hospital, Qingyuan, Guangdong, 511500, China
| | - Yunting Li
- Department of Pediatrics, Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Zihao Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Zhiming Ye
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jianzhu Zhao
- Department of oncology, Sheng Jing Hospital, China Medical University, Shenyang, Liaoning, 110000, China
| | - Yuzheng Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jun Tang
- Department of Thoracic Surgery, Sheng Jing Hospital, China Medical University, Shenyang, Liaoning, 110000, China.
| | - Maojin Yao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Thoracic Surgery and Oncology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|
5
|
Kristenson L, Badami C, Ljungberg A, Islamagic E, Tian Y, Xie G, Hussein BA, Pesce S, Tang KW, Thorén FB. Deletion of the TMEM30A gene enables leukemic cell evasion of NK cell cytotoxicity. Proc Natl Acad Sci U S A 2024; 121:e2316447121. [PMID: 38557174 PMCID: PMC11009675 DOI: 10.1073/pnas.2316447121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024] Open
Abstract
Natural killer (NK) cell immunotherapy has gained attention as a promising strategy for treatment of various malignancies. In this study, we used a genome-wide CRISPR screen to identify genes that provide protection or susceptibility to NK cell cytotoxicity. The screen confirmed the role of several genes in NK cell regulation, such as genes involved in interferon-γ signaling and antigen presentation, as well as genes encoding the NK cell receptor ligands B7-H6 and CD58. Notably, the gene TMEM30A, encoding CDC50A-beta-subunit of the flippase shuttling phospholipids in the plasma membrane, emerged as crucial for NK cell killing. Accordingly, a broad range of TMEM30A knock-out (KO) leukemia and lymphoma cells displayed increased surface levels of phosphatidylserine (PtdSer). TMEM30A KO cells triggered less NK cell degranulation, cytokine production and displayed lower susceptibility to NK cell cytotoxicity. Blockade of PtdSer or the inhibitory receptor TIM-3, restored the NK cell ability to eliminate TMEM30A-mutated cells. The key role of the TIM-3 - PtdSer interaction for NK cell regulation was further substantiated by disruption of the receptor gene in primary NK cells, which significantly reduced the impact of elevated PtdSer in TMEM30A KO leukemic cells. Our study underscores the potential significance of agents targeting the interaction between PtdSer and TIM-3 in the realm of cancer immunotherapy.
Collapse
Affiliation(s)
- Linnea Kristenson
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 90, Sweden
| | - Chiara Badami
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 90, Sweden
| | - Angelica Ljungberg
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
| | - Erna Islamagic
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 90, Sweden
| | - Yarong Tian
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 46, Sweden
| | - Guojiang Xie
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 46, Sweden
| | - Brwa Ali Hussein
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 90, Sweden
| | - Silvia Pesce
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Dipartimento di Medicina Sperimentale, Università di Genova, Genoa16132, Italy
| | - Ka-Wei Tang
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 46, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg413 46, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg413 90, Sweden
| | - Fredrik B. Thorén
- Tumor Immunology (TIMM) Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg413 90, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg413 90, Sweden
| |
Collapse
|
6
|
Li S, Wang T, Xiao X, Zheng X, Sun H, Sun R, Ma H, Tian Z, Zheng X. Blockade of CD300A enhances the ability of human NK cells to lyse hematologic malignancies. Cancer Biol Med 2024; 21:j.issn.2095-3941.2023.0341. [PMID: 38425216 PMCID: PMC11033714 DOI: 10.20892/j.issn.2095-3941.2023.0341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 12/11/2023] [Indexed: 03/02/2024] Open
Abstract
OBJECTIVE The human cluster of differentiation (CD)300A, a type-I transmembrane protein with immunoreceptor tyrosine-based inhibitory motifs, was investigated as a potential immune checkpoint for human natural killer (NK) cells targeting hematologic malignancies (HMs). METHODS We implemented a stimulation system involving the CD300A ligand, phosphatidylserine (PS), exposed to the outer surface of malignant cells. Additionally, we utilized CD300A overexpression, a CD300A blocking system, and a xenotransplantation model to evaluate the impact of CD300A on NK cell efficacy against HMs in in vitro and in vivo settings. Furthermore, we explored the association between CD300A and HM progression in patients. RESULTS Our findings indicated that PS hampers the function of NK cells. Increased CD300A expression inhibited HM lysis by NK cells. CD300A overexpression shortened the survival of HM-xenografted mice by impairing transplanted NK cells. Blocking PS-CD300A signals with antibodies significantly amplified the expression of lysis function-related proteins and effector cytokines in NK cells, thereby augmenting the ability to lyse HMs. Clinically, heightened CD300A expression correlated with shorter survival and an "exhausted" phenotype of intratumoral NK cells in patients with HMs or solid tumors. CONCLUSIONS These results propose CD300A as a potential target for invigorating NK cell-based treatments against HMs.
Collapse
Affiliation(s)
- Shuangcheng Li
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Tianci Wang
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Xinghui Xiao
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Xiaodong Zheng
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Haoyu Sun
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Rui Sun
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Hongdi Ma
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
| | - Zhigang Tian
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Hefei TG ImmunoPharma Corporation Limited, Hefei 230601, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Beijing 100864, China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaohu Zheng
- Hefei National Research Center for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
7
|
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
|
8
|
Koizumi H, Nakahashi-Oda C, Lyu W, Yamashita-Kanemaru Y, Tabuchi K, Shibuya K, Shibuya A. Development of Monoclonal Antibodies Specific to Either CD300A R111 or CD300A Q111 or Both. Monoclon Antib Immunodiagn Immunother 2023; 42:182-185. [PMID: 37902989 DOI: 10.1089/mab.2023.0017] [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: 11/01/2023] Open
Abstract
CD300A is a member of the CD300 immunoglobulin (Ig)-like receptor family consisting of eight molecules in humans, all of which contain one Ig-like domain in the extracellular portion. Upon binding its ligand phosphatidylserine or phosphatidylethanolamine, CD300A mediates an inhibitory signal through the immunoreceptor tyrosine-based inhibitory motif in the cytoplasmic portion. The CD300 family molecules are highly homologous to each other. In addition, CD300A has a single nucleotide polymorphism (rs2272111), which is a nonsense mutation encoding glutamine (CD300AQ111) instead of arginine (CD300AR111) at residue 111 in the Ig-like domain of CD300A. In this study, we successfully generated monoclonal antibodies (mAbs) specific to either CD300AR111 or CD300AQ111 or both. These mAbs are useful for the analysis of CD300A genotype by flow cytometry and the development of an antibody drug for the treatment of various diseases.
Collapse
Affiliation(s)
- Hitoshi Koizumi
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Otolaryngology-Head and Neck Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Chigusa Nakahashi-Oda
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Ibaraki, Japan
| | - Wenxin Lyu
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | | | - Keiji Tabuchi
- Department of Otolaryngology-Head and Neck Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kazuko Shibuya
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Ibaraki, Japan
| | - Akira Shibuya
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- TNAX Biopharma Corporation, Tsukuba, Ibaraki, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), Tsukuba, Japan
| |
Collapse
|
9
|
Khaleafi R, Zeleznjak J, Cordela S, Drucker S, Rovis TL, Jonjic S, Bar-On Y. Reovirus infection of tumor cells reduces the expression of NKG2D ligands, leading to impaired NK-cell cytotoxicity and functionality. Front Immunol 2023; 14:1231782. [PMID: 37753084 PMCID: PMC10518469 DOI: 10.3389/fimmu.2023.1231782] [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: 05/30/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023] Open
Abstract
In recent years, reoviruses have been of major interest in immunotherapy because of their oncolytic properties. Preclinical and clinical trials, in which reovirus was used for the treatment of melanoma and glioblastoma, have paved the way for future clinical use of reovirus. However, little is known about how reovirus infection affects the tumor microenvironment and immune response towards infected tumor cells. Studies have shown that reovirus can directly stimulate natural killer (NK) cells, but how reovirus affects cellular ligands on tumor cells, which are ultimately key to tumor recognition and elimination by NK cells, has not been investigated. We tested how reovirus infection affects the binding of the NK Group-2 member D (NKG2D) receptor, which is a dominant mediator of NK cell anti-tumor activity. Using models of human-derived melanoma and glioblastoma tumors, we demonstrated that NKG2D ligands are downregulated in tumor cells post-reovirus-infection due to the impaired translation of these ligands in reovirus-infected cells. Moreover, we showed that downregulation of NKG2D ligands significantly impaired the binding of NKG2D to infected tumor cells. We further demonstrated that reduced recognition of NKG2D ligands significantly alters NK cell anti-tumor cytotoxicity in human primary NK cells and in the NK cell line NK-92. Thus, this study provides novel insights into reovirus-host interactions and could lead to the development of novel reovirus-based therapeutics that enhance the anti-tumor immune response.
Collapse
Affiliation(s)
- Raghad Khaleafi
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Jelena Zeleznjak
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Sapir Cordela
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Shani Drucker
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tihana Lenac Rovis
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjic
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Yotam Bar-On
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
10
|
Liu X, Liu H, Deng Y. Efferocytosis: An Emerging Therapeutic Strategy for Type 2 Diabetes Mellitus and Diabetes Complications. J Inflamm Res 2023; 16:2801-2815. [PMID: 37440994 PMCID: PMC10335275 DOI: 10.2147/jir.s418334] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Increasing evidence indicates that chronic, low-grade inflammation is a significant contributor to the fundamental pathogenesis of type 2 diabetes mellitus (T2DM). Efferocytosis, an effective way to eliminate apoptotic cells (ACs), plays a critical role in inflammation resolution. Massive accumulation of ACs and the proliferation of persistent inflammation caused by defective efferocytosis have been proven to be closely associated with pancreatic islet β cell destruction, adipose tissue inflammation, skeletal muscle dysfunction, and liver metabolism abnormalities, which together are considered the most fundamental pathological mechanism underlying T2DM. Therefore, here we outline the association between the molecular mechanisms of efferocytosis in glucose homeostasis, T2DM, and its complications, and we analyzed the present constraints and potential future prospects for therapeutic targets in T2DM and its complications.
Collapse
Affiliation(s)
- Xun Liu
- Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Hua Liu
- Southern Theater General Hospital of the Chinese People’s Liberation Army, Guangzhou, Guangdong, 510010, People’s Republic of China
| | - Yihui Deng
- Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| |
Collapse
|
11
|
Cruz Amaya J, Walcheck B, Smith-Gagen J, Lombardi VC, Hudig D. Detection of Antibody-Dependent Cell-Mediated Cytotoxicity-Supporting Antibodies by NK-92-CD16A Cell Externalization of CD107a: Recognition of Antibody Afucosylation and Assay Optimization. Antibodies (Basel) 2023; 12:44. [PMID: 37489366 PMCID: PMC10366760 DOI: 10.3390/antib12030044] [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: 05/24/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/26/2023] Open
Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC) by natural killer (NK) lymphocytes eliminates cells infected with viruses. Anti-viral ADCC requires three components: (1) antibody; (2) effector lymphocytes with the Fc-IgG receptor CD16A; and (3) viral proteins in infected cell membranes. Fc-afucosylated antibodies bind with greater affinity to CD16A than fucosylated antibodies; individuals' variation in afucosylation contributes to differences in ADCC. Current assays for afucosylated antibodies involve expensive methods. We report an improved bioassay for antibodies that supports ADCC, which encompasses afucosylation. This assay utilizes the externalization of CD107a by NK-92-CD16A cells after antibody recognition. We used anti-CD20 monoclonal antibodies, GA101 WT or glycoengineered (GE), 10% or ~50% afucosylated, and CD20-positive Raji target cells. CD107a increased detection 7-fold compared to flow cytometry to detect Raji-bound antibodies. WT and GE antibody effective concentrations (EC50s) for CD107a externalization differed by 20-fold, with afucosylated GA101-GE more detectable. The EC50s for CD107a externalization vs. 51Cr cell death were similar for NK-92-CD16A and blood NK cells. Notably, the % CD107a-positive cells were negatively correlated with dead Raji cells and were nearly undetectable at high NK:Raji ratios required for cytotoxicity. This bioassay is very sensitive and adaptable to assess anti-viral antibodies but unsuitable as a surrogate assay to monitor cell death after ADCC.
Collapse
Affiliation(s)
- Judith Cruz Amaya
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Bruce Walcheck
- Department of Veterinary and Biological Sciences, Center for Immunology and Masonic Cancer Center, University of Minnesota, 295J AS/VM Building, 1988 Fitch Avenue, Saint Paul, MN 55108, USA
| | - Julie Smith-Gagen
- School of Community Health Sciences, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Vincent C. Lombardi
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| | - Dorothy Hudig
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, 1664 N. Virginia St., Reno, NV 89557, USA
| |
Collapse
|
12
|
Wang D, Dai J, Suo C, Wang S, Zhang Y, Chen X. Molecular subtyping of esophageal squamous cell carcinoma by large-scale transcriptional profiling: Characterization, therapeutic targets, and prognostic value. Front Genet 2022; 13:1033214. [DOI: 10.3389/fgene.2022.1033214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/28/2022] [Indexed: 11/11/2022] Open
Abstract
The tumor heterogeneity of the transcriptional profiles is independent of genetic variation. Several studies have successfully identified esophageal squamous cell carcinoma (ESCC) subtypes based on the somatic mutation profile and copy number variations on the genome. However, transcriptome-based classification is limited. In this study, we classified 141 patients with ESCC into three subtypes (Subtype 1, Subtype 2, and Subtype 3) via tumor sample gene expression profiling. Differential gene expression (DGE) analysis of paired tumor and normal samples for each subtype revealed significant difference among subtypes. Moreover, the degree of change in the expression levels of most genes gradually increased from Subtype 1 to Subtype 3. Gene set enrichment analysis (GSEA) identified the representative pathways in each subtype: Subtype 1, abnormal Wnt signaling pathway activation; Subtype 2, inhibition of glycogen metabolism; and Subtype 3, downregulation of neutrophil degranulation process. Weighted gene co-expression network analysis (WGCNA) was used to elucidate the finer regulation of biological pathways and discover hub genes. Subsequently, nine hub genes (CORO1A, CD180, SASH3, CD52, CD300A, CD14, DUSP1, KIF14, and MCM2) were validated to be associated with survival in ESCC based on the RNA sequencing (RNA-seq) data from The Cancer Genome Atlas (TCGA) database. The clustering analysis of ESCC granted better understanding of the molecular characteristics of ESCC and led to the discover of new potential therapeutic targets that may contribute to the clinical treatment of ESCC.
Collapse
|
13
|
Yu G, Lu W, Chen X, Li Y, Long J, Zheng Z, Yin C, Xu D. Single-cell RNA sequencing to explore composition of peripheral blood NK cells in patients with chronic myeloid leukemia in treatment-free remission. Leuk Lymphoma 2022; 63:2604-2615. [PMID: 35695125 DOI: 10.1080/10428194.2022.2086243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This study was to explore the role of NK cell subsets and gene expression in maintaining TFR status. We identified six types of NK cells in the PBMCs over both groups (healthy controls and patients with TFR). Gene Oncology analysis showed that up regulated genes were enriched in the categories of "immune response," "reaction to tumor cells," and "cytolysis." In addition, we found that the three NK cell subsets, mature and terminal NK cells, CD56 bright NK cells, and transitional NK cells, contained many significantly up regulated genes in both groups, and that CD56 bright NK cells and transitional NK cells in patients with CML-TFR were in a proliferating and activated state. Through single-cell RNA sequencing analysis, we confirmed that the mature and terminal, CD56 bright, and transitional subsets of NK cells play an indispensable role in maintaining TFR in patients with CML.
Collapse
Affiliation(s)
- Guopan Yu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weixiang Lu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaofan Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanlin Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaxin Long
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhongxin Zheng
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changxin Yin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dan Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Natural killer cells and immune-checkpoint inhibitor therapy: Current knowledge and new challenges. Mol Ther Oncolytics 2022; 24:26-42. [PMID: 34977340 PMCID: PMC8693432 DOI: 10.1016/j.omto.2021.11.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The discovery of immune checkpoints (ICs) and the development of specific blockers to relieve immune effector cells from this inhibiting mechanism has changed the view of anti-cancer therapy. In addition to cytotoxic T lymphocyte antigen 4 (CTLA4) and programmed death 1 (PD1), classical ICs of T lymphocytes and recently described also on a fraction of natural killer (NK) cells, several NK cell receptors, including killer immunoglobulin-like inhibitory receptors (KIRs) and NGK2A, have been recognized as checkpoint members typical of the NK cell population. This offers the opportunity of a dual-checkpoint inhibition approach, targeting classical and non-classical ICs and leading to a synergistic therapeutic effect. In this review, we will overview and discuss this new perspective, focusing on the most relevant candidates for this role among the variety of potential NK ICs. Beside listing and defining classical ICs expressed also by NK cells, or non-classical ICs either on T or on NK cells, we will address their role in NK cell survival, chronic stimulation or functional exhaustion, and the potential relevance of this phenomenon on anti-tumor immune response. Furthermore, NK ICs will be proposed as possible new targets for the development of efficient combined immunotherapy, not forgetting the relevant concerns that may be raised on NK IC blockade. Finally, the impact of epigenetic drugs in such a complex therapeutic picture will be briefly addressed.
Collapse
|
15
|
Horses for Courses in the Era of CARs: Advancing CAR T and CAR NK Cell Therapies. J Pers Med 2021; 11:jpm11111182. [PMID: 34834534 PMCID: PMC8621371 DOI: 10.3390/jpm11111182] [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: 10/05/2021] [Revised: 11/01/2021] [Accepted: 11/08/2021] [Indexed: 11/23/2022] Open
Abstract
The adoptive transfer of allogeneic CAR NK cells holds great promise as an anticancer modality due to the relative ease of manufacturing and genetic modification of NK cells, which translates into affordable pricing. Compared to the pronounced efficacy of CAR T cell therapy in the treatment of B cell malignancies, rigorous clinical and preclinical assessment of the antitumor properties of CAR NK cells has been lagging behind. In this brief review, we summarize the biological features of NK cells that may help define the therapeutic niche of CAR NK cells as well as create more potent NK cell-based anticancer products. In addition, we compare T cells and NK cells as the carriers of CARs using the data of single-cell transcriptomic analysis.
Collapse
|
16
|
Karmakar S, Pal P, Lal G. Key Activating and Inhibitory Ligands Involved in the Mobilization of Natural Killer Cells for Cancer Immunotherapies. Immunotargets Ther 2021; 10:387-407. [PMID: 34754837 PMCID: PMC8570289 DOI: 10.2147/itt.s306109] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are the most potent arm of the innate immune system and play an important role in immunity, alloimmunity, autoimmunity, and cancer. NK cells recognize “altered-self” cells due to oncogenic transformation or stress due to viral infection and target to kill them. The effector functions of NK cells depend on the interaction of the activating and inhibitory receptors on their surface with their cognate ligand expressed on the target cells. These activating and inhibitory receptors interact with major histocompatibility complex I (MHC I) expressed on the target cells and make decisions to mount an immune response. NK cell immune response includes cytolytic activity and secretion of cytokines to help with the ongoing immune response. The advancement of our knowledge on the expression of inhibitory and activating molecules led us to exploit these molecules in the treatment of cancer. This review discusses the importance of activating and inhibitory receptors on NK cells and their clinical importance in cancer immunotherapy.
Collapse
Affiliation(s)
- Surojit Karmakar
- National Centre for Cell Science (NCCS), Pune, MH, 411007, India
| | - Pradipta Pal
- National Centre for Cell Science (NCCS), Pune, MH, 411007, India
| | - Girdhari Lal
- National Centre for Cell Science (NCCS), Pune, MH, 411007, India
| |
Collapse
|
17
|
Buckle I, Guillerey C. Inhibitory Receptors and Immune Checkpoints Regulating Natural Killer Cell Responses to Cancer. Cancers (Basel) 2021; 13:cancers13174263. [PMID: 34503073 PMCID: PMC8428224 DOI: 10.3390/cancers13174263] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Recent years marked the discovery and increased understanding of the role immune checkpoints play in immunity against cancer. This has revolutionized cancer treatment, saving the lives of many patients. For numerous years the spotlight of success has been directed towards T cells; however, it is now appreciated that other cells play vital roles in this protection. In this review we focused on cytotoxic lymphocytes Natural Killer (NK) cells, which are known to be well equipped in the fight against cancer. We explored the role of well-described and newly emerging inhibitory receptors, including immune checkpoints in regulating NK cell activity against cancer. The knowledge summarized in this review should guide the development of immunotherapies targeting inhibitory receptors with the aim of restoring NK cell responses in cancer patients. Abstract The discovery of immune checkpoints provided a breakthrough for cancer therapy. Immune checkpoints are inhibitory receptors that are up-regulated on chronically stimulated lymphocytes and have been shown to hinder immune responses to cancer. Monoclonal antibodies against the checkpoint molecules PD-1 and CTLA-4 have shown early clinical success against melanoma and are now approved to treat various cancers. Since then, the list of potential candidates for immune checkpoint blockade has dramatically increased. The current paradigm stipulates that immune checkpoint blockade therapy unleashes pre-existing T cell responses. However, there is accumulating evidence that some of these immune checkpoint molecules are also expressed on Natural Killer (NK) cells. In this review, we summarize our latest knowledge about targetable NK cell inhibitory receptors. We discuss the HLA-binding receptors KIRS and NKG2A, receptors binding to nectin and nectin-like molecules including TIGIT, CD96, and CD112R, and immune checkpoints commonly associated with T cells such as PD-1, TIM-3, and LAG-3. We also discuss newly discovered pathways such as IL-1R8 and often overlooked receptors such as CD161 and Siglecs. We detail how these inhibitory receptors might regulate NK cell responses to cancer, and, where relevant, we discuss their implications for therapeutic intervention.
Collapse
|
18
|
Formentini M, Navas A, Hassouneh F, Lopez-Sejas N, Alonso C, Tarazona R, Solana R, Pera A. Impact of CMV and age on T cell subsets defined by CD161, CD300a, and/or CD57 expression in healthy Andalusians. J Gerontol A Biol Sci Med Sci 2021; 76:1946-1953. [PMID: 33993242 DOI: 10.1093/gerona/glab140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Indexed: 11/14/2022] Open
Abstract
Immunosenescence affects innate and adaptive immunity impairing the response to pathogens and vaccines. Chronic infection with cytomegalovirus (CMV) has been shown to drive 'early immunosenescence' and can considerably impact both the function and phenotype of immune cells, especially T cells. We have previously shown that the expression of CD57, CD300a, and CD161 was differentially affected by age and chronic CMV infection, indicating that these markers are a hallmark of CMV infection and T cell ageing. The aim of this present study was to clarify whether these three markers define distinct T cell subpopulations with a specific functional and molecular signature. Specifically, we analyzed the effect of age and chronic CMV infection on the functionality of T cells according to CD161, CD300a, and CD57 expression. We found that these markers defined different T cell subsets, both at the phenotypic and functional levels. CD57 was the best biomarker for CD4+ T cell cytotoxicity and was a hallmark of CMV infection. CD300a+ T cells were heterogeneous and included different cell subsets. The population of CD161+ T cells dramatically decreased with age, independently of CMV infection, and represented a sign of age-associated immune system alterations. The latter could contribute to an increased risk of autoimmune disease and infection in older adults. Our results underline the importance of better understanding the factors involved in the immunosenescence process to be able to uncover new biomarkers and open new avenues for the investigation and development of novel age-related disease therapies.
Collapse
Affiliation(s)
- Martina Formentini
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
| | - Ana Navas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Immunology and Allergy service, Reina Sofia University Hospital, Córdoba, Spain
| | - Fakhri Hassouneh
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
| | - Nelson Lopez-Sejas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
| | - Corona Alonso
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Immunology and Allergy service, Reina Sofia University Hospital, Córdoba, Spain
| | - Raquel Tarazona
- Immunology Unit, Department of Physiology, University of Extremadura, Cáceres, Spain
| | - Rafael Solana
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Immunology and Allergy service, Reina Sofia University Hospital, Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
| | - Alejandra Pera
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
| |
Collapse
|
19
|
Cao Y, Ao T, Wang X, Wei W, Fan J, Tian X. CD300a and CD300f molecules regulate the function of leukocytes. Int Immunopharmacol 2021; 93:107373. [PMID: 33548578 DOI: 10.1016/j.intimp.2021.107373] [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: 11/19/2020] [Revised: 01/02/2021] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
The CD300 molecule family is a type I transmembrane glycoprotein expressed on cell membrane of human and other mammals, and of its eight members, only CD300a and CD300f are classified as inhibitory receptors. CD300a and CD300f play an important role in regulating the function of leukocytes, such as activation, proliferation, differentiation, migration and immunity function. They are considered as potential targets for studying the development and progression of inflammation, infection and other diseases. Here, we review the expression and regulatory mechanisms of CD300a and CD300f on leukocytes, as well as their effects on relevant diseases.
Collapse
Affiliation(s)
- Yue Cao
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Tianrang Ao
- Department of Cardiology, Peking Union Medical College Hospital, Tsinghua University, Beijing 100730, China
| | - Xiaohong Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Wumei Wei
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Jun Fan
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaohong Tian
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
20
|
Soteriou C, Kalli AC, Connell SD, Tyler AII, Thorne JL. Advances in understanding and in multi-disciplinary methodology used to assess lipid regulation of signalling cascades from the cancer cell plasma membrane. Prog Lipid Res 2020; 81:101080. [PMID: 33359620 DOI: 10.1016/j.plipres.2020.101080] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/31/2022]
Abstract
The lipid bilayer is a functional component of cells, forming a stable platform for the initiation of key biological processes, including cell signalling. There are distinct changes in the lipid composition of cell membranes during oncogenic transformation resulting in aberrant activation and inactivation of signalling transduction pathways. Studying the role of the cell membrane in cell signalling is challenging, since techniques are often limited to by timescale, resolution, sensitivity, and averaging. To overcome these limitations, combining 'computational', 'wet-lab' and 'semi-dry' approaches offers the best opportunity to resolving complex biological processes involved in membrane organisation. In this review, we highlight analytical tools that have been applied for the study of cell signalling initiation from the cancer cell membranes through computational microscopy, biological assays, and membrane biophysics. The cancer therapeutic potential of extracellular membrane-modulating agents, such as cholesterol-reducing agents is also discussed, as is the need for future collaborative inter-disciplinary research for studying the role of the cell membrane and its components in cancer therapy.
Collapse
Affiliation(s)
- C Soteriou
- School of Food Science and Nutrition, University of Leeds, Leeds LS29JT, UK; Leeds Institute of Cardiovascular and Metabolic Medicine and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK; Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK
| | - A C Kalli
- Leeds Institute of Cardiovascular and Metabolic Medicine and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - S D Connell
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK
| | - A I I Tyler
- School of Food Science and Nutrition, University of Leeds, Leeds LS29JT, UK
| | - J L Thorne
- School of Food Science and Nutrition, University of Leeds, Leeds LS29JT, UK.
| |
Collapse
|
21
|
Evasion of the Cell-Mediated Immune Response by Alphaherpesviruses. Viruses 2020; 12:v12121354. [PMID: 33256093 PMCID: PMC7761393 DOI: 10.3390/v12121354] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022] Open
Abstract
Alphaherpesviruses cause various diseases and establish life-long latent infections in humans and animals. These viruses encode multiple viral proteins and miRNAs to evade the host immune response, including both innate and adaptive immunity. Alphaherpesviruses evolved highly advanced immune evasion strategies to be able to replicate efficiently in vivo and produce latent infections with recurrent outbreaks. This review describes the immune evasion strategies of alphaherpesviruses, especially against cytotoxic host immune responses. Considering these strategies, it is important to evaluate whether the immune evasion mechanisms in cell cultures are applicable to viral propagation and pathogenicity in vivo. This review focuses on cytotoxic T lymphocytes (CTLs), natural killer cells (NK cells), and natural killer T cells (NKT cells), which are representative immune cells that directly damage virus-infected cells. Since these immune cells recognize the ligands expressed on their target cells via specific activating and/or inhibitory receptors, alphaherpesviruses make several ligands that may be targets for immune evasion. In addition, alphaherpesviruses suppress the infiltration of CTLs by downregulating the expression of chemokines at infection sites in vivo. Elucidation of the alphaherpesvirus immune evasion mechanisms is essential for the development of new antiviral therapies and vaccines.
Collapse
|
22
|
Szlasa W, Zendran I, Zalesińska A, Tarek M, Kulbacka J. Lipid composition of the cancer cell membrane. J Bioenerg Biomembr 2020; 52:321-342. [PMID: 32715369 PMCID: PMC7520422 DOI: 10.1007/s10863-020-09846-4] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022]
Abstract
Cancer cell possesses numerous adaptations to resist the immune system response and chemotherapy. One of the most significant properties of the neoplastic cells is the altered lipid metabolism, and consequently, the abnormal cell membrane composition. Like in the case of phosphatidylcholine, these changes result in the modulation of certain enzymes and accumulation of energetic material, which could be used for a higher proliferation rate. The changes are so prominent, that some lipids, such as phosphatidylserines, could even be considered as the cancer biomarkers. Additionally, some changes of biophysical properties of cell membranes lead to the higher resistance to chemotherapy, and finally to the disturbances in signalling pathways. Namely, the increased levels of certain lipids, like for instance phosphatidylserine, lead to the attenuation of the immune system response. Also, changes in lipid saturation prevent the cells from demanding conditions of the microenvironment. Particularly interesting is the significance of cell membrane cholesterol content in the modulation of metastasis. This review paper discusses the roles of each lipid type in cancer physiology. The review combined theoretical data with clinical studies to show novel therapeutic options concerning the modulation of cell membranes in oncology.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Iga Zendran
- Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | | | - Mounir Tarek
- Université de Lorraine, CNRS, LPCT, F-54000, Nancy, France
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wrocław, Poland.
| |
Collapse
|
23
|
Sivori S, Della Chiesa M, Carlomagno S, Quatrini L, Munari E, Vacca P, Tumino N, Mariotti FR, Mingari MC, Pende D, Moretta L. Inhibitory Receptors and Checkpoints in Human NK Cells, Implications for the Immunotherapy of Cancer. Front Immunol 2020; 11:2156. [PMID: 33013909 PMCID: PMC7494755 DOI: 10.3389/fimmu.2020.02156] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/07/2020] [Indexed: 12/30/2022] Open
Abstract
The highly destructive mechanisms by which the immune system faces microbial infections is under the control of a series of inhibitory receptors. While most of these receptors prevent unwanted/excessive responses of individual effector cells, others play a more general role in immunity, acting as true inhibitory checkpoints controlling both innate and adaptive immunity. Regarding human NK cells, their function is finely regulated by HLA-class I-specific inhibitory receptors which allow discrimination between HLA-I+, healthy cells and tumor or virus-infected cells displaying loss or substantial alterations of HLA-I molecules, including allelic losses that are sensed by KIRs. A number of non-HLA-specific receptors have been identified which recognize cell surface or extracellular matrix ligands and may contribute to the physiologic control of immune responses and tolerance. Among these receptors, Siglec 7 (p75/AIRM-1), LAIR-1 and IRp60, recognize ligands including sialic acids, extracellular matrix/collagen or aminophospholipids, respectively. These ligands may be expressed at the surface of tumor cells, thus inhibiting NK cell function. Expression of the PD-1 checkpoint by NK cells requires particular cytokines (IL-15, IL-12, IL-18) together with cortisol, a combination that may occur in the microenvironment of different tumors. Blocking of single or combinations of inhibitory receptors unleashes NK cells and restore their anti-tumor activity, with obvious implications for tumor immunotherapy.
Collapse
Affiliation(s)
- Simona Sivori
- Department of Experimental Medicine (DIMES) and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Mariella Della Chiesa
- Department of Experimental Medicine (DIMES) and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Simona Carlomagno
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Linda Quatrini
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Enrico Munari
- Department of Pathology, IRCCS Sacro Cuore Don Calabria, Negrar, Italy
| | - Paola Vacca
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Nicola Tumino
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | | | - Maria Cristina Mingari
- Department of Experimental Medicine (DIMES) and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.,UOC Immunology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniela Pende
- UOC Immunology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| |
Collapse
|
24
|
Vitallé J, Terrén I, Gamboa-Urquijo L, Orrantia A, Tarancón-Díez L, Genebat M, Leal M, Ruiz-Mateos E, Borrego F, Zenarruzabeitia O. Polyfunctional HIV-1 specific response by CD8+ T lymphocytes expressing high levels of CD300a. Sci Rep 2020; 10:6070. [PMID: 32269232 PMCID: PMC7142067 DOI: 10.1038/s41598-020-63025-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/17/2020] [Indexed: 01/12/2023] Open
Abstract
CD300a receptor is found on different CD8+ T cell subsets and its expression has been associated to a more cytotoxic molecular signature. CD300a has an important role in some viral infections and its expression levels are known to be modulated by human immunodeficiency virus (HIV)−1 infection on several cell types. The main objective of this work was to investigate CD300a expression and its regulation during HIV-1 specific CD8+ T cell responses. CD300a receptor expression was analysed by multiparametric flow cytometry on CD8+ T lymphocytes from HIV negative donors, naive HIV-1+ individuals and HIV-1+ subjects under suppressive combined antiretroviral therapy (cART). HIV-1 specific CD8+ T cell response was studied by stimulating cells with HIV-1 derived peptides or with a Gag HIV-1 peptide. Our results showed that HIV-1 specific CD8+ T cells expressing higher levels of CD300a were more polyfunctional showing an increased degranulation and cytokine production. Moreover, we observed an up-regulation of CD300a expression after Gag HIV-1 peptide stimulation. Finally, our results demonstrated an inverse correlation between CD300a expression on CD8+ T lymphocytes and HIV disease progression markers. In conclusion, CD300a expression is associated to a better and more polyfunctional HIV-1 specific CD8+ T cell response.
Collapse
Affiliation(s)
- Joana Vitallé
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, 48903, Barakaldo, Spain
| | - Iñigo Terrén
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, 48903, Barakaldo, Spain
| | - Leire Gamboa-Urquijo
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, 48903, Barakaldo, Spain
| | - Ane Orrantia
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, 48903, Barakaldo, Spain
| | - Laura Tarancón-Díez
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, University of Seville, CSIC, 41013, Seville, Spain.,Laboratory of Molecular Immuno-Biology, Gregorio Marañón University Hospital, Health Research Institute, 28007, Madrid, Spain
| | - Miguel Genebat
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, University of Seville, CSIC, 41013, Seville, Spain
| | - Manuel Leal
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, University of Seville, CSIC, 41013, Seville, Spain.,Internal Medicine Service, Santa Ángela de la Cruz Viamed Hospital, 41014, Sevilla, Spain
| | - Ezequiel Ruiz-Mateos
- Clinic Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, University of Seville, CSIC, 41013, Seville, Spain
| | - Francisco Borrego
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, 48903, Barakaldo, Spain.,Ikerbasque, Basque Foundation for Science, 48013, Bilbao, Spain
| | - Olatz Zenarruzabeitia
- Biocruces Bizkaia Health Research Institute, Immunopathology Group, 48903, Barakaldo, Spain.
| |
Collapse
|
25
|
Glassman FY, Dingman R, Yau HC, Balu-Iyer SV. Biological Function and Immunotherapy Utilizing Phosphatidylserine-based Nanoparticles. Immunol Invest 2020; 49:858-874. [PMID: 32204629 DOI: 10.1080/08820139.2020.1738456] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Phosphatidylserine (PS) is a naturally occurring anionic phospholipid that is primarily located in the inner leaflet of eukaryotic cell membranes. The role of PS during apoptosis is one of the most studied biological functions of PS. Externalization of PS during apoptosis mediates an "eat me" signal for phagocytic uptake, leading to clearance of apoptotic cells and thus maintain self-tolerance by immunological ignorance. However, an emerging view is that PS exposure-mediated cellular uptake is not an immunologically silent event, but rather promoting an active tolerance towards self and foreign proteins. This biological property of PS has been exploited by parasites and viruses in order to evade immune surveillance of the host immune system. Further, this novel immune regulatory property of PS that results in tolerance induction can be harnessed for clinical applications, such as to treat autoimmune conditions and to reduce immunogenicity of therapeutic proteins. This review attempts to provide an overview of the biological functions of PS in the immune response and its potential therapeutic applications.
Collapse
Affiliation(s)
- Fiona Y Glassman
- Department of Pharmaceutical Sciences, University at Buffalo, the State University of New York , Buffalo, New York, USA.,Clinical Pharmacology and Pharmacometrics, Currently at CSL Behring , King of Prussia, Pennsylvania, USA
| | - Robert Dingman
- Department of Pharmaceutical Sciences, University at Buffalo, the State University of New York , Buffalo, New York, USA
| | - Helena C Yau
- Department of Film and Media Studies, Washington University in St. Louis , St. Louis, Missouri, USA
| | - Sathy V Balu-Iyer
- Department of Pharmaceutical Sciences, University at Buffalo, the State University of New York , Buffalo, New York, USA
| |
Collapse
|
26
|
Terrén I, Orrantia A, Mikelez-Alonso I, Vitallé J, Zenarruzabeitia O, Borrego F. NK Cell-Based Immunotherapy in Renal Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12020316. [PMID: 32013092 PMCID: PMC7072691 DOI: 10.3390/cancers12020316] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/14/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells are cytotoxic lymphocytes that are able to kill tumor cells without prior sensitization. It has been shown that NK cells play a pivotal role in a variety of cancers, highlighting their relevance in tumor immunosurveillance. NK cell infiltration has been reported in renal cell carcinoma (RCC), the most frequent kidney cancer in adults, and their presence has been associated with patients’ survival. However, the role of NK cells in this disease is not yet fully understood. In this review, we summarize the biology of NK cells and the mechanisms through which they are able to recognize and kill tumor cells. Furthermore, we discuss the role that NK cells play in renal cell carcinoma, and review current strategies that are being used to boost and exploit their cytotoxic capabilities.
Collapse
Affiliation(s)
- Iñigo Terrén
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Ane Orrantia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Idoia Mikelez-Alonso
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
- CIC biomaGUNE, 20014 Donostia-San Sebastián, Spain
| | - Joana Vitallé
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
| | - Francisco Borrego
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain; (I.T.); (A.O.); (I.M.-A.); (J.V.); (O.Z.)
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
- Correspondence: ; Tel.: +34-94-600-6000 (ext. 7079)
| |
Collapse
|
27
|
Yang L, Wang M, Cheng A, Yang Q, Wu Y, Jia R, Liu M, Zhu D, Chen S, Zhang S, Zhao X, Huang J, Wang Y, Xu Z, Chen Z, Zhu L, Luo Q, Liu Y, Yu Y, Zhang L, Tian B, Pan L, Rehman MU, Chen X. Innate Immune Evasion of Alphaherpesvirus Tegument Proteins. Front Immunol 2019; 10:2196. [PMID: 31572398 PMCID: PMC6753173 DOI: 10.3389/fimmu.2019.02196] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 08/30/2019] [Indexed: 12/24/2022] Open
Abstract
Alphaherpesviruses are a large family of highly successful human and animal DNA viruses that can establish lifelong latent infection in neurons. All alphaherpesviruses have a protein-rich layer called the tegument that, connects the DNA-containing capsid to the envelope. Tegument proteins have a variety of functions, playing roles in viral entry, secondary envelopment, viral capsid nuclear transportation during infection, and immune evasion. Recently, many studies have made substantial breakthroughs in characterizing the innate immune evasion of tegument proteins. A wide range of antiviral tegument protein factors that control incoming infectious pathogens are induced by the type I interferon (IFN) signaling pathway and other innate immune responses. In this review, we discuss the immune evasion of tegument proteins with a focus on herpes simplex virus type I.
Collapse
Affiliation(s)
- Linjiang Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mafeng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dekang Zhu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shaqiu Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xinxin Zhao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Juan Huang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yin Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhiwen Xu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhengli Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ling Zhu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qihui Luo
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yunya Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yanling Yu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ling Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bin Tian
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Leichang Pan
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Mujeeb Ur Rehman
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaoyue Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China.,Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
28
|
Vitallé J, Terrén I, Orrantia A, Zenarruzabeitia O, Borrego F. CD300 receptor family in viral infections. Eur J Immunol 2018; 49:364-374. [DOI: 10.1002/eji.201847951] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/02/2018] [Accepted: 11/26/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Joana Vitallé
- Immunopathology GroupBiocruces Bizkaia Health Research Institute Barakaldo Bizkaia Spain
| | - Iñigo Terrén
- Immunopathology GroupBiocruces Bizkaia Health Research Institute Barakaldo Bizkaia Spain
| | - Ane Orrantia
- Immunopathology GroupBiocruces Bizkaia Health Research Institute Barakaldo Bizkaia Spain
| | - Olatz Zenarruzabeitia
- Immunopathology GroupBiocruces Bizkaia Health Research Institute Barakaldo Bizkaia Spain
| | - Francisco Borrego
- Immunopathology GroupBiocruces Bizkaia Health Research Institute Barakaldo Bizkaia Spain
- IkerbasqueBasque Foundation for Science Bilbao Bizkaia Spain
- Basque Center for Transfusion and Human Tissues Galdakao Spain
| |
Collapse
|
29
|
Tang Z, Cai H, Wang R, Cui Y. Overexpression of CD300A inhibits progression of NSCLC through downregulating Wnt/β-catenin pathway. Onco Targets Ther 2018; 11:8875-8883. [PMID: 30573974 PMCID: PMC6290927 DOI: 10.2147/ott.s185521] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background CD300A, a type I transmembrane glycoprotein receptor, plays an important role in immune response. Recent studies have reported that CD300A is involved in the development of hematological malignancies. Purpose The objective of this study was to investigate the role of CD300A in the progression of non-small-cell lung cancer (NSCLC) and explore the associated mechanism. Materials and methods Gene Expression Profiling Interactive Analysis (GEPIA) was used to analyze the expression of CD300A in NSCLC and its prognostic value. NSCLC cell lines A549 and H1650 were transfected with siRNA-CD300A or pcDNA3.1-CD300A vector to down- or up-regulate the expression of CD300A. Cell Counting Kit 8, colony formation and Transwell assays were used to assess the effects of CD300A on cell proliferation and migration capacities. Flow cytometry was performed to examine rate of apoptosis, and the protein levels of associated proteins was detected using Western blot assay. Results From GEPIA analysis, we observed that expression of CD300A mRNA was downregulated in NSCLC and positively correlated with the overall survival of NSCLC patients. Overexpression of CD300A significantly suppressed cell growth and migration capacities of A549 and H1650 cells and induced cell apoptosis via regulating apoptosis-related proteins. Moreover, decreasing level of CD300A promoted cell growth and migration and blocked apoptosis of NSCLC cells. Furthermore, upregulation of CD300A led to significant decrease in expression level of Wnt3 and β-catenin, the pivotal components in Wnt/β-catenin signaling pathway, and an increase in expression of E-cad, a key protein in tumor metastasis, in A549 and H1650 cells; while depletion of CD300A up-regulated the Wnt/β-catenin signaling pathway. In conclusion, the present study highlighted an anti-oncogenic role of CD300A in the progression of NSCLC via inhibiting Wnt/β-catenin pathway, suggesting that CD300A might be a potential target for the treatment of NSCLC Conclusion CD300A plays an anti-oncogenic role in the progression of NSCLC through inhibiting the Wnt/β-catenin pathway, suggesting that CD300A might be a potential target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Ze Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, People's Republic of China,
| | - Hongfei Cai
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, People's Republic of China,
| | - Rui Wang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, People's Republic of China,
| | - Youbin Cui
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, People's Republic of China,
| |
Collapse
|
30
|
Meng M, Li L, Li R, Wang W, Chen Y, Xie Y, Han R, Zhu K, Huang W, Yang L, Li S, Shi J, Tan W, Gao H, Zhao Y, Yang L, Tan J, Hou Z. A dynamic transcriptomic atlas of cytokine-induced killer cells. J Biol Chem 2018; 293:19600-19612. [PMID: 30333226 PMCID: PMC6314136 DOI: 10.1074/jbc.ra118.003280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 10/11/2018] [Indexed: 12/31/2022] Open
Abstract
Several clinical immunotherapy trials with cytokine-induced killer (CIK) cells have been reported. However, molecular evidence of cell expansion, acquisition of tumor cytotoxicity, and safety of CIK cells is required before putting them to clinical use. Here, we performed dynamic transcriptomic analyses of CIKs generated from primary peripheral blood mononuclear cells exposed to interferon-γ, OKT3, and interleukin-2. CIK mRNAs were extracted and sequenced at days 0, 1, 7, and 14 and subjected to bioinformatics analyses. Using weighted correlation network analysis (WGCNA), we identified two major gene modules that mediate immune cell activation and mitosis. We found that activation and cytotoxicity of CIK cells likely rely on cluster of differentiation 8 (CD8) and its protein partner LCK proto-oncogene, Src family tyrosine kinase (LCK). A time-course series analysis revealed that CIK cells have relatively low immunogenicity because of decreased expression of some self-antigens. Importantly, we identified several crucial activating receptors and auxiliary adhesion receptors expressed on CIK cells that may function as tumor sensors. Interestingly, cytotoxicity-associated genes, including those encoding PRF1, GZMB, FASL, and several cytokines, were up-regulated in mature CIK cells. Most immune-checkpoint molecules and inflammatory tumor-promoting factors were down-regulated in the CIK cells, suggesting efficacy and safety in future clinical trials. Notably, insulin-like growth factor 1 (IGF-1) was highly expressed in CIK cells and may promote cytotoxicity, although it also could facilitate tumorigenesis. The transcriptomic atlas of CIK cells presented here may inform efforts to improve CIK-associated tumor cytotoxicity and safety in clinical trials.
Collapse
Affiliation(s)
- Mingyao Meng
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Lin Li
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Ruhong Li
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Wenju Wang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Yang Chen
- the Ministry of Education (MOE) Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yanhua Xie
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Rui Han
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Kai Zhu
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Wenwen Huang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Lili Yang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Shuo Li
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Jianlin Shi
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Weiwei Tan
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Hui Gao
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Yiyi Zhao
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Li Yang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Jing Tan
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China, .,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Zongliu Hou
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China, .,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| |
Collapse
|
31
|
Du X, Liu B, Ding Q, He D, Zhang R, Yang F, Fan H, Teng L, Xin T. CD300A inhibits tumor cell growth by downregulating AKT phosphorylation in human glioblastoma multiforme. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3471-3478. [PMID: 31949725 PMCID: PMC6962892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 05/17/2018] [Indexed: 06/10/2023]
Abstract
Glioblastoma multiforme (GBM) is a primary malignant tumor of the central nervous system with the highest incidence and dismal prognosis. As a member of the CD300 glycoprotein family, CD300A plays a role in cell proliferation, apoptosis, differentiation, and immune response, but its role in solid tumors remains unknown. In this study, CD300A was observed to be overexpressed in human GBM samples using real-time PCR and western blotting. To investigate the role of CD300A in GBM, CCK8, transwell and flow cytometry analysis were performed to examine the proliferation, migration and apoptosis in GBM cell lines, respectively. From our results, knockdown of CD300A blocks cell proliferation and migration, and induces cell apoptosis in human GBM cells U251MG and U87MG. Further, we assessed AKT expression level in CD300A knockdown and negative control cells. The phosphorylation level of AKT was significantly suppressed in CD300A knockdown cells in comparison to negative control cells, suggesting that CD300A promoted tumor cell growth through the AKT pathway. In conclusion, our findings expand the knowledge of CD300A as an oncogene in solid tumor, and provide experimental and theoretical basis for further clinical application.
Collapse
Affiliation(s)
- Xinrui Du
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, China
- Department of Neurosurgery, Zibo Central HospitalZibo, China
| | - Bin Liu
- Department of Neurosurgery, Kyoto University Graduate School of MedicineKyoto, Japan
| | - Qian Ding
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, China
| | - Dong He
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, China
| | - Rui Zhang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, China
| | - Fan Yang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, China
| | - Haitao Fan
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, China
| | - Liangzhu Teng
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, China
| | - Tao Xin
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, China
- Department of Neurosurgery, Jiangxi Provincial People’s HospitalNanchang, China
| |
Collapse
|
32
|
Kantrowitz J, Sinjab A, Xu L, McDowell TL, Sivakumar S, Lang W, Nunomura-Nakamura S, Fukuoka J, Nemer G, Darwiche N, Chami H, Tfayli A, Wistuba II, Scheet P, Fujimoto J, Spira AE, Kadara H. Genome-Wide Gene Expression Changes in the Normal-Appearing Airway during the Evolution of Smoking-Associated Lung Adenocarcinoma. Cancer Prev Res (Phila) 2018; 11:237-248. [PMID: 29382653 DOI: 10.1158/1940-6207.capr-17-0295] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/07/2017] [Accepted: 01/17/2018] [Indexed: 12/14/2022]
Abstract
Smoking perpetuates in cytologically normal airways a molecular "field of injury" that is pertinent to lung cancer and early detection. The evolution of airway field changes prior to lung oncogenesis is poorly understood largely due to the long latency of lung cancer in smokers. Here, we studied airway expression changes prior to lung cancer onset in mice with knockout of the Gprc5a gene (Gprc5a-/-) and tobacco carcinogen (NNK) exposure and that develop the most common type of lung cancer, lung adenocarcinoma, within 6 months following exposure. Airway epithelial brushings were collected from Gprc5a-/- mice before exposure and at multiple times post-NNK until time of lung adenocarcinoma development and then analyzed by RNA sequencing. Temporal airway profiles were identified by linear models and analyzed by comparative genomics in normal airways of human smokers with and without lung cancer. We identified significantly altered profiles (n = 926) in the NNK-exposed mouse normal airways relative to baseline epithelia, a subset of which were concordantly modulated with smoking status in the human airway. Among airway profiles that were significantly modulated following NNK, we found that expression changes (n = 22) occurring as early as 2 months following exposure were significantly associated with lung cancer status when examined in airways of human smokers. Furthermore, a subset of a recently reported human bronchial gene classifier (Percepta; n = 56) was enriched in the temporal mouse airway profiles. We underscore evolutionarily conserved profiles in the normal-appearing airway that develop prior to lung oncogenesis and that comprise viable markers for early lung cancer detection in suspect smokers. Cancer Prev Res; 11(4); 237-48. ©2018 AACR.
Collapse
Affiliation(s)
- Jacob Kantrowitz
- Section of Computational Biomedicine, School of Medicine, Boston University, Boston, Massachusetts
| | - Ansam Sinjab
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Li Xu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tina L McDowell
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Smruthy Sivakumar
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wenhua Lang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sayuri Nunomura-Nakamura
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Graduate School of Biomedical Science, Nagasaki University, Nagasaki, Japan
| | - Junya Fukuoka
- Graduate School of Biomedical Science, Nagasaki University, Nagasaki, Japan
| | - Georges Nemer
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hassan Chami
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Arafat Tfayli
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paul Scheet
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Avrum E Spira
- Section of Computational Biomedicine, School of Medicine, Boston University, Boston, Massachusetts
| | - Humam Kadara
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
33
|
Rozenberg P, Reichman H, Moshkovits I, Munitz A. CD300 family receptors regulate eosinophil survival, chemotaxis, and effector functions. J Leukoc Biol 2017; 104:21-29. [PMID: 29345367 DOI: 10.1002/jlb.2mr1117-433r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/17/2022] Open
Abstract
The CD300 family of receptors is an evolutionary conserved receptor family that belongs to the Ig superfamily and is expressed predominantly by the myeloid lineage. Over the past couple of years, accumulating data have shown that eosinophils express various Ig superfamily receptors that regulate key checkpoints in their biology including their maturation, transition from the bone marrow to the peripheral blood, migration, adhesion, survival, and effector functions in response to numerous activating signals such as IL-4, IL-33, and bacteria. In this review, we will present the emerging roles of CD300 family receptors and specifically CD300a and CD300f in the regulation of these eosinophil activities. The structure and expression pattern of these molecules will be discussed and their involvement in suppressing or co-activating eosinophil functions in health and disease will be illustrated.
Collapse
Affiliation(s)
- Perri Rozenberg
- Department of Clinical Microbiology and Immunology, the Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Hadar Reichman
- Department of Clinical Microbiology and Immunology, the Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Itay Moshkovits
- Department of Internal Medicine "T" and the Research Center for Digestive Disorders and Liver Diseases, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ariel Munitz
- Department of Clinical Microbiology and Immunology, the Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
34
|
Alaterre E, Raimbault S, Goldschmidt H, Bouhya S, Requirand G, Robert N, Boireau S, Seckinger A, Hose D, Klein B, Moreaux J. CD24, CD27, CD36 and CD302 gene expression for outcome prediction in patients with multiple myeloma. Oncotarget 2017; 8:98931-98944. [PMID: 29228738 PMCID: PMC5716778 DOI: 10.18632/oncotarget.22131] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 08/27/2017] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is a B cell neoplasia characterized by clonal plasma cell (PC) proliferation. Minimal residual disease monitoring by multi-parameter flow cytometry is a powerful tool for predicting treatment efficacy and MM outcome. In this study, we compared CD antigens expression between normal and malignant plasma cells to identify new potential markers to discriminate normal from malignant plasma cells, new potential therapeutic targets for monoclonal-based treatments and new prognostic factors. Nine genes were significantly overexpressed and 16 were significantly downregulated in MMC compared with BMPC (ratio ≥2; FDR CD24, CD27, CD36 and CD302) was associated with a prognostic value in two independent cohorts of patients with MM (HM cohort and TT2 cohort, n=345). The expression level of these four genes was then used to develop a CD gene risk score that classified patients in two groups with different survival (P = 2.06E-6) in the HM training cohort. The prognostic value of the CD gene risk score was validated in two independent cohorts of patients with MM (TT2 cohort and HOVON65/GMMGHD4 cohort, n=282 patients). The CD gene risk score remained a prognostic factor that separated patients in two groups with significantly different overall survival also when using publicly available data from a cohort of relapsing patients treated with bortezomib (n=188). In conclusion, the CD gene risk score allows identifying high risk patients with MM based on CD24, CD27, CD36 and CD302 expression and could represent a powerful tool for simple outcome prediction in MM.
Collapse
Affiliation(s)
- Elina Alaterre
- HORIBA Medical, Parc Euromédecine, Montpellier, France.,Institute of Human Genetics, CNRS-UM UMR9002, Montpellier, France
| | | | - Hartmut Goldschmidt
- Medizinische Klinik und Poliklinik V, Universitätsklinikum Heidelberg, Heidelberg, Germany.,Nationales Centrum für Tumorerkrankungen, Heidelberg, Germany
| | - Salahedine Bouhya
- CHU Montpellier, Department of Clinical Hematology, Montpellier, France
| | - Guilhem Requirand
- Department of Biological Haematology, CHU Montpellier, Montpellier, France.,Institute of Human Genetics, CNRS-UM UMR9002, Montpellier, France
| | - Nicolas Robert
- Department of Biological Haematology, CHU Montpellier, Montpellier, France.,Institute of Human Genetics, CNRS-UM UMR9002, Montpellier, France
| | - Stéphanie Boireau
- Department of Biological Haematology, CHU Montpellier, Montpellier, France.,Institute of Human Genetics, CNRS-UM UMR9002, Montpellier, France
| | - Anja Seckinger
- Medizinische Klinik und Poliklinik V, Universitätsklinikum Heidelberg, Heidelberg, Germany.,Nationales Centrum für Tumorerkrankungen, Heidelberg, Germany
| | - Dirk Hose
- Medizinische Klinik und Poliklinik V, Universitätsklinikum Heidelberg, Heidelberg, Germany.,Nationales Centrum für Tumorerkrankungen, Heidelberg, Germany
| | - Bernard Klein
- Department of Biological Haematology, CHU Montpellier, Montpellier, France.,Institute of Human Genetics, CNRS-UM UMR9002, Montpellier, France.,University of Montpellier, UFR Medecine, Montpellier, France
| | - Jérôme Moreaux
- Department of Biological Haematology, CHU Montpellier, Montpellier, France.,Institute of Human Genetics, CNRS-UM UMR9002, Montpellier, France.,University of Montpellier, UFR Medecine, Montpellier, France
| |
Collapse
|
35
|
Hassouneh F, Lopez-Sejas N, Campos C, Sanchez-Correa B, Tarazona R, Solana R, Pera A. Differential Effect of Cytomegalovirus Infection with Age on the Expression of CD57, CD300a, and CD161 on T-Cell Subpopulations. Front Immunol 2017. [PMID: 28626460 PMCID: PMC5454039 DOI: 10.3389/fimmu.2017.00649] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Immunosenescence is a progressive deterioration of the immune system with aging. It affects both innate and adaptive immunity limiting the response to pathogens and to vaccines. As chronic cytomegalovirus (CMV) infection is probably one of the major driving forces of immunosenescence, and its persistent infection results in functional and phenotypic changes to the T-cell repertoire, the aim of this study was to analyze the effect of CMV-seropositivity and aging on the expression of CD300a and CD161 inhibitory receptors, along with the expression of CD57 marker on CD4+, CD8+, CD8+CD56+ (NKT-Like) and CD4−CD8− (DN) T-cell subsets. Our results showed that, regardless of the T-cell subset, CD57−CD161−CD300a+ T-cells expand with age in CMV-seropositive individuals, whereas CD57−CD161+CD300a+ T-cells decrease. Similarly, CD57+CD161−CD300a+ T-cells expand with age in CMV-seropositive individuals in all subsets except in DN cells and CD57−CD161+CD300a− T-cells decrease in all T-cell subsets except in CD4+ T-cells. Besides, in young individuals, CMV latent infection associates with the expansion of CD57+CD161−CD300a+CD4+, CD57−CD161−CD300a+CD4+, CD57+CD161−CD300a+CD8+, CD57−CD161−CD300a+CD8+, CD57+CD161−CD300a+NKT-like, and CD57+CD161−CD300a+DN T-cells. Moreover, in young individuals, CD161 expression on T-cells is not affected by CMV infection. Changes of CD161 expression were only associated with age in the context of CMV latent infection. Besides, CD300a+CD57+CD161+ and CD300a−CD57+CD161+ phenotypes were not found in any of the T-cell subsets studied except in the DN subpopulation, indicating that in the majority of T-cells, CD161 and CD57 do not co-express. Thus, our results show that CMV latent infection impact on the immune system depends on the age of the individual, highlighting the importance of including CMV serology in any study regarding immunosenescence.
Collapse
Affiliation(s)
- Fakhri Hassouneh
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Cordoba, Cordoba, Spain
| | - Nelson Lopez-Sejas
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Cordoba, Cordoba, Spain
| | - Carmen Campos
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Cordoba, Cordoba, Spain
| | | | - Raquel Tarazona
- Immunology Unit, Department of Physiology, University of Extremadura, Cáceres, Spain
| | - Rafael Solana
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Cordoba, Cordoba, Spain.,Immunology Unit, Department of Physiology, University of Extremadura, Cáceres, Spain
| | - Alejandra Pera
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Cordoba, Cordoba, Spain.,Division of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Brighton, United Kingdom
| |
Collapse
|
36
|
Lopez-Sejas N, Campos C, Hassouneh F, Sanchez-Correa B, Tarazona R, Pera A, Solana R. Effect of CMV and Aging on the Differential Expression of CD300a, CD161, T-bet, and Eomes on NK Cell Subsets. Front Immunol 2016; 7:476. [PMID: 27872625 PMCID: PMC5097920 DOI: 10.3389/fimmu.2016.00476] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 10/19/2016] [Indexed: 12/27/2022] Open
Abstract
Natural killer (NK) cells are innate lymphoid cells involved in the defense against virus-infected cells and tumor cells. NK cell phenotype and function is affected with age and cytomegalovirus (CMV) latent infection. Aging affects the frequency and phenotype of NK cells, and CMV infection also contributes to these alterations. Thus, a reduction of CD56bright NK cell subpopulation associated with age and an expansion of memory-like NK cells CD56dimCD57+NKG2C+ probably related to CMV seropositivity have been described. NK cells express T-bet and Eomes transcription factors that are necessary for the development of NK cells. Here, we analyze the effect of age and CMV seropositivity on the expression of CD300a and CD161 inhibitory receptors, and T-bet and Eomes transcription factors in NK cell subsets defined by the expression of CD56 and CD57. CD300a is expressed by the majority of NK cells. CD56bright NK cells express higher levels of CD300a than CD56dim NK cells. An increase in the expression of CD300a was associated with age, whereas a decreased expression of CD161 in CD56dim NK cells was associated with CMV seropositivity. In CD56dim NK cells, an increased percentage of CD57+CD300a+ and a reduction in the percentage of CD161+CD300a+ cells were found to be associated with CMV seropositivity. Regarding T-bet and Eomes transcription factors, CMV seropositivity was associated with a decrease of T-bethi in CD56dimCD57+ NK cells from young individuals, whereas Eomes expression was increased with CMV seropositivity in both CD56bright and CD56dimCD57+/− (from middle age and young individuals, respectively) and was decreased with aging in all NK subsets from the three group of age. In conclusion, CMV infection and age induce significant changes in the expression of CD300a and CD161 in NK cell subsets defined by the expression of CD56 and CD57. T-bet and Eomes are differentially expressed on NK cell subsets, and their expression is affected by CMV latent infection and aging.
Collapse
Affiliation(s)
- Nelson Lopez-Sejas
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Cordoba , Cordoba , Spain
| | - Carmen Campos
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Cordoba , Cordoba , Spain
| | - Fakhri Hassouneh
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Cordoba , Cordoba , Spain
| | | | - Raquel Tarazona
- Immunology Unit, Department of Physiology, University of Extremadura , Cáceres , Spain
| | - Alejandra Pera
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Cordoba , Cordoba , Spain
| | - Rafael Solana
- Maimonides Biomedicine Institute of Cordoba (IMIBIC), Reina Sofia Hospital, University of Cordoba , Cordoba , Spain
| |
Collapse
|
37
|
Assessment of CD37 B-cell antigen and cell of origin significantly improves risk prediction in diffuse large B-cell lymphoma. Blood 2016; 128:3083-3100. [PMID: 27760757 DOI: 10.1182/blood-2016-05-715094] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 10/11/2016] [Indexed: 01/21/2023] Open
Abstract
CD37 (tetraspanin TSPAN26) is a B-cell surface antigen widely expressed on mature B cells. CD37 is involved in immune regulation and tumor suppression but its function has not been fully elucidated. We assessed CD37 expression in de novo diffuse large B-cell lymphoma (DLBCL), and investigated its clinical and biologic significance in 773 patients treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) and 231 patients treated with CHOP. We found that CD37 loss (CD37-) in ∼60% of DLBCL patients showed significantly decreased survival after R-CHOP treatment, independent of the International Prognostic Index (IPI), germinal center B-cell-like (GCB)/activated B-cell-like (ABC) cell of origin, nodal/extranodal primary origin, and the prognostic factors associated with CD37-, including TP53 mutation, NF-κBhigh, Mychigh, phosphorylated STAT3high, survivinhigh, p63-, and BCL6 translocation. CD37 positivity predicted superior survival, abolishing the prognostic impact of high IPI and above biomarkers in GCB-DLBCL but not in ABC-DLBCL. Combining risk scores for CD37- status and ABC cell of origin with the IPI, defined as molecularly adjusted IPI for R-CHOP (M-IPI-R), or IPI plus immunohistochemistry (IHC; IPI+IHC) for CD37, Myc, and Bcl-2, significantly improved risk prediction over IPI alone. Gene expression profiling suggested that decreased CD20 and increased PD-1 levels in CD37- DLBCL, ICOSLG upregulation in CD37+ GCB-DLBCL, and CD37 functions during R-CHOP treatment underlie the pivotal role of CD37 status in clinical outcomes. In conclusion, CD37 is a critical determinant of R-CHOP outcome in DLBCL especially in GCB-DLBCL, representing its importance for optimal rituximab action and sustained immune responses. The combined molecular and clinical prognostic indices, M-IPI-R and IPI+IHC, have remarkable predictive values in R-CHOP-treated DLBCL.
Collapse
|
38
|
|
39
|
Suppression of CD300A inhibits the growth of diffuse large B-cell lymphoma. Oncotarget 2016; 6:31191-202. [PMID: 26435477 PMCID: PMC4741597 DOI: 10.18632/oncotarget.5152] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 08/20/2015] [Indexed: 12/13/2022] Open
Abstract
CD300A is a type I transmembrane receptor protein which has shown inhibitory effects on B-cell receptor-mediated signals. In an analysis of public dataset, we found that CD300A mRNA levels were inversely correlated with the overall survival time of patients with diffuse large B-cell lymphoma (DLBCL). To decipher the role of CD300A in DLBCL, we knocked down the expression levels of CD300A in DLBCL cells and found that decreasing levels of CD300A significantly inhibited cell proliferation of OCI-Ly01, Farage, and SUDHL-4 cells, but not of VAL, OCI-Ly10, or SUDHL-8 cells. Mechanistically, reduced expression of CD300A resulted in a marked attenuation of AKT phosphorylation, a key molecular event in tumorigenesis, in OCI-Ly01, Farage, and SUDHL-4 cells. Pharmacologic inhibition of PI3K displayed a similar inhibitory effect on cell proliferation. Furthermore, using a xenograft animal model, we found that decreasing levels of CD300A in OCI-Ly01 and Farage cells significantly inhibited tumor formation in vivo. Collectively, our results suggested an oncogenic role of CD300A in DLBCL which could serve as a potential biomarker and therapeutic target for this malignant disease.
Collapse
|
40
|
Dimitrova M, Zenarruzabeitia O, Borrego F, Simhadri VR. CD300c is uniquely expressed on CD56 bright Natural Killer Cells and differs from CD300a upon ligand recognition. Sci Rep 2016; 6:23942. [PMID: 27040328 PMCID: PMC4819222 DOI: 10.1038/srep23942] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 03/17/2016] [Indexed: 12/22/2022] Open
Abstract
Paired receptors on NK cells recognize similar ligands with varied strength of binding ability and perform different functions. The CD300 molecules are emerging as novel immune regulators in health and disease due to their interaction with their lipid-nature ligands. Particularly, the paired receptors CD300c and CD300a have been shown to elicit activating and inhibitory capabilities, respectively. In the current study, we seek to investigate the expression and function of CD300c on human NK cells. We demonstrate that IL-2 and IL-15 treatment significantly induce CD300c expression exclusively on CD56(bright) NK cells. CD300c up-regulation requires STAT5 and its expression is inhibited by IL-4. Consistently, IL-2 secreted from activated CD4(+) T cells specifically induces the expression of CD300c on CD56(bright) NK cells. Crosslinking CD300c with a specific antibody enhances the proficiency of CD56(bright) NK cells to degranulate and induce chemokine and cytokine secretion. We also show the differential binding of CD300a and CD300c to their ligands phosphatidylethanolamine (PE) and phosphatidylserine (PS) and their differential ability to affect CD56(bright) NK cell functions. Our results provide an insight into the novel set of paired receptors CD300a and CD300c that are distinctively expressed on CD56(bright) NK cells with varied effector functions.
Collapse
Affiliation(s)
- Milena Dimitrova
- Division of Biotechnology Review and Research-I, Office of Biotechnology Products Review and Research, CDER, Food and Drug Administration, USA
| | - Olatz Zenarruzabeitia
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain.,Immunotherapy Group, Basque Center for Transfusion and Human Tissues, Galdakao, Spain
| | - Francisco Borrego
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain.,Immunotherapy Group, Basque Center for Transfusion and Human Tissues, Galdakao, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Venkateswara R Simhadri
- Division of Biotechnology Review and Research-I, Office of Biotechnology Products Review and Research, CDER, Food and Drug Administration, USA
| |
Collapse
|
41
|
Pietra G, Vitale C, Pende D, Bertaina A, Moretta F, Falco M, Vacca P, Montaldo E, Cantoni C, Mingari MC, Moretta A, Locatelli F, Moretta L. Human natural killer cells: news in the therapy of solid tumors and high-risk leukemias. Cancer Immunol Immunother 2016; 65:465-76. [PMID: 26289090 PMCID: PMC11028670 DOI: 10.1007/s00262-015-1744-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/18/2015] [Indexed: 12/28/2022]
Abstract
It is well established that natural killer (NK) cells play an important role in the immunity against cancer, while the involvement of other recently identified, NK-related innate lymphoid cells is still poorly defined. In the haploidentical hematopoietic stem cell transplantation for the therapy of high-risk leukemias, NK cells have been shown to exert a key role in killing leukemic blasts residual after conditioning. While the clinical results in the cure of leukemias are excellent, the exploitation of NK cells in the therapy of solid tumors is still limited and unsatisfactory. In solid tumors, NK cell function may be inhibited via different mechanisms, occurring primarily at the tumor site. The cellular interactions in the tumor microenvironment involve tumor cells, stromal cells and resident or recruited leukocytes and may favor tumor evasion from the host's defenses. In this context, a number of cytokines, growth factors and enzymes synthesized by tumor cells, stromal cells, suppressive/regulatory myeloid and lymphoid cells may substantially impair the function of different tumor-reactive effector cells, including NK cells. The identification and characterization of such mechanisms may offer clues for the development of new immunotherapeutic strategies to restore effective anti-tumor responses. In order to harness NK cell-based immunotherapies, several approaches have been proposed, including reinforcement of NK cell cytotoxicity by means of specific cytokines, antibodies or drugs. These new tools may improve NK cell function and/or increase tumor susceptibility to NK-mediated killing. Hence, the integration of NK-based immunotherapies with conventional anti-tumor therapies may increase chances of successful cancer treatment.
Collapse
Affiliation(s)
- Gabriella Pietra
- IRCCS AOU San Martino-IST, Genoa, Italy
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Chiara Vitale
- IRCCS AOU San Martino-IST, Genoa, Italy
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | | | | | - Francesca Moretta
- Department of Medicine, University of Verona, Verona, Italy
- Ospedale Sacro Cuore, Negrar, Verona, Italy
| | - Michela Falco
- Istituto Giannina Gaslini, Via G. Gaslini n. 5, 16147, Genoa, Italy
| | - Paola Vacca
- Department of Experimental Medicine, University of Genova, Genoa, Italy
| | - Elisa Montaldo
- Istituto Giannina Gaslini, Via G. Gaslini n. 5, 16147, Genoa, Italy
| | - Claudia Cantoni
- Department of Experimental Medicine, University of Genova, Genoa, Italy
- Istituto Giannina Gaslini, Via G. Gaslini n. 5, 16147, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genova, Genoa, Italy
| | - Maria Cristina Mingari
- IRCCS AOU San Martino-IST, Genoa, Italy
- Department of Experimental Medicine, University of Genova, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genova, Genoa, Italy
| | - Alessandro Moretta
- Department of Experimental Medicine, University of Genova, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genova, Genoa, Italy
| | | | | |
Collapse
|
42
|
van Rees DJ, Szilagyi K, Kuijpers TW, Matlung HL, van den Berg TK. Immunoreceptors on neutrophils. Semin Immunol 2016; 28:94-108. [PMID: 26976825 PMCID: PMC7129252 DOI: 10.1016/j.smim.2016.02.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 02/24/2016] [Accepted: 02/26/2016] [Indexed: 12/12/2022]
Abstract
Neutrophil activities must be tightly controlled to maintain immune homeostasis. Activating and inhibitory receptors balance the outcome of immune cell activation. Immunoreceptors contain Ig-like extracellular domains and signal via ITAMs or ITIMs. Syk or SHP/SHIP mediate downstream signaling after immunoreceptor activation. Targeting immunoreceptors provides opportunities for therapeutic interventions.
Neutrophils play a critical role in the host defense against infection, and they are able to perform a variety of effector mechanisms for this purpose. However, there are also a number of pathological conditions, including autoimmunity and cancer, in which the activities of neutrophils can be harmful to the host. Thus the activities of neutrophils need to be tightly controlled. As in the case of other immune cells, many of the neutrophil effector functions are regulated by a series of immunoreceptors on the plasma membrane. Here, we review what is currently known about the functions of the various individual immunoreceptors and their signaling in neutrophils. While these immunoreceptors allow for the recognition of a diverse range of extracellular ligands, such as cell surface structures (like proteins, glycans and lipids) and extracellular matrix components, they commonly signal via conserved ITAM or ITIM motifs and their associated downstream pathways that depend on the phosphorylation of tyrosine residues in proteins and/or inositol lipids. This allows for a balanced homeostatic regulation of neutrophil effector functions. Given the number of available immunoreceptors and their fundamental importance for neutrophil behavior, it is perhaps not surprising that pathogens have evolved means to evade immune responses through some of these pathways. Inversely, some of these receptors evolved to specifically recognize these pathogens. Finally, some interactions mediated by immunoreceptors in neutrophils have been identified as promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Dieke J van Rees
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katka Szilagyi
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanke L Matlung
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Timo K van den Berg
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Pseudorabies Virus US3 Protein Kinase Protects Infected Cells from NK Cell-Mediated Lysis via Increased Binding of the Inhibitory NK Cell Receptor CD300a. J Virol 2015; 90:1522-33. [PMID: 26581992 DOI: 10.1128/jvi.02902-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Several reports have indicated that natural killer (NK) cells are of particular importance in the innate response against herpesvirus infections. As a consequence, herpesviruses have developed diverse mechanisms for evading NK cells, although few such mechanisms have been identified for the largest herpesvirus subfamily, the alphaherpesviruses. The antiviral activity of NK cells is regulated by a complex array of interactions between activating/inhibitory receptors on the NK cell surface and the corresponding ligands on the surfaces of virus-infected cells. Here we report that the US3 protein kinase of the alphaherpesvirus pseudorabies virus (PRV) displays previously uncharacterized immune evasion properties: it triggers the binding of the inhibitory NK cell receptor CD300a to the surface of the infected cell, thereby providing increased CD300a-mediated protection of infected cells against NK cell-mediated lysis. US3-mediated CD300a binding was found to depend on aminophospholipid ligands of CD300a and on group I p21-activated kinases. These data identify a novel alphaherpesvirus strategy for evading NK cells and demonstrate, for the first time, a role for CD300a in regulating NK cell activity upon contact with virus-infected target cells. IMPORTANCE Herpesviruses have developed fascinating mechanisms to evade elimination by key elements of the host immune system, contributing to their ability to cause lifelong infections with recurrent reactivation events. Natural killer (NK) cells are central in the innate antiviral response. Here we report that the US3 protein kinase of the alphaherpesvirus pseudorabies virus displays a previously uncharacterized capacity for evasion of NK cells. Expression of US3 protects infected cells from NK cell-mediated lysis via increased binding of the inhibitory NK cell receptor CD300a. We show that this US3-mediated increase in CD300a binding depends on aminophospholipids and on cellular p21-activated kinases (PAKs). The identification of this novel NK cell evasion strategy may contribute to the design of improved herpesvirus vaccines and may also have significance for other PAK- and CD300a-modulating viruses and cancer cells.
Collapse
|
44
|
Rysavy NM, Shimoda LMN, Dixon AM, Speck M, Stokes AJ, Turner H, Umemoto EY. Beyond apoptosis: the mechanism and function of phosphatidylserine asymmetry in the membrane of activating mast cells. BIOARCHITECTURE 2015; 4:127-37. [PMID: 25759911 DOI: 10.1080/19490992.2014.995516] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Loss of plasma membrane asymmetry is a hallmark of apoptosis, but lipid bilayer asymmetry and loss of asymmetry can contribute to numerous cellular functions and responses that are independent of programmed cell death. Exofacial exposure of phosphatidylserine occurs in lymphocytes and mast cells after antigenic stimulation and in the absence of apoptosis, suggesting that there is a functional requirement for phosphatidylserine exposure in immunocytes. In this review we examine current ideas as to the nature of this functional role in mast cell activation. Mechanistically, there is controversy as to the candidate proteins responsible for phosphatidylserine translocation from the internal to external leaflet, and here we review the candidacies of mast cell PLSCR1 and TMEM16F. Finally we examine the potential relationship between functionally important mast cell membrane perturbations and phosphatidylserine exposure during activation.
Collapse
Key Words
- ABCA, ABC binding cassette family A
- CRAC, calcium release activated channel
- GPMV, giant plasma membrane vesicle
- ITIM, immunoreceptor tyrosine based inhibitory motif
- PLA2, phospholipase A2
- PLSCR, phospholipid scramblase
- PMA, phorbol 12,13-myristate acetate
- RBL, rat basophilic leukemia
- RFU, relative fluorescence units
- ROI, region of interest
- TMEM, transmembrane protein
- TMEM16F
- WGA, wheat germ agglutinin
- mast cells
- membrane lipids
- phosphatidylserine
Collapse
Affiliation(s)
- Noel M Rysavy
- a Laboratory of Immunology and Signal Transduction ; Department of Biology; Chaminade University ; Honolulu , Hawai'i USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Cantoni C, Grauwet K, Pietra G, Parodi M, Mingari MC, Maria AD, Favoreel H, Vitale M. Role of NK cells in immunotherapy and virotherapy of solid tumors. Immunotherapy 2015; 7:861-82. [PMID: 26314197 DOI: 10.2217/imt.15.53] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Although natural killer (NK) cells are endowed with powerful cytolytic activity against cancer cells, their role in different therapies against solid tumors has not yet been fully elucidated. Their interactions with various elements of the tumor microenvironment as well as their possible effects in contributing to and/or limiting oncolytic virotherapy render this potential immunotherapeutic tool still difficult to exploit at the bedside. Here, we will review the current literature with the aim of providing new hints to manage this powerful cell type in future innovative therapies, such as the use of NK cells in combination with new cytokines, specific mAbs (inducing ADCC), Tyr-Kinase inhibitors, immunomodulatory drugs and/or the design of oncolytic viruses aimed at optimizing the effect of NK cells in virotherapy.
Collapse
Affiliation(s)
- Claudia Cantoni
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,Istituto Giannina Gaslini, Genova, Italy
| | - Korneel Grauwet
- Laboratory of Immunology, Department of Virology, Parasitology & Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | - Gabriella Pietra
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy
| | - Monica Parodi
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy
| | - Andrea De Maria
- Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,IRCCS AOU San Martino-IST Genova, Genova, Italy.,Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Herman Favoreel
- Laboratory of Immunology, Department of Virology, Parasitology & Immunology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | | |
Collapse
|
46
|
Zenarruzabeitia O, Vitallé J, Eguizabal C, Simhadri VR, Borrego F. The Biology and Disease Relevance of CD300a, an Inhibitory Receptor for Phosphatidylserine and Phosphatidylethanolamine. THE JOURNAL OF IMMUNOLOGY 2015; 194:5053-60. [PMID: 25980030 DOI: 10.4049/jimmunol.1500304] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The CD300a inhibitory receptor belongs to the CD300 family of cell surface molecules that regulate a diverse array of immune cell processes. The inhibitory signal of CD300a depends on the phosphorylation of tyrosine residues embedded in ITIMs of the cytoplasmic tail. CD300a is broadly expressed on myeloid and lymphoid cells, and its expression is differentially regulated depending on the cell type. The finding that CD300a recognizes phosphatidylserine and phosphatidylethanolamine, two aminophospholipids exposed on the outer leaflet of dead and activated cells, has shed new light on its role in the modulation of immune functions and in its participation in the host response to several diseases states, such as infectious diseases, cancer, allergy, and chronic inflammatory diseases. This review summarizes the literature on CD300a expression, regulation, signaling pathways, and ligand interaction, as well as its role in fine tuning immune cell functions and its clinical relevance.
Collapse
Affiliation(s)
| | - Joana Vitallé
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo 48903, Spain
| | - Cristina Eguizabal
- Cell Therapy and Stem Cell Group, Basque Center for Transfusion and Human Tissues, Galdakao 48960, Spain
| | - Venkateswara R Simhadri
- Division of Biotechnology Review and Research-I, Office of Biotechnology Products Review and Research, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993; and
| | - Francisco Borrego
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo 48903, Spain; Ikerbasque, Basque Foundation for Science 48903, Bilbao, Spain
| |
Collapse
|
47
|
Goldberger T, Mandelboim O. The use of microRNA by human viruses: lessons from NK cells and HCMV infection. Semin Immunopathol 2014; 36:659-74. [PMID: 25234555 DOI: 10.1007/s00281-014-0447-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 08/28/2014] [Indexed: 12/21/2022]
Abstract
Depending on ethnicity and on social conditions, between 40 and 90 % of the population is infected with human cytomegalovirus (HCMV). In immunocompetent patients, the virus may cause an acute disease and then revert to a state of latency, which enables its coexistence with the human host. However, in cases of immunosuppression or in neonatal infections, HCMV can cause serious long-lasting illnesses. HCMV has developed multiple mechanisms in order to escape its elimination by the immune system, specifically by two killer cell types of the adaptive and the innate immune systems; cytotoxic T lymphocytes (CTL) and natural killer (NK) cells, respectively. Another fascinating aspect of HCMV is that like other highly developed herpesviruses, it expresses its own unique set of microRNAs. Here, we initially describe how the activity of NK cells is regulated under normal conditions and during infection. Then, we discuss what is currently known about HCMV microRNA-mediated interactions, with special emphasis on immune modulation and NK cell evasion. We further illustrate the significant modulation of cellular microRNAs during HCMV infection. Although, the full target spectrum of HCMV microRNAs is far from being completely elucidated, it can already be concluded that HCMV uses its "multitasking" microRNAs to globally affect its own life cycle, as well as important cellular and immune-related pathways.
Collapse
Affiliation(s)
- Tal Goldberger
- The Lautenberg Center of General and Tumor Immunology, The Hebrew University Hadassah Medical School, IMRIC, Jerusalem, 91120, Israel
| | | |
Collapse
|
48
|
Vitale M, Cantoni C, Pietra G, Mingari MC, Moretta L. Effect of tumor cells and tumor microenvironment on NK-cell function. Eur J Immunol 2014; 44:1582-92. [DOI: 10.1002/eji.201344272] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/21/2014] [Accepted: 04/24/2014] [Indexed: 01/05/2023]
Affiliation(s)
- Massimo Vitale
- IRCCS Azienda Ospedaliera Universitaria San Martino-IST; Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
| | - Claudia Cantoni
- Dipartimento di Medicina Sperimentale; Università di Genova; Genova Italy
- Centro di Eccellenza per la Ricerca Biomedica; Genova Italy
- Istituto Giannina Gaslini; Genova Italy
| | - Gabriella Pietra
- IRCCS Azienda Ospedaliera Universitaria San Martino-IST; Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
- Dipartimento di Medicina Sperimentale; Università di Genova; Genova Italy
| | - Maria Cristina Mingari
- IRCCS Azienda Ospedaliera Universitaria San Martino-IST; Istituto Nazionale per la Ricerca sul Cancro; Genova Italy
- Dipartimento di Medicina Sperimentale; Università di Genova; Genova Italy
- Centro di Eccellenza per la Ricerca Biomedica; Genova Italy
| | | |
Collapse
|
49
|
Bar-On Y, Seidel E, Tsukerman P, Mandelboim M, Mandelboim O. Influenza virus uses its neuraminidase protein to evade the recognition of two activating NK cell receptors. J Infect Dis 2014; 210:410-8. [PMID: 24532603 DOI: 10.1093/infdis/jiu094] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Natural Killer (NK) cells play a central role in the defense against viral infections and in the elimination of transformed cells. The recognition of pathogen-infected and tumor cells is controlled by inhibitory and activating receptors. We have previously shown that among the activating (killer) NK cell receptors the natural cytotoxicity receptors, NKp44 and NKp46, interact with the viral hemagglutinin (HA) protein expressed on the cell surface of influenza-virus-infected cells. We further showed that the interaction between NKp44/NKp46 and viral HA is sialic-acid dependent and that the recognition of HA by NKp44 and NKp46 leads to the elimination of the infected cells. Here we demonstrate that the influenza virus developed a counter-attack mechanism and that the virus uses its neuraminidase (NA) protein to prevent the recognition of HA by both the NKp44 and NKp46 receptors, resulting in reduced elimination of the infected cells by NK cells.
Collapse
Affiliation(s)
- Yotam Bar-On
- The Lautenberg Center for General and Tumor Immunology, The BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC), the Hebrew University Hadassah Medical School, Jerusalem
| | - Einat Seidel
- The Lautenberg Center for General and Tumor Immunology, The BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC), the Hebrew University Hadassah Medical School, Jerusalem
| | - Pinchas Tsukerman
- The Lautenberg Center for General and Tumor Immunology, The BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC), the Hebrew University Hadassah Medical School, Jerusalem
| | - Michal Mandelboim
- Central Virology Laboratory, Ministry of Health, Public Health Services, Chaim, Sheba Medical Center, Ramat-Gan, Israel
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, The BioMedical Research Institute Israel-Canada of the Faculty of Medicine (IMRIC), the Hebrew University Hadassah Medical School, Jerusalem
| |
Collapse
|