1
|
Wang M, Wu H, Jiang W, Ren Y, Yuan X, Wang Y, Zhou J, Feng W, Wang Y, Xu T, Zhang D, Fang Y, He C, Li W. Differences in nature killer cell response and interference with mitochondrial DNA induced apoptosis in moxifloxacin environment. Int Immunopharmacol 2024; 132:111970. [PMID: 38608472 DOI: 10.1016/j.intimp.2024.111970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024]
Abstract
OBJECTIVES As antibiotics become more prevalent, accuracy and safety are critical. Moxifloxacin (MXF) have been reported to have immunomodulatory effects on a variety of immune cells and even anti-proliferative and pro-apoptotic effects, but the mechanism of action is not fully clear. METHODS Peripheral blood mononuclear cells (PBMC) from experimental groups of healthy adults (n = 3) were treated with MXF (10ug/ml) in vitro for 24 h. Single-cell sequencing was performed to investigate differences in the response of each immune cell to MXF. Flow cytometry determined differential gene expression in subsets of most damaged NK cells. Pseudo-time analysis identified drivers that influence MXF-stimulated cell differentiation. Detection of mitochondrial DNA and its involvement in the mitochondrial respiratory chain pathway clarifies the origin of MXF-induced stress injury. RESULTS Moxifloxacin-environmental NK cells are markedly reduced: a new subset of NK cells emerges, and immediate-early-response genes in this subset indicate the presence of an early activation response. The inhibitory receptor-dominant subset shows enhanced activation, leading to increased expression of cytokines and chemokines. The near-mature subset showed greater cytotoxicity and the most pronounced cellular damage. CD56bright cells responded by antagonizing the regulation of activation and inhibitory signals, demonstrating a strong cleavage capacity. The severe depletion of mitochondrial genes was focused on apoptosis induced by the mitochondrial respiratory chain complex. CONCLUSION NK cells exhibit heightened sensitivity to the MXF environment. Different NK subsets upregulate the expression of cytokines and chemokines through different activation pathways. Concurrently, MXF induces impairment of the mitochondrial oxidative phosphorylation system, culminating in apoptosis.
Collapse
Affiliation(s)
- Mengqing Wang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Hao Wu
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Weiwei Jiang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yunfei Ren
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xiaowei Yuan
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yanan Wang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Jian Zhou
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Wei Feng
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yusen Wang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Tianpeng Xu
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Danying Zhang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yunhao Fang
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Chao He
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Wenfang Li
- Department of Emergency and Critical Care, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
2
|
Ziehe D, Wolf A, Rahmel T, Nowak H, Haberl H, Bergmann L, Rump K, Dyck B, Palmowski L, Marko B, Witowski A, Willemsen KM, Pfaender S, Eisenacher M, Anft M, Babel N, Bracht T, Sitek B, Bayer M, Zarbock A, von Groote T, Putensen C, Ehrentraut SF, Weisheit C, Adamzik M, Unterberg M, Koos B. Exploring the relationship between HCMV serostatus and outcomes in COVID-19 sepsis. Front Immunol 2024; 15:1386586. [PMID: 38779663 PMCID: PMC11109369 DOI: 10.3389/fimmu.2024.1386586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Background Sepsis, a life-threatening condition caused by the dysregulated host response to infection, is a major global health concern. Understanding the impact of viral or bacterial pathogens in sepsis is crucial for improving patient outcomes. This study aimed to investigate the human cytomegalovirus (HCMV) seropositivity as a risk factor for development of sepsis in patients with COVID-19. Methods A multicenter observational study enrolled 95 intensive care patients with COVID-19-induced sepsis and 80 post-surgery individuals as controls. HCMV serostatus was determined using an ELISA test. Comprehensive clinical data, including demographics, comorbidities, and 30-day mortality, were collected. Statistical analyses evaluated the association between HCMV seropositivity and COVID-19 induced sepsis. Results The prevalence of HCMV seropositivity did not significantly differ between COVID-19-induced sepsis patients (78%) and controls (71%, p = 0.382) in the entire cohort. However, among patients aged ≤60 years, HCMV seropositivity was significantly higher in COVID-19 sepsis patients compared to controls (86% vs 61%, respectively; p = 0.030). Nevertheless, HCMV serostatus did not affect 30-day survival. Discussion These findings confirm the association between HCMV seropositivity and COVID-19 sepsis in non-geriatric patients. However, the lack of an independent effect on 30-day survival can be explained by the cross-reactivity of HCMV specific CD8+ T-cells towards SARS-CoV-2 peptides, which might confer some protection to HCMV seropositive patients. The inclusion of a post-surgery control group strengthens the generalizability of the findings. Further research is needed to elucidate the underlying mechanisms of this association, explore different patient populations, and identify interventions for optimizing patient management. Conclusion This study validates the association between HCMV seropositivity and severe COVID-19-induced sepsis in non-geriatric patients, contributing to the growing body of evidence on viral pathogens in sepsis. Although HCMV serostatus did not independently influence 30-day survival, future investigations should focus on unraveling the intricate interplay between HCMV, immune responses, and COVID-19. These insights will aid in risk stratification and the development of targeted interventions for viral sepsis.
Collapse
Affiliation(s)
- Dominik Ziehe
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Alexander Wolf
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Tim Rahmel
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Hartmuth Nowak
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Helge Haberl
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Lars Bergmann
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Katharina Rump
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Birte Dyck
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Lars Palmowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Britta Marko
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Andrea Witowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Katrin Maria Willemsen
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Stephanie Pfaender
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Martin Eisenacher
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
- Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr University Bochum, Bochum, Germany
| | - Moritz Anft
- Center for Translational Medicine, Medical Clinic I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Nina Babel
- Center for Translational Medicine, Medical Clinic I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Thilo Bracht
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Barbara Sitek
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Malte Bayer
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Alexander Zarbock
- Klinik für Anästhesiologie, Operative Intensivmedizin und Schmerztherapie, Universitätsklinikum Münster, Münster, Germany
| | - Thilo von Groote
- Klinik für Anästhesiologie, Operative Intensivmedizin und Schmerztherapie, Universitätsklinikum Münster, Münster, Germany
| | - Christian Putensen
- Klinik für Anästhesiologie und Operative Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Stefan Felix Ehrentraut
- Klinik für Anästhesiologie und Operative Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Christina Weisheit
- Klinik für Anästhesiologie und Operative Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Michael Adamzik
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Matthias Unterberg
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Björn Koos
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany
| |
Collapse
|
3
|
Mele D, Ottolini S, Lombardi A, Conteianni D, Bandera A, Oliviero B, Mantovani S, Cassaniti I, Baldanti F, Gori A, Mondelli MU, Varchetta S. Long-term dynamics of natural killer cells in response to SARS-CoV-2 vaccination: Persistently enhanced activity postvaccination. J Med Virol 2024; 96:e29585. [PMID: 38566585 DOI: 10.1002/jmv.29585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/08/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024]
Abstract
Natural Killer (NK) cells play a significant role in the early defense against virus infections and cancer. Recent studies have demonstrated the involvement of NK cells in both the induction and effector phases of vaccine-induced immunity in various contexts. However, their role in shaping immune responses following SARS-CoV-2 vaccination remains poorly understood. To address this matter, we conducted a comprehensive analysis of NK cell phenotype and function in SARS-CoV-2 unexposed individuals who received the BNT162b2 vaccine. We employed a longitudinal study design and utilized a panel of 53 15-mer overlapping peptides covering the receptor binding domain (RBD) of the SARS-CoV-2 Spike protein to assess NK cell function at 0 and 20 days following the first vaccine, and 30 and 240 days following booster. Additionally, we evaluated the levels of total IgG anti-Spike antibodies and their potential neutralizing ability. Our findings revealed an increased NK cell activity upon re-exposure to RBD when combined with IL12 and IL18 several months after booster. Concurrently, we observed that the frequencies of NKG2A + NK cells declined over the course of the follow-up period, while NKG2C increased only in CMV positive subjects. The finding that NK cell functions are inducible 9 months after vaccination upon re-exposure to RBD and cytokines, sheds light on the role of NK cells in contributing to SARS-CoV-2 vaccine-induced immune protection and pave the way to further studies in the field.
Collapse
Affiliation(s)
- Dalila Mele
- Division of Clinical Immunology - Infectious Diseases, Department of Research, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Sabrina Ottolini
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Andrea Lombardi
- Department of Pathophysiology and Transplantation, University of Milano, Milano, Italy
- Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Daniela Conteianni
- Division of Clinical Immunology - Infectious Diseases, Department of Research, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alessandra Bandera
- Department of Pathophysiology and Transplantation, University of Milano, Milano, Italy
- Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Barbara Oliviero
- Division of Clinical Immunology - Infectious Diseases, Department of Research, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Stefania Mantovani
- Division of Clinical Immunology - Infectious Diseases, Department of Research, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Irene Cassaniti
- Department of Microbiology and Virology, Molecular Virology Unit, Fondazione IRCCS, Policlinico S. Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Fausto Baldanti
- Department of Microbiology and Virology, Molecular Virology Unit, Fondazione IRCCS, Policlinico S. Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Andrea Gori
- Department of Clinical Sciences, Infectious Diseases and Immunopathology, L. Sacco Hospital, Università di Milano, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milano, Milano, Italy
| | - Mario U Mondelli
- Division of Clinical Immunology - Infectious Diseases, Department of Research, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Stefania Varchetta
- Division of Clinical Immunology - Infectious Diseases, Department of Research, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
4
|
Yang X, Wang X, Zhang X, Ding H, Wang H, Huang T, Zhang G, Duan J, Xia W, Su B, Jin C, Wu H, Zhang T. Durable natural killer cell response after three doses of SARS-CoV-2 inactivated vaccine in HIV-infected individuals. Chin Med J (Engl) 2023; 136:2948-2959. [PMID: 38018259 PMCID: PMC10752448 DOI: 10.1097/cm9.0000000000002947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine can induce a potent cellular and humoral immune response to protect against SARS-CoV-2 infection. However, it was unknown whether SARS-CoV-2 vaccination can induce effective natural killer (NK) cell response in people living with human immunodeficiency virus (PLWH) and healthy individuals. METHODS Forty-seven PLWH and thirty healthy controls (HCs) inoculated with SARS-CoV-2 inactivated vaccine were enrolled from Beijing Youan Hospital in this study. The effect of SARS-CoV-2 vaccine on NK cell frequency, phenotype, and function in PLWH and HCs was evaluated by flow cytometry, and the response of NK cells to SARS-CoV-2 Omicron Spike (SARS-2-OS) protein stimulation was also evaluated. RESULTS SARS-CoV-2 vaccine inoculation elicited activation and degranulation of NK cells in PLWH, which peaked at 2 weeks and then decreased to a minimum at 12 weeks after the third dose of vaccine. However, in vitro stimulation of the corresponding peripheral blood monocular cells from PLWH with SARS-2-OS protein did not upregulate the expression of the aforementioned markers. Additionally, the frequencies of NK cells expressing the activation markers CD25 and CD69 in PLWH were significantly lower than those in HCs at 0, 4 and 12 weeks, but the percentage of CD16 + NK cells in PLWH was significantly higher than that in HCs at 2, 4 and 12 weeks after the third dose of vaccine. Interestingly, the frequency of CD16 + NK cells was significantly negatively correlated with the proportion of CD107a + NK cells in PLWH at each time point after the third dose. Similarly, this phenomenon was also observed in HCs at 0, 2, and 4 weeks after the third dose. Finally, regardless of whether NK cells were stimulated with SARS-2-OS or not, we did not observe any differences in the expression of NK cell degranulation markers between PLWH and HCs. CONCLUSION s:SARS-CoV-2 vaccine elicited activation and degranulation of NK cells, indicating that the inoculation of SARS-CoV-2 vaccine enhances NK cell immune response.
Collapse
Affiliation(s)
- Xiaodong Yang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Xiuwen Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Xin Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Haifeng Ding
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Hu Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Tao Huang
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Guanghui Zhang
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Junyi Duan
- Tian Yuan Studio, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Wei Xia
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Cong Jin
- National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
5
|
Majewska-Szczepanik M, Kowalczyk P, Askenase PW, Szczepanik M. Natural killer cell-mediated contact dermatitis-like reaction induced by treatment with TLR3 ligand poly(I:C). Contact Dermatitis 2023; 89:230-240. [PMID: 37463838 PMCID: PMC10530359 DOI: 10.1111/cod.14380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/20/2023]
Abstract
INTRODUCTION Poly(I:C) is recognised by endosomal Toll-like receptor 3 (TLR3) and activates cytotoxic CD8(+) lymphocytes and natural killer (NK) cells. It has been shown that the viral TLR3 agonist induces robust and long-lasting T-cell-mediated responses. In addition, TLR3 modulates the contact hypersensitivity reaction. OBJECTIVE This study aimed to determine whether poly(I:C) injection can induce NK-mediated hapten reactivity in mice. METHODS Mice were treated with poly(I:C), and their response to dinitrofluorobenzene hapten was measured by assessing ear swelling and serum interferon gamma (IFN-γ) production. Adoptive cell transfer and cell sorting were used to investigate the mechanism of the reaction, and the phenotype of poly(I:C)-activated liver NK cells was determined by flow cytometry analysis. RESULTS The results showed that poly(I:C) administration increased ear swelling, serum IFN-γ levels and the response to hapten in both immunocompetent and T- and B-cell-deficient mice. Only liver poly(I:C)-activated DX5(+) NK cells were able to transfer reactivity to hapten into a naive recipient. Induction of liver NK cells after poly(I:C) administration was TLR3/TRIF- and IFN-γ-dependent, interleukin 12-independent, and not modulated by MyD88. CONCLUSION This study provides new insights into how poly(I:C) stimulates NK-mediated reactivity to hapten and suggests that liver NK cells may modulate the immune response to non-pathogenic factors during viral infection.
Collapse
Affiliation(s)
- Monika Majewska-Szczepanik
- Department of Medical Physiology, Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-126 Krakow, Poland
- Department of Internal Medicine, Section of Allergy and Clinical Immunology, Yale University School of Medicine, New Haven, USA
| | - Paulina Kowalczyk
- Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Philip W Askenase
- Department of Internal Medicine, Section of Allergy and Clinical Immunology, Yale University School of Medicine, New Haven, USA
| | - Marian Szczepanik
- Department of Internal Medicine, Section of Allergy and Clinical Immunology, Yale University School of Medicine, New Haven, USA
- Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-034 Krakow, Poland
| |
Collapse
|
6
|
Al B, Suen TK, Placek K, Netea MG. Innate (learned) memory. J Allergy Clin Immunol 2023; 152:551-566. [PMID: 37385546 DOI: 10.1016/j.jaci.2023.06.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/24/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
With the growing body of evidence, it is now clear that not only adaptive immune cells but also innate immune cells can mount a more rapid and potent nonspecific immune response to subsequent exposures. This process is known as trained immunity or innate (learned) immune memory. This review discusses the different immune and nonimmune cell types of the central and peripheral immune systems that can develop trained immunity. This review highlights the intracellular signaling and metabolic and epigenetic mechanisms underlying the formation of innate immune memory. Finally, this review explores the health implications together with the potential therapeutic interventions harnessing trained immunity.
Collapse
Affiliation(s)
- Burcu Al
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn
| | - Tsz K Suen
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn
| | - Katarzyna Placek
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn
| | - Mihai G Netea
- Department of Molecular Immunology and Cell Biology, Life and Medical Sciences Institute, University of Bonn; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen.
| |
Collapse
|
7
|
Preechanukul A, Kronsteiner B, Saiprom N, Rochaikun K, Moonmueangsan B, Phunpang R, Ottiwet O, Kongphrai Y, Wapee S, Chotivanich K, Morakot C, Janon R, Dunachie SJ, Chantratita N. Identification and function of a novel human memory-like NK cell population expressing CD160 in melioidosis. iScience 2023; 26:107234. [PMID: 37520720 PMCID: PMC10372747 DOI: 10.1016/j.isci.2023.107234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/01/2023] [Accepted: 06/23/2023] [Indexed: 08/01/2023] Open
Abstract
NK cells are endowed with immunological memory to a range of pathogens but the development of NK cell memory in bacterial infections remains elusive. Here, we establish an assay inducing memory-like NK cell response to Burkholderia pseudomallei, the causative agent of the severe bacterial disease called melioidosis, and explore NK cell memory in a melioidosis patient cohort. We show that NK cells require bacteria-primed monocytes to acquire memory-like properties, demonstrated by bacteria-specific responses, features that strongly associate with CD160 expression. Induction of this memory-like NK cell is partly dependent on CD160 and IL-12R. Importantly, CD160 expression identifies memory-like NK cells in a cohort of recovered melioidosis patients with heightened responses maintained at least 3 months post hospital admission and reduced numbers of this cell population independently correlate with recurrent melioidosis. These newly identified memory-like NK cells are a promising target for future vaccine design and for monitoring protection against infection.
Collapse
Affiliation(s)
- Anucha Preechanukul
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- NDM Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Natnaree Saiprom
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kitilak Rochaikun
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Boonthanom Moonmueangsan
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Rungnapa Phunpang
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Orawan Ottiwet
- Department of Medical Technology and Clinical Pathology, Mukdahan Hospital, Mukdahan, Thailand
| | - Yuphin Kongphrai
- Department of Medical Technology and Clinical Pathology, Mukdahan Hospital, Mukdahan, Thailand
| | - Soonthon Wapee
- Department of Medical Technology and Clinical Pathology, Mukdahan Hospital, Mukdahan, Thailand
| | - Kesinee Chotivanich
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Chumpol Morakot
- Department of Medicine, Mukdahan Hospital, Mukdahan, Thailand
| | - Rachan Janon
- Department of Medicine, Mukdahan Hospital, Mukdahan, Thailand
| | - Susanna J. Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- NDM Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Narisara Chantratita
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| |
Collapse
|
8
|
Lee J, Chang WLW, Scott JM, Hong S, Lee T, Deere JD, Park PH, Sparger EE, Dandekar S, Hartigan-O'Connor DJ, Barry PA, Kim S. FcRγ- NK Cell Induction by Specific Cytomegalovirus and Expansion by Subclinical Viral Infections in Rhesus Macaques. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:443-452. [PMID: 37314415 PMCID: PMC10932550 DOI: 10.4049/jimmunol.2200380] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 05/25/2023] [Indexed: 06/15/2023]
Abstract
"Adaptive" NK cells, characterized by FcRγ deficiency and enhanced responsiveness to Ab-bound, virus-infected cells, have been found in certain hCMV-seropositive individuals. Because humans are exposed to numerous microbes and environmental agents, specific relationships between hCMV and FcRγ-deficient NK cells (also known as g-NK cells) have been challenging to define. Here, we show that a subgroup of rhesus CMV (RhCMV)-seropositive macaques possesses FcRγ-deficient NK cells that stably persist and display a phenotype resembling human FcRγ-deficient NK cells. Moreover, these macaque NK cells resembled human FcRγ-deficient NK cells with respect to functional characteristics, including enhanced responsiveness to RhCMV-infected target in an Ab-dependent manner and hyporesponsiveness to tumor and cytokine stimulation. These cells were not detected in specific pathogen-free (SPF) macaques free of RhCMV and six other viruses; however, experimental infection of SPF animals with RhCMV strain UCD59, but not RhCMV strain 68-1 or SIV, led to induction of FcRγ-deficient NK cells. In non-SPF macaques, coinfection by RhCMV with other common viruses was associated with higher frequencies of FcRγ-deficient NK cells. These results support a causal role for specific CMV strain(s) in the induction of FcRγ-deficient NK cells and suggest that coinfection by other viruses further expands this memory-like NK cell pool.
Collapse
Affiliation(s)
- Jaewon Lee
- Graduate Group of Immunology, University of California, Davis, Davis, CA
- Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA
| | - W L William Chang
- Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA
- California National Primate Research Center, Davis, CA
| | - Jeannine M Scott
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI
| | - Suyeon Hong
- Immunobiology Interdepartmental Graduate Program, Iowa State University, Ames, IA
| | - Taehyung Lee
- Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA
| | - Jesse D Deere
- Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA
| | - Peter H Park
- Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA
| | - Ellen E Sparger
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA
| | - Satya Dandekar
- California National Primate Research Center, Davis, CA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA
| | - Dennis J Hartigan-O'Connor
- California National Primate Research Center, Davis, CA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA
| | - Peter A Barry
- Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA
- California National Primate Research Center, Davis, CA
- Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, CA
| | - Sungjin Kim
- Graduate Group of Immunology, University of California, Davis, Davis, CA
- Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA
| |
Collapse
|
9
|
Zafarani A, Razizadeh MH, Pashangzadeh S, Amirzargar MR, Taghavi-Farahabadi M, Mahmoudi M. Natural killer cells in COVID-19: from infection, to vaccination and therapy. Future Virol 2023:10.2217/fvl-2022-0040. [PMID: 36936055 PMCID: PMC10013930 DOI: 10.2217/fvl-2022-0040] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 01/31/2023] [Indexed: 03/15/2023]
Abstract
Natural killer (NK) cells are among the most important innate immunity members, which are the first cells that fight against infected cells. The function of these cells is impaired in patients with COVID-19 and they are not able to prevent the spread of the disease or destroy the infected cells. Few studies have evaluated the effects of COVID-19 vaccines on NK cells, though it has been demonstrated that DNA vaccines and BNT162b2 can affect NK cell response. In the present paper, the effects of SARS-CoV-2 on the NK cells during infection, the effect of vaccination on NK cells, and the NK cell-based therapies were reviewed.
Collapse
Affiliation(s)
- Alireza Zafarani
- 1Department of Hematology & Blood Banking, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Salar Pashangzadeh
- 3Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
- 4Immunology Today, Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mohammad Reza Amirzargar
- 1Department of Hematology & Blood Banking, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Taghavi-Farahabadi
- 5Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahmoudi
- 6Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Author for correspondence: Tel.: +98 936 002 0731;
| |
Collapse
|
10
|
Wagstaffe HR, Anzala O, Kibuuka H, Anywaine Z, Sirima SB, Thiébaut R, Richert L, Levy Y, Lacabaratz C, Bockstal V, Luhn K, Douoguih M, Goodier MR. NK Cell Subset Redistribution and Antibody Dependent Activation after Ebola Vaccination in Africans. Vaccines (Basel) 2022; 10:vaccines10060884. [PMID: 35746491 PMCID: PMC9230153 DOI: 10.3390/vaccines10060884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/21/2022] Open
Abstract
Natural killer cells play an important role in the control of viral infections both by regulating acquired immune responses and as potent innate or antibody-mediated cytotoxic effector cells. NK cells have been implicated in control of Ebola virus infections and our previous studies in European trial participants have demonstrated durable activation, proliferation and antibody-dependent NK cell activation after heterologous two-dose Ebola vaccination with adenovirus type 26.ZEBOV followed by modified vaccinia Ankara-BN-Filo. Regional variation in immunity and environmental exposure to pathogens, in particular human cytomegalovirus, have profound impacts on NK cell functional capacity. We therefore assessed the NK cell phenotype and function in African trial participants with universal exposure to HCMV. We demonstrate a significant redistribution of NK cell subsets after vaccine dose two, involving the enrichment of less differentiated CD56dimCD57− and CD56dimFcεR1γ+ (canonical) cells and the increased proliferation of these subsets. Sera taken after vaccine dose two support robust antibody-dependent NK cell activation in a standard NK cell readout; these responses correlate strongly with the concentration of anti-Ebola glycoprotein specific antibodies. These sera also promote comparable IFN-γ production in autologous NK cells taken at baseline and post-vaccine dose two. However, degranulation responses of post-vaccination NK cells were reduced compared to baseline NK cells and these effects could not be directly attributed to alterations in NK cell phenotype after vaccination. These studies demonstrate consistent changes in NK cell phenotypic composition and robust antibody-dependent NK cell function and reveal novel characteristics of these responses after heterologous two dose Ebola vaccination in African individuals.
Collapse
Affiliation(s)
- Helen R. Wagstaffe
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK;
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Omu Anzala
- KAVI—Institute of Clinical Research University of Nairobi, Nairobi 19676, Kenya;
| | - Hannah Kibuuka
- Makerere University—Walter Reed Project, Kampala 16524, Uganda;
| | - Zacchaeus Anywaine
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe P.O. Box 49, Uganda;
| | - Sodiomon B. Sirima
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Unité de Recherche Clinique de Banfora, 1487 Avenue Kumda Yonré, Ouagadougou 01 BP 2208, Burkina Faso;
| | - Rodolphe Thiébaut
- Bordeaux Population Health Research Center, University Bordeaux, Inserm, UMR 1219, 33000 Bordeaux, France; (R.T.); (L.R.)
- CIC 1401, EUCLID/F-CRIN Clinical Trials Platform, F-33000 Bordeaux, France
- Inria SISTM Team, F-33405 Talence, France
| | - Laura Richert
- Bordeaux Population Health Research Center, University Bordeaux, Inserm, UMR 1219, 33000 Bordeaux, France; (R.T.); (L.R.)
- CIC 1401, EUCLID/F-CRIN Clinical Trials Platform, F-33000 Bordeaux, France
- Inria SISTM Team, F-33405 Talence, France
| | - Yves Levy
- Inserm U955, Vaccine Research Institute, Université Paris-Est Créteil, Hôpital Henri Mondor, 94010 Creteil, France; (Y.L.); (C.L.)
| | - Christine Lacabaratz
- Inserm U955, Vaccine Research Institute, Université Paris-Est Créteil, Hôpital Henri Mondor, 94010 Creteil, France; (Y.L.); (C.L.)
| | - Viki Bockstal
- Janssen Vaccines and Prevention, 2333 CP Leiden, The Netherlands; (V.B.); (K.L.); (M.D.)
| | - Kerstin Luhn
- Janssen Vaccines and Prevention, 2333 CP Leiden, The Netherlands; (V.B.); (K.L.); (M.D.)
| | - Macaya Douoguih
- Janssen Vaccines and Prevention, 2333 CP Leiden, The Netherlands; (V.B.); (K.L.); (M.D.)
| | - Martin R. Goodier
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK;
- Flow Cytometry and Immunology Platform, MRC Unit the Gambia at London School of Hygiene and Tropical Medicine, Banjul P.O. Box 273, The Gambia
- Correspondence:
| |
Collapse
|
11
|
Sugawara S, Reeves RK, Jost S. Learning to Be Elite: Lessons From HIV-1 Controllers and Animal Models on Trained Innate Immunity and Virus Suppression. Front Immunol 2022; 13:858383. [PMID: 35572502 PMCID: PMC9094575 DOI: 10.3389/fimmu.2022.858383] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/18/2022] [Indexed: 12/23/2022] Open
Abstract
Although antiretroviral therapy (ART) has drastically changed the lives of people living with human immunodeficiency virus-1 (HIV-1), long-term treatment has been associated with a vast array of comorbidities. Therefore, a cure for HIV-1 remains the best option to globally eradicate HIV-1/acquired immunodeficiency syndrome (AIDS). However, development of strategies to achieve complete eradication of HIV-1 has been extremely challenging. Thus, the control of HIV-1 replication by the host immune system, namely functional cure, has long been studied as an alternative approach for HIV-1 cure. HIV-1 elite controllers (ECs) are rare individuals who naturally maintain undetectable HIV-1 replication levels in the absence of ART and whose immune repertoire might be a desirable blueprint for a functional cure. While the role(s) played by distinct human leukocyte antigen (HLA) expression and CD8+ T cell responses expressing cognate ligands in controlling HIV-1 has been widely characterized in ECs, the innate immune phenotype has been decidedly understudied. Comparably, in animal models such as HIV-1-infected humanized mice and simian Immunodeficiency Virus (SIV)-infected non-human primates (NHP), viremic control is known to be associated with specific major histocompatibility complex (MHC) alleles and CD8+ T cell activity, but the innate immune response remains incompletely characterized. Notably, recent work demonstrating the existence of trained innate immunity may provide new complementary approaches to achieve an HIV-1 cure. Herein, we review the known characteristics of innate immune responses in ECs and available animal models, identify gaps of knowledge regarding responses by adaptive or trained innate immune cells, and speculate on potential strategies to induce EC-like responses in HIV-1 non-controllers.
Collapse
|
12
|
Sharpe HR, Provine NM, Bowyer GS, Moreira Folegatti P, Belij-Rammerstorfer S, Flaxman A, Makinson R, Hill AV, Ewer KJ, Pollard AJ, Klenerman P, Gilbert S, Lambe T. CMV-associated T cell and NK cell terminal differentiation does not affect immunogenicity of ChAdOx1 vaccination. JCI Insight 2022; 7:e154187. [PMID: 35192547 PMCID: PMC8986084 DOI: 10.1172/jci.insight.154187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/02/2022] [Indexed: 11/17/2022] Open
Abstract
Cytomegalovirus (CMV) is a globally ubiquitous pathogen with a seroprevalence of approximately 50% in the United Kingdom. CMV infection induces expansion of immunosenescent T cell and NK cell populations, with these cells demonstrating lower responsiveness to activation and reduced functionality upon infection and vaccination. In this study, we found that CMV+ participants had normal T cell responses after a single-dose or homologous vaccination with the viral vector chimpanzee adenovirus developed by the University of Oxford (ChAdOx1). CMV seropositivity was associated with reduced induction of IFN-γ-secreting T cells in a ChAd-Modified Vaccinia Ankara (ChAd-MVA) viral vector vaccination trial. Analysis of participants receiving a single dose of ChAdOx1 demonstrated that T cells from CMV+ donors had a more terminally differentiated profile of CD57+PD1+CD4+ T cells and CD8+ T cells expressing less IL-2Rα (CD25) and fewer polyfunctional CD4+ T cells 14 days after vaccination. NK cells from CMV-seropositive individuals also had a reduced activation profile. Overall, our data suggest that although CMV infection enhances immunosenescence of T and NK populations, it does not affect antigen-specific T cell IFN-γ secretion or antibody IgG production after vaccination with the current ChAdOx1 nCoV-19 vaccination regimen, which has important implications given the widespread use of this vaccine, particularly in low- and middle-income countries with high CMV seroprevalence.
Collapse
Affiliation(s)
| | - Nicholas M. Provine
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, United Kingdom
| | | | | | | | | | | | | | | | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Paul Klenerman
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, United Kingdom
| | | | - Teresa Lambe
- Jenner Institute and
- Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Ietto G, Mortara L, Dalla Gasperina D, Iovino D, Azzi L, Baj A, Ageno W, Genoni AP, Acquati F, Gallazzi M, Spina G, Coco G, Pierin F, Noonan D, Vigezzi A, Monti E, Iori V, Masci F, Franchi C, Di Saverio S, Carcano G. Study of immune-mediated mechanisms in patients tested positive for SARS-CoV-2: phenotypic and functional analysis of monocytes, NK and T cells in the blood of subjects affected by COVID 19. JMIR Res Protoc 2021; 11:e29892. [PMID: 34854818 PMCID: PMC8793914 DOI: 10.2196/29892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 01/01/2023] Open
Abstract
Background The novel coronavirus has a high mortality rate (over 1% for patients older than 50 years). This can only be partially ascribed to other comorbidities. A possible explanation is a factor that assures a prompt response to SARS-CoV-2 in younger people, independent from the novelty of the virus itself. A factor is believed to stimulate the immune system and provide immunity against more antigens. The only external stimulation received by healthy people is vaccination (eg, the diphtheria, tetanus, and pertussis [DTP] vaccine). One hypothesis is that vaccination helps develop specific immunity but generates sprouting immunity against antigens in transit. The underlying immunological phenomena are the “bystander effect” and “trained immunity.” The developed immunity gives protection for years until it naturally fades out. After the fifth decade of life, the immune system is almost incompetent when a viral infection occurs, and thus, at this stage, the novel coronavirus can enter the body and cause acute respiratory distress syndrome. Objective The initial aim is to demonstrate that blood monocytes and natural killer cells show overpowering hyperactivity, while CD4+ and CD8+ T cells experience impediments to their defensive functions in patients with severe SARS-CoV-2 infection. The secondary objectives are to correlate clinical data and vaccination history with laboratory immune patterns in order to identify protective factors. Subsequently, we are also interested in characterizing the phenotypes and state of the degree of activation of peripheral blood mononuclear cells, including monocytes, natural killer cells, and CD4+ and CD8+ T cells, in healthy subjects vaccinated with the Pfizer vaccine. Methods Data will be collected using the following 3 approaches: (1) an experimental analysis to study the innate immune response and to identify genetic profiles; (2) an epidemiological analysis to identify the patients’ vaccination history; and (3) a clinical analysis to detect the immunological profile. Results The protocol was approved by the Ethics Committee on April 16, 2020, and the study started on April 27, 2020. As of February 2021, enrollment has been completed. Immunological analysis is ongoing, and we expect to complete this analysis by December 2022. Conclusions We will recognize different populations of patients, each one with a specific immunological pattern in terms of cytokines, soluble factor serum levels, and immune cell activity. Anamnestic data, such as preceding vaccinations and comorbidities, biochemical findings like lymphocyte immunophenotyping, and pre-existing persistent cytomegalovirus infection, allow depicting the risk profile of severe COVID-19. Proof of the roles of these immunological phenomena in the development of COVID-19 can be the basis for the implementation of therapeutic immunomodulatory treatments. Trial Registration ClinicalTrials.gov NCT04375176; https://clinicaltrials.gov/ct2/show/NCT04375176 International Registered Report Identifier (IRRID) DERR1-10.2196/29892
Collapse
Affiliation(s)
- Giuseppe Ietto
- General Emergency and Transplant Surgery Department, University of Insubria, V. Guicciardini, 9, Varese, IT
| | - Lorenzo Mortara
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, IT
| | | | - Domenico Iovino
- General Emergency and Transplant Surgery Department, University of Insubria, V. Guicciardini, 9, Varese, IT
| | - Lorenzo Azzi
- Department of Medicine and Surgery, University of Insubria, Varese, IT
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, Varese, IT
| | - Walter Ageno
- Department of Medicine and Surgery, University of Insubria, Varese, IT
| | - Angelo Paolo Genoni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, IT
| | - Francesco Acquati
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, IT
| | - Matteo Gallazzi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, IT
| | - Giorgia Spina
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, IT
| | - Grace Coco
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, IT
| | - Federica Pierin
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, IT
| | - Douglas Noonan
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, IT.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Multi Medica, Milan, Italy, Milan, IT
| | - Andrea Vigezzi
- General Emergency and Transplant Surgery Department, University of Insubria, V. Guicciardini, 9, Varese, IT
| | - Elisa Monti
- General Emergency and Transplant Surgery Department, University of Insubria, V. Guicciardini, 9, Varese, IT
| | - Valentina Iori
- General Emergency and Transplant Surgery Department, University of Insubria, V. Guicciardini, 9, Varese, IT
| | - Federica Masci
- General Emergency and Transplant Surgery Department, University of Insubria, V. Guicciardini, 9, Varese, IT
| | - Caterina Franchi
- General Emergency and Transplant Surgery Department, University of Insubria, V. Guicciardini, 9, Varese, IT
| | | | - Giulio Carcano
- General Emergency and Transplant Surgery Department, University of Insubria, V. Guicciardini, 9, Varese, IT
| |
Collapse
|
14
|
de Sousa LP, Ribeiro-Gomes FL, de Almeida RF, Souza TME, Werneck GL, Souza DO, Daniel-Ribeiro CT. Immune system challenge improves recognition memory and reverses malaria-induced cognitive impairment in mice. Sci Rep 2021; 11:14857. [PMID: 34290279 PMCID: PMC8295320 DOI: 10.1038/s41598-021-94167-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/05/2021] [Indexed: 11/09/2022] Open
Abstract
The immune system plays a role in the maintenance of healthy neurocognitive function. Different patterns of immune response triggered by distinct stimuli may affect nervous functions through regulatory or deregulatory signals, depending on the properties of the exogenous immunogens. Here, we investigate the effect of immune stimulation on cognitive-behavioural parameters in healthy mice and its impact on cognitive sequelae resulting from non-severe experimental malaria. We show that immune modulation induced by a specific combination of immune stimuli that induce a type 2 immune response can enhance long-term recognition memory in healthy adult mice subjected to novel object recognition task (NORT) and reverse a lack of recognition ability in NORT and anxiety-like behaviour in a light/dark task that result from a single episode of mild Plasmodium berghei ANKA malaria. Our findings suggest a potential use of immunogens for boosting and recovering recognition memory that may be impaired by chronic and infectious diseases and by the effects of ageing.
Collapse
Affiliation(s)
- Luciana Pereira de Sousa
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Fiocruz. Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 2104-360, Brazil
| | - Flávia Lima Ribeiro-Gomes
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Fiocruz. Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 2104-360, Brazil
| | - Roberto Farina de Almeida
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Minas Gerais, Brazil
| | - Tadeu Mello E Souza
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Guilherme Loureiro Werneck
- Departamento de Epidemiologia of Instituto de Medicina Social, Universidade do Estado do Rio de Janeiro and Instituto de Estudos de Saúde Coletiva da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo Onofre Souza
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Fiocruz. Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 2104-360, Brazil.
| |
Collapse
|
15
|
Bajpai G, Nahrendorf M. Infectious and lifestyle modifiers of immunity and host resilience. Immunity 2021; 54:1110-1122. [PMID: 34107270 DOI: 10.1016/j.immuni.2021.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/25/2021] [Accepted: 05/11/2021] [Indexed: 12/30/2022]
Abstract
The interindividual heterogeneity of the immune system likely determines the personal risk for acquiring infections and developing diseases with inflammatory components. In addition to genetic factors, the immune system's heterogeneity is driven by diverging exposures of leukocytes and their progenitors to infections, vaccinations, and health behavior, including lifestyle-related stimuli such as diet, physical inactivity, and psychosocial stress. We review how such experiences alter immune cell responses to concurrent and subsequent challenges, leading to either improved host resilience or disease susceptibility due to a muted or overzealous immune system, with a primary focus on the contribution of innate immune cells. We explore the involvement of diverse mechanisms, including trained immunity, and their relevance for infections and cardiovascular disease, as these prevalent conditions are heavily influenced by immune cell abundance and phenotypic adaptions. Understanding the mechanistic bases of immune modulations by prior or co-exposures may lead to new therapies targeting dysfunctional inflammation.
Collapse
Affiliation(s)
- Geetika Bajpai
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
16
|
Phenotypic and Functional Characteristics of a Novel Influenza Virus Hemagglutinin-Specific Memory NK Cell. J Virol 2021; 95:JVI.00165-21. [PMID: 33827945 DOI: 10.1128/jvi.00165-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/28/2021] [Indexed: 12/30/2022] Open
Abstract
Immune memory represents the most efficient defense against invasion and transmission of infectious pathogens. In contrast to memory T and B cells, the roles of innate immunity in recall responses remain inconclusive. In this study, we identified a novel mouse spleen NK cell subset expressing NKp46 and NKG2A induced by intranasal influenza virus infection. These memory NK cells specifically recognize N-linked glycosylation sites on influenza hemagglutinin (HA) protein. Different from memory-like NK cells reported previously, these NKp46+ NKG2A+ memory NK cells exhibited HA-specific silence of cytotoxicity but increase of gamma interferon (IFN-γ) response against influenza virus-infected cells, which could be reversed by pifithrin-μ, a p53-heat shock protein 70 (HSP70) signaling inhibitor. During recall responses, splenic NKp46+ NKG2A+ NK cells were recruited to infected lung and modulated viral clearance of virus and CD8+ T cell distribution, resulting in improved clinical outcomes. This long-lived NK memory bridges innate and adaptive immune memory response and promotes the homeostasis of local environment during recall response.IMPORTANCE In this study, we demonstrate a novel hemagglutinin (HA)-specific NKp46+ NKG2A+ NK cell subset induced by influenza A virus infection. These memory NK cells show virus-specific decreased cytotoxicity and increased gamma interferon (IFN-γ) on reencountering the same influenza virus antigen. In addition, they modulate host recall responses and CD8 T cell distribution, thus bridging the innate immune and adaptive immune responses during influenza virus infection.
Collapse
|
17
|
Rees-Spear C, McCoy LE. Vaccine responses in ageing and chronic viral infection. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab007. [PMID: 36845567 PMCID: PMC9914503 DOI: 10.1093/oxfimm/iqab007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Over the last few decades, changing population demographics have shown that there are a growing number of individuals living past the age of 60. With this expanding older population comes an increase in individuals that are more susceptible to chronic illness and disease. An important part of maintaining health in this population is through prophylactic vaccination, however, there is growing evidence that vaccines may be less effective in the elderly. Furthermore, with the success of anti-viral therapies, chronic infections such as HIV are becoming increasingly prevalent in older populations and present a relatively unstudied population with respect to the efficacy of vaccination. Here we will examine the evidence for age-associated reduction in antibody and cellular responsiveness to a variety of common vaccines and investigate the underlying causes attributed to this phenomenon, such as inflammation and senescence. We will also discuss the impact of chronic viral infections on immune responses in both young and elderly patients, particularly those living with HIV, and how this affects vaccinations in these populations.
Collapse
Affiliation(s)
- Chloe Rees-Spear
- Division of Infection and Immunity, University College London, London, UK
| | - Laura E McCoy
- Division of Infection and Immunity, University College London, London, UK,Correspondence address. Division of Infection and Immunity, University College London, London, UK. E-mail:
| |
Collapse
|
18
|
Perera Molligoda Arachchige AS. Human NK cells: From development to effector functions. Innate Immun 2021; 27:212-229. [PMID: 33761782 PMCID: PMC8054151 DOI: 10.1177/17534259211001512] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
NK cells are the major lymphocyte subset of the innate immune system that mediates antiviral and anti-tumor responses. It is well established that they develop mechanisms to distinguish self from non-self during the process of NK cell education. Unlike T and B cells, natural killer cells lack clonotypic receptors and are activated after recognizing their target via germline-encoded receptors through natural cytotoxicity, cytokine stimulation, and Ab-dependent cellular cytotoxicity. Subsequently, they utilize cytotoxic granules, death receptor ligands, and cytokines to perform their effector functions. In this review, we provide a general overview of human NK cells, as opposed to murine NK cells, discussing their ontogeny, maturation, receptor diversity, types of responses, and effector functions. Furthermore, we also describe recent advances in human NK cell biology, including tissue-resident NK cell populations, NK cell memory, and novel approaches used to target NK cells in cancer immunotherapy.
Collapse
|
19
|
Sheppard S, Sun JC. Virus-specific NK cell memory. J Exp Med 2021; 218:211913. [PMID: 33755720 PMCID: PMC7992500 DOI: 10.1084/jem.20201731] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/25/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
NK cells express a limited number of germline-encoded receptors that identify infected or transformed cells, eliciting cytotoxicity, effector cytokine production, and in some circumstances clonal proliferation and memory. To maximize the functional diversity of NK cells, the array and expression level of surface receptors vary between individual NK cell “clones” in mice and humans. Cytomegalovirus infection in both species can expand a population of NK cells expressing receptors critical to the clearance of infected cells and generate a long-lived memory pool capable of targeting future infection with greater efficacy. Here, we discuss the pathways and factors that regulate the generation and maintenance of effector and memory NK cells and propose how this understanding may be harnessed therapeutically.
Collapse
Affiliation(s)
- Sam Sheppard
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Joseph C Sun
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY.,Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY
| |
Collapse
|
20
|
Wight A, Parsons BD, Rahim MMA, Makrigiannis AP. A Central Role for Ly49 Receptors in NK Cell Memory. THE JOURNAL OF IMMUNOLOGY 2021; 204:2867-2875. [PMID: 32423924 DOI: 10.4049/jimmunol.2000196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022]
Abstract
In the past decade, the study of NK cells was transformed by the discovery of three ways these "innate" immune cells display adaptive immune behavior, including the ability to form long-lasting, Ag-specific memories of a wide variety of immunogens. In this review, we examine these types of NK cell memory, highlighting their unique features and underlying similarities. We explore those similarities in depth, focusing on the role that Ly49 receptors play in various types of NK cell memory. From this Ly49 dependency, we will build a model by which we understand the three types of NK cell memory as aspects of what is ultimately the same adaptive immune process, rather than separate facets of NK cell biology. We hope that a defined model for NK cell memory will empower collaboration between researchers of these three fields to further our understanding of this surprising and clinically promising immune response.
Collapse
Affiliation(s)
- Andrew Wight
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA 02215
| | - Brendon D Parsons
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; and
| | - Mir Munir A Rahim
- Department of Biomedical Sciences, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| | - Andrew P Makrigiannis
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; and
| |
Collapse
|
21
|
Expansions of adaptive-like NK cells with a tissue-resident phenotype in human lung and blood. Proc Natl Acad Sci U S A 2021; 118:2016580118. [PMID: 33836578 PMCID: PMC7980282 DOI: 10.1073/pnas.2016580118] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Respiratory diseases are leading causes of death worldwide. However, the local immune cell composition in the human lung and individual outliers within the population still remain largely undescribed. We here identify adaptive-like NK cell expansions with tissue-resident traits in lung and blood in approximately 20% of individuals. This particular NK cell subset, which differed from adaptive-like CD16+ blood NK cells, was hyperresponsive to target cell stimulation. Individuals with such in vivo-primed, expanded NK cells will likely experience a different course of acute lung disease such as viral infections. Furthermore, we believe that target cell-hyperresponsive tissue-resident NK cells represent a future tool in the treatment of lung cancer. Human adaptive-like “memory” CD56dimCD16+ natural killer (NK) cells in peripheral blood from cytomegalovirus-seropositive individuals have been extensively investigated in recent years and are currently explored as a treatment strategy for hematological cancers. However, treatment of solid tumors remains limited due to insufficient NK cell tumor infiltration, and it is unknown whether large expansions of adaptive-like NK cells that are equipped for tissue residency and tumor homing exist in peripheral tissues. Here, we show that human lung and blood contains adaptive-like CD56brightCD16− NK cells with hallmarks of tissue residency, including expression of CD49a. Expansions of adaptive-like lung tissue-resident NK (trNK) cells were found to be present independently of adaptive-like CD56dimCD16+ NK cells and to be hyperresponsive toward target cells. Together, our data demonstrate that phenotypically, functionally, and developmentally distinct subsets of adaptive-like NK cells exist in human lung and blood. Given their tissue-related character and hyperresponsiveness, human lung adaptive-like trNK cells might represent a suitable alternative for therapies targeting solid tumors.
Collapse
|
22
|
Wagstaffe HR, Clutterbuck EA, Bockstal V, Stoop JN, Luhn K, Douoguih M, Shukarev G, Snape MD, Pollard AJ, Riley EM, Goodier MR. Ebola virus glycoprotein stimulates IL-18-dependent natural killer cell responses. J Clin Invest 2021; 130:3936-3946. [PMID: 32315287 PMCID: PMC7324188 DOI: 10.1172/jci132438] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 04/16/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND NK cells are activated by innate cytokines and viral ligands to kill virus-infected cells. These functions are enhanced during secondary immune responses and after vaccination by synergy with effector T cells and virus-specific antibodies. In human Ebola virus infection, clinical outcome is strongly associated with the initial innate cytokine response, but the role of NK cells has not been thoroughly examined. METHODS The novel 2-dose heterologous Adenovirus type 26.ZEBOV (Ad26.ZEBOV) and modified vaccinia Ankara-BN-Filo (MVA-BN-Filo) vaccine regimen is safe and provides specific immunity against Ebola glycoprotein, and is currently in phase 2 and 3 studies. Here, we analyzed NK cell phenotype and function in response to Ad26.ZEBOV, MVA-BN-Filo vaccination regimen and in response to in vitro Ebola glycoprotein stimulation of PBMCs isolated before and after vaccination. RESULTS We show enhanced NK cell proliferation and activation after vaccination compared with baseline. Ebola glycoprotein–induced activation of NK cells was dependent on accessory cells and TLR-4–dependent innate cytokine secretion (predominantly from CD14+ monocytes) and enriched within less differentiated NK cell subsets. Optimal NK cell responses were dependent on IL-18 and IL-12, whereas IFN-γ secretion was restricted by high concentrations of IL-10. CONCLUSION This study demonstrates the induction of NK cell effector functions early after Ad26.ZEBOV, MVA-BN-Filo vaccination and provides a mechanism for the activation and regulation of NK cells by Ebola glycoprotein. TRIAL REGISTRATION ClinicalTrials.gov NCT02313077. FUNDING United Kingdom Medical Research Council Studentship in Vaccine Research, Innovative Medicines Initiative 2 Joint Undertaking, EBOVAC (grant 115861) and Crucell Holland (now Janssen Vaccines and Prevention B.V.), European Union’s Horizon 2020 research and innovation programme and European Federation of Pharmaceutical Industries and Associations (EFPIA).
Collapse
Affiliation(s)
- Helen R Wagstaffe
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Elizabeth A Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals and National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Viki Bockstal
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | | | - Kerstin Luhn
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | | | | | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals and National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford University Hospitals and National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Eleanor M Riley
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Martin R Goodier
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
23
|
Durable natural killer cell responses after heterologous two-dose Ebola vaccination. NPJ Vaccines 2021; 6:19. [PMID: 33514756 PMCID: PMC7846750 DOI: 10.1038/s41541-021-00280-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 01/04/2021] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are implicated among immune effectors after vaccination against viral pathogens, including Ebola virus. The two-dose heterologous Ebola virus vaccine regimen, adenovirus type 26.ZEBOV followed by modified vaccinia Ankara-BN-Filo (EBOVAC2 consortium, EU Innovative Medicines Initiative), induces NK cell activation and anti-Ebola glycoprotein (GP) antibody-dependent NK cell activation post-dose 1, which is further elevated post-dose 2. Here, in a multicentre, phase 2 clinical trial (EBL2001), we demonstrate durable ex vivo NK cell activation 180 days after dose 2, with responses enriched in CD56bright NK cells. In vitro antibody-dependent responses to immobilised Ebola GP increased after dose 1, and remained elevated compared to pre-vaccination levels in serum collected 180 days later. Peak NK cell responses were observed post-dose 2 and NK cell IFN-γ responses remained significantly elevated at 180 days post-dose 2. Individual variation in NK cell responses were influenced by both anti-Ebola GP antibody concentrations and intrinsic interindividual differences in NK cell functional capacity. In summary, this study demonstrates durable NK cell responses after Ad26.ZEBOV, MVA-BN-Filo Ebola virus vaccination and could inform the immunological evaluation of future iterations of the vaccine regimen and vaccination schedules.
Collapse
|
24
|
Marotel M, Villard M, Drouillard A, Tout I, Besson L, Allatif O, Pujol M, Rocca Y, Ainouze M, Roblot G, Viel S, Gomez M, Loustaud V, Alain S, Durantel D, Walzer T, Hasan U, Marçais A. Peripheral natural killer cells in chronic hepatitis B patients display multiple molecular features of T cell exhaustion. eLife 2021; 10:60095. [PMID: 33507150 PMCID: PMC7870135 DOI: 10.7554/elife.60095] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/28/2021] [Indexed: 12/11/2022] Open
Abstract
Antiviral effectors such as natural killer (NK) cells have impaired functions in chronic hepatitis B (CHB) patients. The molecular mechanism responsible for this dysfunction remains poorly characterised. We show that decreased cytokine production capacity of peripheral NK cells from CHB patients was associated with reduced expression of NKp30 and CD16, and defective mTOR pathway activity. Transcriptome analysis of patients NK cells revealed an enrichment for transcripts expressed in exhausted T cells suggesting that NK cell dysfunction and T cell exhaustion employ common mechanisms. In particular, the transcription factor TOX and several of its targets were over-expressed in NK cells of CHB patients. This signature was predicted to be dependent on the calcium-associated transcription factor NFAT. Stimulation of the calcium-dependent pathway recapitulated features of NK cells from CHB patients. Thus, deregulated calcium signalling could be a central event in both T cell exhaustion and NK cell dysfunction occurring during chronic infections.
Collapse
Affiliation(s)
- Marie Marotel
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Marine Villard
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France.,Service d'Immunologie biologique, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Annabelle Drouillard
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Issam Tout
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Laurie Besson
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France.,Service d'Immunologie biologique, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Omran Allatif
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Marine Pujol
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Yamila Rocca
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Michelle Ainouze
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Guillaume Roblot
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Sébastien Viel
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France.,Service d'Immunologie biologique, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Melissa Gomez
- CHU Limoges, Service d'Hépatogastroentérologie, U1248 INSERM, Université Limoges, Limoges, France
| | - Veronique Loustaud
- CHU Limoges, Service d'Hépatogastroentérologie, U1248 INSERM, Université Limoges, Limoges, France
| | - Sophie Alain
- Département de Microbiologie, CHU de Limoges, Faculté de médecine-Université de Limoges, Limoges, France
| | - David Durantel
- Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM, U1052, CNRS, Université de Lyon, Lyon, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Uzma Hasan
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie, Team Innate Immunity in Infectious and Autoimmune Diseases, Univ Lyon, Inserm, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| |
Collapse
|
25
|
Goodier MR, Riley EM. Regulation of the human NK cell compartment by pathogens and vaccines. Clin Transl Immunology 2021; 10:e1244. [PMID: 33505682 PMCID: PMC7813579 DOI: 10.1002/cti2.1244] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/17/2022] Open
Abstract
Natural killer cells constitute a phenotypically diverse population of innate lymphoid cells with a broad functional spectrum. Classically defined as cytotoxic lymphocytes with the capacity to eliminate cells lacking self‐MHC or expressing markers of stress or neoplastic transformation, critical roles for NK cells in immunity to infection in the regulation of immune responses and as vaccine‐induced effector cells have also emerged. A crucial feature of NK cell biology is their capacity to integrate signals from pathogen‐, tumor‐ or stress‐induced innate pathways and from antigen‐specific immune responses. The extent to which innate and acquired immune mediators influence NK cell effector function is influenced by the maturation and differentiation state of the NK cell compartment; moreover, NK cell differentiation is driven in part by exposure to infection. Pathogens can thus mould the NK cell response to maximise their own success and/or minimise the damage they cause. Here, we review recent evidence that pathogen‐ and vaccine‐derived signals influence the differentiation, adaptation and subsequent effector function of human NK cells.
Collapse
Affiliation(s)
- Martin R Goodier
- Department of Infection Biology London School of Hygiene and Tropical Medicine London UK
| | - Eleanor M Riley
- Institute of Immunology and Infection Research School of Biological Sciences University of Edinburgh Edinburgh UK
| |
Collapse
|
26
|
Barnes S, Schilizzi O, Audsley KM, Newnes HV, Foley B. Deciphering the Immunological Phenomenon of Adaptive Natural Killer (NK) Cells and Cytomegalovirus (CMV). Int J Mol Sci 2020; 21:ijms21228864. [PMID: 33238550 PMCID: PMC7700325 DOI: 10.3390/ijms21228864] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022] Open
Abstract
Natural killer (NK) cells play a significant and vital role in the first line of defense against infection through their ability to target cells without prior sensitization. They also contribute significantly to the activation and recruitment of both innate and adaptive immune cells through the production of a range of cytokines and chemokines. In the context of cytomegalovirus (CMV) infection, NK cells and CMV have co-evolved side by side to employ several mechanisms to evade one another. However, during this co-evolution the discovery of a subset of long-lived NK cells with enhanced effector potential, increased antibody-dependent responses and the potential to mediate immune memory has revolutionized the field of NK cell biology. The ability of a virus to imprint on the NK cell receptor repertoire resulting in the expansion of diverse, highly functional NK cells to this day remains a significant immunological phenomenon that only occurs in the context of CMV. Here we review our current understanding of the development of these NK cells, commonly referred to as adaptive NK cells and their current role in transplantation, infection, vaccination and cancer immunotherapy to decipher the complex role of CMV in dictating NK cell functional fate.
Collapse
Affiliation(s)
- Samantha Barnes
- Telethon Kids Institute, University of Western Australia, Perth Children’s Hospital, Nedlands, WA 6009, Australia; (S.B.); (O.S.); (K.M.A.); (H.V.N.)
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Ophelia Schilizzi
- Telethon Kids Institute, University of Western Australia, Perth Children’s Hospital, Nedlands, WA 6009, Australia; (S.B.); (O.S.); (K.M.A.); (H.V.N.)
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Katherine M. Audsley
- Telethon Kids Institute, University of Western Australia, Perth Children’s Hospital, Nedlands, WA 6009, Australia; (S.B.); (O.S.); (K.M.A.); (H.V.N.)
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Hannah V. Newnes
- Telethon Kids Institute, University of Western Australia, Perth Children’s Hospital, Nedlands, WA 6009, Australia; (S.B.); (O.S.); (K.M.A.); (H.V.N.)
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Bree Foley
- Telethon Kids Institute, University of Western Australia, Perth Children’s Hospital, Nedlands, WA 6009, Australia; (S.B.); (O.S.); (K.M.A.); (H.V.N.)
- Correspondence:
| |
Collapse
|
27
|
Soleimanian S, Yaghobi R. Harnessing Memory NK Cell to Protect Against COVID-19. Front Pharmacol 2020; 11:1309. [PMID: 32973527 PMCID: PMC7468462 DOI: 10.3389/fphar.2020.01309] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023] Open
Abstract
The worldwide struggle against the coronavirus disease 2019 (COVID-19) as a public health crisis continues to sweep across the globe. Up to now, effective antiviral treatment against COVID-19 is not available. Therefore, throughout virus infections, a thorough clarification of the virus-host immune system interactions will be most probably helpful to encounter these challenges. Emerging evidence suggests that just like SARS and MERS, COVID-19 primarily suppresses the innate immune system, enabling its stable propagation during the early stage of infection. Consequently, proinflammatory cytokines and chemokines have been increasing during infection progression associated with severe lung pathology. It is imperative to consider hyper inflammation in vaccine designing, as vaccine-induced immune responses must have a protective role against infection without leading to immunopathology. Among the front-line responders to viral infections, Natural Killer (NK) cells have immense therapeutic potential, forming a bridge between innate and adaptive responses. A subset of NK cells exhibits putatively increased effector functions against viruses following pathogen-specific and immunization. Memory NK cells have higher cytotoxicity and effector activity, compared with the conventional NK cells. As a pioneering strategy, prompt accumulation and long-term maintenance of these memory NK cells could be an efficacious viral treatment. According to the high prevalence of human cytomegalovirus (HCMV) infection in the world, it remains to be determined whether HCMV adaptive NK cells could play a protective role against this new emerging virus. In addition, the new adaptive-like KIR+NKG2C+ NK cell subset (the adaptive-like lung tissue residue [tr]NK cell) in the context of the respiratory infection at this site could specifically exhibit the expansion upon COVID-19. Another aspect of NK cells we should note, utilizing modified NK cells such as allogeneic off-the-shelf CAR-NK cells as a state-of-the-art strategy for the treatment of COVID-19. In this line, we speculate introducing NKG2C into chimeric antigen receptors in NK cells might be a potential approach in future viral immunotherapy for emerging viruses. In this contribution, we will briefly discuss the current status and future perspective of NK cells, which provide to successfully exploit NK cell-mediated antiviral activity that may offer important new tools in COVID-19 treatment.
Collapse
Affiliation(s)
| | - Ramin Yaghobi
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
28
|
Frank K, Paust S. Dynamic Natural Killer Cell and T Cell Responses to Influenza Infection. Front Cell Infect Microbiol 2020; 10:425. [PMID: 32974217 PMCID: PMC7461885 DOI: 10.3389/fcimb.2020.00425] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
Influenza viruses have perplexed scientists for over a hundred years. Yearly vaccines limit their spread, but they do not prevent all infections. Therapeutic treatments for those experiencing severe infection are limited; further advances are held back by insufficient understanding of the fundamental immune mechanisms responsible for immunopathology. NK cells and T cells are essential in host responses to influenza infection. They produce immunomodulatory cytokines and mediate the cytotoxic response to infection. An imbalance in NK and T cell responses can lead to two outcomes: excessive inflammation and tissue damage or insufficient anti-viral functions and uncontrolled infection. The main cause of death in influenza patients is the former, mediated by hyperinflammatory responses termed “cytokine storm.” NK cells and T cells contribute to cytokine storm, but they are also required for viral clearance. Many studies have attempted to distinguish protective and pathogenic components of the NK cell and T cell influenza response, but it has become clear that they are dynamic and integrated processes. This review will analyze how NK cell and T cell effector functions during influenza infection affect the host response and correlate with morbidity and mortality outcomes.
Collapse
Affiliation(s)
- Kayla Frank
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States.,The Skaggs Graduate Program in Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, United States
| | - Silke Paust
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States.,The Skaggs Graduate Program in Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
29
|
Shih HI, Wu CJ, Tu YF, Chi CY. Fighting COVID-19: A quick review of diagnoses, therapies, and vaccines. Biomed J 2020; 43:341-354. [PMID: 32532623 PMCID: PMC7260535 DOI: 10.1016/j.bj.2020.05.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by a novel coronavirus, SARS-CoV-2, has infected more than 22 million individuals and resulted in over 780,000 deaths globally. The rapid spread of the virus and the precipitously increasing numbers of cases necessitate the urgent development of accurate diagnostic methods, effective treatments, and vaccines. Here, we review the progress of developing diagnostic methods, therapies, and vaccines for SARS-CoV-2 with a focus on current clinical trials and their challenges. For diagnosis, nucleic acid amplification tests remain the mainstay diagnostics for laboratory confirmation of SARS-CoV-2 infection, while serological antibody tests are used to aid contact tracing, epidemiological, and vaccine evaluation studies. Viral isolation is not recommended for routine diagnostic procedures due to safety concerns. Currently, no single effective drug or specific vaccine is available against SARS-CoV-2. Some candidate drugs targeting different levels and stages of human responses against COVID-19 such as cell membrane fusion, RNA-dependent RNA polymerase, viral protease inhibitor, interleukin 6 blocker, and convalescent plasma may improve the clinical outcomes of critical COVID-19 patients. Other supportive care measures for critical patients are still necessary. Advances in genetic sequencing and other technological developments have sped up the establishment of a variety of vaccine platforms. Accordingly, numerous vaccines are under development. Vaccine candidates against SARS-CoV-2 are mainly based upon the viral spike protein due to its vital role in viral infectivity, and most of these candidates have recently moved into clinical trials. Before the efficacy of such vaccines in humans is demonstrated, strong international coordination and collaboration among studies, pharmaceutical companies, regulators, and governments are needed to limit further damage due the emerging SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Hsin-I Shih
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Jung Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Fang Tu
- School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Yu Chi
- School of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan; Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Doctoral Degree Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
30
|
Jaiswal SR, Malhotra P, Mitra DK, Chakrabarti S. Focusing On A Unique Innate Memory Cell Population Of Natural Killer Cells In The Fight Against COVID-19: Harnessing The Ubiquity Of Cytomegalovirus Exposure. Mediterr J Hematol Infect Dis 2020; 12:e2020047. [PMID: 32670525 PMCID: PMC7340217 DOI: 10.4084/mjhid.2020.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/07/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- Sarita Rani Jaiswal
- Cellular Therapy and Immunology, Manashi Chakrabarti Foundation
- Department of Blood and Marrow Transplantation Dharamshila Narayana Super-speciality Hospital, New Delhi, India
| | - Pankaj Malhotra
- Clinical Hematology, Department of Internal Medicine, Post-Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Dipendra K Mitra
- Department of Immunology, All India Institute of Medical Sciences, New Delhi, India
| | - Suparno Chakrabarti
- Cellular Therapy and Immunology, Manashi Chakrabarti Foundation
- Department of Blood and Marrow Transplantation Dharamshila Narayana Super-speciality Hospital, New Delhi, India
| |
Collapse
|
31
|
Responsiveness to Influenza Vaccination Correlates with NKG2C-Expression on NK Cells. Vaccines (Basel) 2020; 8:vaccines8020281. [PMID: 32517137 PMCID: PMC7349951 DOI: 10.3390/vaccines8020281] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/24/2020] [Accepted: 06/03/2020] [Indexed: 12/24/2022] Open
Abstract
Influenza vaccination often results in a large percentage of low responders, especially in high-risk groups. As a first line of defense, natural killer (NK) cells play a crucial role in the fight against infections. However, their implication with regard to vaccine responsiveness is insufficiently assessed. Therefore, this study aimed at the validation of essential NK cell features potentially associated with differential vaccine responsiveness with a special focus on NKG2C- and/or CD57-expressing NK cells considered to harbor memory-like functions. To this end, 16 healthy volunteers were vaccinated with an adjuvanted pandemic influenza vaccine. Vaccine responders and low responders were classified according to their hemagglutination inhibition antibody titers. A majority of responders displayed enhanced frequencies of NKG2C-expressing NK cells 7- or 14-days post-vaccination as compared to low responders, whereas the expression of CD57 was not differentially modulated. The NK cell cytotoxic potential was found to be confined to CD56dimCD16+ NKG2C-expressing NK cells in the responders but not in the low responders, which was further confirmed by stochastic neighbor embedding analysis. The presented study is the first of its kind that ascribes CD56dimCD16+ NKG2C-expressing NK cells a crucial role in biasing adaptive immune responses upon influenza vaccination and suggests NKG2C as a potential biomarker in predicting pandemic influenza vaccine responsiveness.
Collapse
|
32
|
Effects of Porphyra tenera Supplementation on the Immune System: A Randomized, Double-Blind, and Placebo-Controlled Clinical Trial. Nutrients 2020; 12:nu12061642. [PMID: 32498269 PMCID: PMC7352330 DOI: 10.3390/nu12061642] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/21/2020] [Accepted: 05/29/2020] [Indexed: 12/24/2022] Open
Abstract
Objective: The purpose of this study was to determine if Porphyra tenera extract (PTE) has immune-enhancing effects and is safe in healthy adults. Methods: Subjects who met the inclusion criteria (3 × 103 ≤ peripheral blood leukocyte level ≥ 8 × 103 cells/µL) were recruited for this study. Enrolled subjects (n = 120) were randomly assigned to either the PTE group (n = 60) and were given 2.5 g/day of PTE (as PTE) in capsule form or the placebo group (n = 60) and were given crystal cellulose capsules with the identical appearance, weight, and flavor as the PTE capsules for 8 weeks. Outcomes were assessed based on measuring natural killer (NK) cell activity, cytokines level, and upper respiratory infection (URI), and safety parameters were assessed at baseline and 8 weeks. Results: Compared with baseline, NK cell activity (%) increased for all effector cell-to-target cell ratios in the PTE group after 8 weeks; however, changes were not observed in the placebo group (p < 0.10). Subgroup analysis of 101 subjects without URI showed that NK cell activity in the PTE group tended to increase for all effector cell/target cell (E:T) ratios (E:T = 12.5:1 p = 0.068; E:T = 25:1 p = 0.036; E:T = 50:1 p = 0.081) compared with the placebo group. A significant difference between the two groups was observed for the E:T = 25:1 ratio, which increased from 20.3 ± 12.0% at baseline to 23.2 ± 12.4% after 8 weeks in the PTE group (p = 0.036). A significant difference was not observed in cytokine between the two groups. Conclusion: PTE supplementation appears to enhance immune function by improving NK cell activity without adverse effects in healthy adults.
Collapse
|
33
|
Pierce S, Geanes ES, Bradley T. Targeting Natural Killer Cells for Improved Immunity and Control of the Adaptive Immune Response. Front Cell Infect Microbiol 2020; 10:231. [PMID: 32509600 PMCID: PMC7248265 DOI: 10.3389/fcimb.2020.00231] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are critical for targeting and killing tumor, virus-infected and stressed cells as a member of the innate immune system. Recently, NK cells have also emerged as key regulators of adaptive immunity and have become a prominent therapeutic target for cancer immunotherapy and infection control. NK cells display a diverse array of phenotypes and function. Determining how NK cells develop and are regulated is critical for understanding their role in both innate and adaptive immunity. In this review we discuss current research approaches into NK cell adaptive immunity and how these cells are being harnessed for improving cancer and vaccination outcomes.
Collapse
Affiliation(s)
- Stephen Pierce
- Center for Pediatric Genomic Medicine, Children's Mercy Kansas City, Kansas City, MO, United States
| | - Eric S Geanes
- Center for Pediatric Genomic Medicine, Children's Mercy Kansas City, Kansas City, MO, United States
| | - Todd Bradley
- Center for Pediatric Genomic Medicine, Children's Mercy Kansas City, Kansas City, MO, United States.,Departments of Pediatrics and Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States.,Department of Pediatrics, University of Missouri Kansas City Medical School, Kansas City, MO, United States
| |
Collapse
|
34
|
Loiseau C, Doumbo OK, Traore B, Brady JL, Proietti C, de Sousa KP, Crompton PD, Doolan DL. A novel population of memory-activated natural killer cells associated with low parasitaemia in Plasmodium falciparum-exposed sickle-cell trait children. Clin Transl Immunology 2020; 9:e1125. [PMID: 32257211 PMCID: PMC7114700 DOI: 10.1002/cti2.1125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 01/10/2023] Open
Abstract
Objectives The sickle‐cell trait phenotype is associated with protection from malaria. Multiple molecular mechanisms have been proposed to explain this protection, but the role of the host immune system has been poorly investigated. We hypothesised that cellular immunity to malaria may develop differently in sickle‐cell trait children (HbAS) and children with normal haemoglobin (HbAA) repeatedly exposed to Plasmodium falciparum (Pf). Methods Paired samples collected prior to the Pf transmission season and during the first malaria episode of the ensuing transmission season from HbAS and HbAA children were analysed by multiplex bead‐based assay and comprehensive multi‐dimensional flow cytometry profiling. Results Cellular immune profiles were enriched in HbAS relative to HbAA children before the start of the Pf transmission season, with a distinct NK subset. These cells were identified as a novel subset of memory‐activated NK cells characterised by reduced expression of the ecto‐enzyme CD38 as well as co‐expression of high levels of HLA‐DR and CD45RO. The frequency of this NK subset before the transmission season was negatively correlated with parasite density quantified during the first malaria episode of the ensuing transmission season. Functional assessment revealed that these CD38dim CD45RO+ HLA‐DR+ NK cells represent a important source of IFN‐γ. Conclusion Our data suggest that this novel memory‐activated NK cell subset may contribute to an accelerated and enhanced IFN‐γ‐mediated immune response and to control of parasite density in individuals with the sickle‐cell trait. This distinct cellular immune profile may contribute to predispose HbAS children to a relative protection from malaria.
Collapse
Affiliation(s)
- Claire Loiseau
- Centre for Molecular Therapeutics Australian Institute of Tropical Health & Medicine James Cook University Cairns QLD Australia
| | - Ogobara K Doumbo
- Mali International Center of Excellence in Research University of Sciences, Technique and Technology of Bamako Bamako Mali
| | - Boubacar Traore
- Mali International Center of Excellence in Research University of Sciences, Technique and Technology of Bamako Bamako Mali
| | - Jamie L Brady
- Centre for Molecular Therapeutics Australian Institute of Tropical Health & Medicine James Cook University Cairns QLD Australia
| | - Carla Proietti
- Centre for Molecular Therapeutics Australian Institute of Tropical Health & Medicine James Cook University Cairns QLD Australia
| | - Karina P de Sousa
- Centre for Molecular Therapeutics Australian Institute of Tropical Health & Medicine James Cook University Cairns QLD Australia.,Present address: School of Life and Medical Sciences Biosciences Research Group University of Hertfordshire Hatfield UK
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section Laboratory of Immunogenetics National Institute of Allergy and Infectious Diseases National Institutes of Health Rockville MD USA
| | - Denise L Doolan
- Centre for Molecular Therapeutics Australian Institute of Tropical Health & Medicine James Cook University Cairns QLD Australia
| |
Collapse
|
35
|
Brillantes M, Beaulieu AM. Memory and Memory-Like NK Cell Responses to Microbial Pathogens. Front Cell Infect Microbiol 2020; 10:102. [PMID: 32269968 PMCID: PMC7109401 DOI: 10.3389/fcimb.2020.00102] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
NK cells are cytotoxic lymphocytes that provide systemic defense against pathogens and malignancy. Although historically considered cells of the innate immune system, NK cells are now known to be capable of memory or memory-like immune responses in certain settings. Memory NK responses were initially reported over a decade ago in studies involving mouse models of cytomegalovirus infection and delayed-type hypersensitivity reactions to chemical haptens and viral antigens. Since then, a growing body of literature suggests that memory or memory-like NK cell responses may occur in a broader range of immunological settings, including in response to various viral and bacterial infections, and some immunization protocols. Memory-like NK cell responses have also now been reported in humans and non-human primates. Here, we summarize recent studies demonstrating memory or memory-like responses by NK cells in settings of infection and immunization against infectious agents.
Collapse
Affiliation(s)
- Marc Brillantes
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers—The State University of New Jersey, Newark, NJ, United States
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers—The State University of New Jersey, Newark, NJ, United States
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Rutgers—The State University of New Jersey, Newark, NJ, United States
| | - Aimee M. Beaulieu
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers—The State University of New Jersey, Newark, NJ, United States
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers—The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
36
|
Mooney JP, Qendro T, Keith M, Philbey AW, Groves HT, Tregoning JS, Goodier MR, Riley EM. Natural Killer Cells Dampen the Pathogenic Features of Recall Responses to Influenza Infection. Front Immunol 2020; 11:135. [PMID: 32117282 PMCID: PMC7019041 DOI: 10.3389/fimmu.2020.00135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/20/2020] [Indexed: 12/25/2022] Open
Abstract
Despite evidence of augmented Natural Killer (NK) cell responses after influenza vaccination, the role of these cells in vaccine-induced immunity remains unclear. Here, we hypothesized that NK cells might increase viral clearance but possibly at the expense of increased severity of pathology. On the contrary, we found that NK cells serve a homeostatic role during influenza virus infection of vaccinated mice, allowing viral clearance with minimal pathology. Using a diphtheria toxin receptor transgenic mouse model, we were able to specifically deplete NKp46+ NK cells through the administration of diphtheria toxin. Using this model, we assessed the effect of NK cell depletion prior to influenza challenge in vaccinated and unvaccinated mice. NK-depleted, vaccinated animals lost significantly more weight after viral challenge than vaccinated NK intact animals, indicating that NK cells ameliorate disease in vaccinated animals. However, there was also a significant reduction in viral load in NK-depleted, unvaccinated animals indicating that NK cells also constrain viral clearance. Depletion of NK cells after vaccination, but 21 days before infection, did not affect viral clearance or weight loss—indicating that it is the presence of NK cells during the infection itself that promotes homeostasis. Further work is needed to identify the mechanism(s) by which NK cells regulate adaptive immunity in influenza-vaccinated animals to allow efficient and effective virus control whilst simultaneously minimizing inflammation and pathology.
Collapse
Affiliation(s)
- Jason P Mooney
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Tedi Qendro
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Marianne Keith
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Adrian W Philbey
- Easter Bush Pathology, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Helen T Groves
- Department of Medicine, Imperial College London, London, United Kingdom
| | - John S Tregoning
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Martin R Goodier
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Eleanor M Riley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
37
|
Goodier MR, Wolf AS, Riley EM. Differentiation and adaptation of natural killer cells for anti-malarial immunity. Immunol Rev 2019; 293:25-37. [PMID: 31762040 DOI: 10.1111/imr.12798] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/07/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
Natural killer cells employ a diverse arsenal of effector mechanisms to target intracellular pathogens. Differentiation of natural killer (NK) cell activation pathways occurs along a continuum from reliance on innate pro-inflammatory cytokines and stress-induced host ligands through to interaction with signals derived from acquired immune responses. Importantly, the degree of functional differentiation of the NK cell lineage influences the magnitude and specificity of interactions with host cells infected with viruses, bacteria, fungi, and parasites. Individual humans possess a vast diversity of distinct NK cell clones, each with the capacity to vary along this functional differentiation pathway, which - when combined - results in unique individual responses to different infections. Here we summarize these NK cell differentiation events, review evidence for direct interaction of malaria-infected host cells with NK cells and assess how innate inflammatory signals induced by malaria parasite-associated molecular patterns influence the indirect activation and function of NK cells. Finally, we discuss evidence that anti-malarial immunity develops in parallel with advancing NK differentiation, coincident with a loss of reliance on inflammatory signals, and a refined capacity of NK cells to target malaria parasites more precisely, particularly through antibody-dependent mechanisms.
Collapse
Affiliation(s)
- Martin R Goodier
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Asia-Sophia Wolf
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.,Department of Infection and Immunity, University College London, London, UK
| | - Eleanor M Riley
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.,The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| |
Collapse
|
38
|
Gyurova IE, Schlums H, Sucharew H, Ambroggio L, Ochayon DE, Win HT, Bryceson YT, Bernstein DI, Waggoner SN. Dynamic Changes in Natural Killer Cell Subset Frequencies in the Absence of Cytomegalovirus Infection. Front Immunol 2019; 10:2728. [PMID: 31824507 PMCID: PMC6882915 DOI: 10.3389/fimmu.2019.02728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/07/2019] [Indexed: 01/02/2023] Open
Abstract
Individuals lacking functional natural killer (NK) cells suffer severe, recurrent infections with cytomegalovirus (CMV), highlighting the critical role of NK cells in antiviral defense. Therefore, ongoing attempts to develop an efficacious vaccine to prevent CMV infection should potentially aim to elicit NK-cell antiviral responses as an accessory to conventional T- and B-cell based approaches. In this regard, CMV infection provokes marked phenotypic and functional differentiation of the NK-cell compartment, including development of adaptive NK cells that exhibit enhanced antiviral activity. We examined longitudinal blood samples collected from 40 CMV-seronegative adolescents to ascertain whether a CMV glycoprotein B (gB) vaccine in the absence of CMV infection can stimulate differentiation or expansion of CMV-associated subsets of NK cells. Study participants uniformly lacked the CMV-dependent NKG2C+ subset of NK cells, suggesting that an adjuvanted CMV gB vaccine alone is an inadequate stimulus for sustained expansion of these cells. In contrast, we observed unexpected dynamic fluctuations in the frequency of NK cells lacking FcRγ, EAT-2, and SYK, which were independent of vaccination or CMV infection. Whereas, FcRγneg NK cells in CMV infection are reported to express increased levels of the maturation marker CD57, the FcRγneg NK cells observed in our CMV-negative vaccine cohort express less CD57 than their FcRγ+ counterparts. The FcRγneg NK cells in CMV-negative individuals were also functionally distinct from this subset in CMV infection, exhibiting comparable IFN-γ production and degranulation as FcRγ+ NK cells in response to cytokine or antibody-dependent stimuli. These results suggest that frequencies of some NK cell subsets may increase in response to unknown environmental or inflammatory cues distinct from that which occurs after CMV infection. Greater understanding of the nature of the signals driving CMV-independent accumulation of these subsets should permit development of mechanisms to facilitate vaccine-driven expansion of CMV-reactive NK cells.
Collapse
Affiliation(s)
- Ivayla E Gyurova
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, OH, United States
| | - Heinrich Schlums
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Heidi Sucharew
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Lilliam Ambroggio
- Sections of Emergency Medicine and Hospital Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Denver, Denver, CO, United States
| | - David E Ochayon
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Hannah Than Win
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Yenan T Bryceson
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - David I Bernstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Stephen N Waggoner
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
39
|
Wagstaffe HR, Pickering H, Houghton J, Mooney JP, Wolf AS, Prevatt N, Behrens RH, Holland MJ, Riley EM, Goodier MR. Influenza Vaccination Primes Human Myeloid Cell Cytokine Secretion and NK Cell Function. THE JOURNAL OF IMMUNOLOGY 2019; 203:1609-1618. [PMID: 31427444 DOI: 10.4049/jimmunol.1801648] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/16/2019] [Indexed: 12/14/2022]
Abstract
Cytokine-induced memory-like (CIML) NK cells generated in response to proinflammatory cytokines in vitro and in vivo can also be generated by vaccination, exhibiting heightened responses to cytokine stimulation months after their initial induction. Our previous study demonstrated that in vitro human NK cell responses to inactivated influenza virus were also indirectly augmented by very low doses of IL-15, which increased induction of myeloid cell-derived cytokine secretion. These findings led us to hypothesize that IL-15 stimulation could reveal a similar effect for active influenza vaccination and influence CIML NK cell effector functions. In this study, 51 healthy adults were vaccinated with seasonal influenza vaccine, and PBMC were collected before and up to 30 d after vaccination. Myeloid and lymphoid cell cytokine secretion was measured after in vitro PBMC restimulation with low-dose IL-15, alone or in combination with inactivated H3N2 virus; the associated NK cell response was assessed by flow cytometry. PBMC collected 30 d postvaccination showed heightened cytokine production in response to IL-15 compared with PBMC collected at baseline; these responses were further enhanced when IL-15 was combined with H3N2. NK cell activation in response to IL-15 alone (CD25) and H3N2 plus IL-15 (CD25 and IFN-γ) was enhanced postvaccination. We also observed proliferation of less-differentiated NK cells with downregulation of cytokine receptors as early as 3 d after vaccination, suggesting cytokine stimulation in vivo. We conclude that vaccination-induced "training" of accessory cells combines with the generation of CIML NK cells to enhance the overall NK cell response postvaccination.
Collapse
Affiliation(s)
- Helen R Wagstaffe
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Harry Pickering
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Joanna Houghton
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Jason P Mooney
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom.,The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom; and
| | - Asia-Sophia Wolf
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom.,Division of Infection and Immunity, University College London, London WC1E 6JF, United Kingdom
| | - Natalie Prevatt
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Ron H Behrens
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Martin J Holland
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Eleanor M Riley
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom.,The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom; and
| | - Martin R Goodier
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom;
| |
Collapse
|
40
|
Manickam C, Shah SV, Nohara J, Ferrari G, Reeves RK. Monkeying Around: Using Non-human Primate Models to Study NK Cell Biology in HIV Infections. Front Immunol 2019; 10:1124. [PMID: 31191520 PMCID: PMC6540610 DOI: 10.3389/fimmu.2019.01124] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/03/2019] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are the major innate effectors primed to eliminate virus-infected and tumor or neoplastic cells. Recent studies also suggest nuances in phenotypic and functional characteristics among NK cell subsets may further permit execution of regulatory and adaptive roles. Animal models, particularly non-human primate (NHP) models, are critical for characterizing NK cell biology in disease and under homeostatic conditions. In HIV infection, NK cells mediate multiple antiviral functions via upregulation of activating receptors, inflammatory cytokine secretion, and antibody dependent cell cytotoxicity through antibody Fc-FcR interaction and others. However, HIV infection can also reciprocally modulate NK cells directly or indirectly, leading to impaired/ineffective NK cell responses. In this review, we will describe multiple aspects of NK cell biology in HIV/SIV infections and their association with viral control and disease progression, and how NHP models were critical in detailing each finding. Further, we will discuss the effect of NK cell depletion in SIV-infected NHP and the characteristics of newly described memory NK cells in NHP models and different mouse strains. Overall, we propose that the role of NK cells in controlling viral infections remains incompletely understood and that NHP models are indispensable in order to efficiently address these deficits.
Collapse
Affiliation(s)
- Cordelia Manickam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Spandan V. Shah
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Junsuke Nohara
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Guido Ferrari
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - R. Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Ragon Institute of Massachusetts General Hospital, MIT, and Harvard, Cambridge, MA, United States
| |
Collapse
|
41
|
Ghofrani J, Lucar O, Dugan H, Reeves RK, Jost S. Semaphorin 7A modulates cytokine-induced memory-like responses by human natural killer cells. Eur J Immunol 2019; 49:1153-1166. [PMID: 31016720 DOI: 10.1002/eji.201847931] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 03/05/2019] [Accepted: 04/04/2019] [Indexed: 12/24/2022]
Abstract
Cytokine-induced memory-like (CIML) NK cells are endowed with the capacity to mediate enhanced effector functions upon cytokine or activating receptor restimulation for several weeks following short-term preactivation with IL-12, IL-15, and IL-18. Promising results from a first-in-human clinical trial highlighted the clinical potential of CIML NK cells as adoptive immunotherapy for patients with hematologic malignancies. However, the mechanisms underlying CIML NK cell differentiation and increased functionality remain incompletely understood. Semaphorin 7A (SEMA7A) is a potent immunomodulator expressed in activated lymphocytes and myeloid cells. In this study, we show that SEMA7A is substantially upregulated on NK cells stimulated with cytokines, and specifically marks activated NK cells with a strong potential to release IFN-γ. In particular, preactivation of NK cells with IL-12+IL-15+IL-18 resulted in greater than tenfold upregulation of SEMA7A and enhanced expression of the ligand for SEMA7A, integrin-β1, on CIML NK cells. Strikingly, preactivation in the presence of antibodies targeting SEMA7A lead to significantly decreased IFN-γ production following restimulation. These results imply a novel mechanism by which cytokine-enhanced SEMA7A/integrin-β1 interaction promotes CIML NK cell differentiation and maintenance of increased functionality. Our data suggest that targeting SEMA7A/integrin-β1 signaling might provide a novel immunotherapeutic approach to potentiate antitumor activity of CIML NK cells.
Collapse
Affiliation(s)
- Joshua Ghofrani
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Olivier Lucar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Haley Dugan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - R Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Ragon Institute of Massachusetts General Hospital, MIT, and Harvard, Cambridge, MA, USA
| | - Stephanie Jost
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Influenza Virus Infection Enhances Antibody-Mediated NK Cell Functions via Type I Interferon-Dependent Pathways. J Virol 2019; 93:JVI.02090-18. [PMID: 30541850 DOI: 10.1128/jvi.02090-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 11/29/2018] [Indexed: 11/20/2022] Open
Abstract
Natural killer (NK) cells are an important component in the control of influenza virus infection, acting to both clear virus-infected cells and release antiviral cytokines. Engagement of CD16 on NK cells by antibody-coated influenza virus-infected cells results in antibody-dependent cellular cytotoxicity (ADCC). Increasing the potency of antibody-mediated NK cell activity could ultimately lead to improved control of influenza virus infection. To understand if NK cells can be functionally enhanced following exposure to influenza virus-infected cells, we cocultured human peripheral blood mononuclear cells (PBMCs) with influenza virus-infected human alveolar epithelial (A549) cells and evaluated the capacity of NK cells to mediate antibody-dependent functions. Preincubation of PBMCs with influenza virus-infected cells markedly enhanced the ability of NK cells to respond to immune complexes containing hemagglutinin (HA) and anti-HA antibodies or transformed allogeneic cells in the presence or absence of a therapeutic monoclonal antibody. Cytokine multiplex, RNA sequencing, supernatant transfer, Transwell, and cytokine-blocking/cytokine supplementation experiments showed that type I interferons released from PBMCs were primarily responsible for the influenza virus-induced enhancement of antibody-mediated NK cell functions. Importantly, the influenza virus-mediated increase in antibody-dependent NK cell functionality was mimicked by the type I interferon agonist poly(I·C). We conclude that the type I interferon secretion induced by influenza virus infection enhances the capacity of NK cells to mediate ADCC and that this pathway could be manipulated to alter the potency of anti-influenza virus therapies and vaccines.IMPORTANCE Protection from severe influenza may be assisted by antibodies that engage NK cells to kill infected cells through ADCC. Studies have primarily focused on antibodies that have ADCC activity, rather than the capacity of NK cells to become activated and mediate ADCC during an influenza virus infection. We found that type I interferon released in response to influenza virus infection primes NK cells to become highly reactive to anti-influenza virus ADCC antibodies. Enhancing the capacity of NK cells to mediate ADCC could assist in controlling influenza virus infections.
Collapse
|
43
|
Kronstad LM, Seiler C, Vergara R, Holmes SP, Blish CA. Differential Induction of IFN-α and Modulation of CD112 and CD54 Expression Govern the Magnitude of NK Cell IFN-γ Response to Influenza A Viruses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2117-2131. [PMID: 30143589 PMCID: PMC6143432 DOI: 10.4049/jimmunol.1800161] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/19/2018] [Indexed: 01/22/2023]
Abstract
In human and murine studies, IFN-γ is a critical mediator immunity to influenza. IFN-γ production is critical for viral clearance and the development of adaptive immune responses, yet excessive production of IFN-γ and other cytokines as part of a cytokine storm is associated with poor outcomes of influenza infection in humans. As NK cells are the main population of lung innate immune cells capable of producing IFN-γ early in infection, we set out to identify the drivers of the human NK cell IFN-γ response to influenza A viruses. We found that influenza triggers NK cells to secrete IFN-γ in the absence of T cells and in a manner dependent upon signaling from both cytokines and receptor-ligand interactions. Further, we discovered that the pandemic A/California/07/2009 (H1N1) strain elicits a seven-fold greater IFN-γ response than other strains tested, including a seasonal A/Victoria/361/2011 (H3N2) strain. These differential responses were independent of memory NK cells. Instead, we discovered that the A/Victoria/361/2011 influenza strain suppresses the NK cell IFN-γ response by downregulating NK-activating ligands CD112 and CD54 and by repressing the type I IFN response in a viral replication-dependent manner. In contrast, the A/California/07/2009 strain fails to repress the type I IFN response or to downregulate CD54 and CD112 to the same extent, which leads to the enhanced NK cell IFN-γ response. Our results indicate that influenza implements a strain-specific mechanism governing NK cell production of IFN-γ and identifies a previously unrecognized influenza innate immune evasion strategy.
Collapse
Affiliation(s)
- Lisa M Kronstad
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305
| | - Christof Seiler
- Department of Statistics, Stanford University, Stanford, CA 94305
| | - Rosemary Vergara
- Immunology Program, School of Medicine, Stanford University Stanford, CA 94305; and
| | - Susan P Holmes
- Department of Statistics, Stanford University, Stanford, CA 94305
| | - Catherine A Blish
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305;
- Immunology Program, School of Medicine, Stanford University Stanford, CA 94305; and
- Chan Zuckerberg BioHub, San Francisco, CA 94158
| |
Collapse
|
44
|
Ram DR, Manickam C, Hueber B, Itell HL, Permar SR, Varner V, Reeves RK. Tracking KLRC2 (NKG2C)+ memory-like NK cells in SIV+ and rhCMV+ rhesus macaques. PLoS Pathog 2018; 14:e1007104. [PMID: 29851983 PMCID: PMC5997355 DOI: 10.1371/journal.ppat.1007104] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/12/2018] [Accepted: 05/15/2018] [Indexed: 01/25/2023] Open
Abstract
Natural killer (NK) cells classically typify the nonspecific effector arm of the innate immune system, but have recently been shown to possess memory-like properties against multiple viral infections, most notably CMV. Expression of the activating receptor NKG2C is elevated on human NK cells in response to infection with CMV as well as HIV, and may delineate cells with memory and memory-like functions. A better understanding of how NKG2C+ NK cells specifically respond to these pathogens could be significantly advanced using nonhuman primate (NHP) models but, to date, it has not been possible to distinguish NKG2C from its inhibitory counterpart, NKG2A, in NHP because of unfaithful antibody cross-reactivity. Using novel RNA-based flow cytometry, we identify for the first time true memory NKG2C+ NK cells in NHP by gene expression (KLRC2), and show that these cells have elevated frequencies and diversify their functional repertoire specifically in response to rhCMV and SIV infections.
Collapse
Affiliation(s)
- Daniel R. Ram
- Center for Virology and Vaccine Research (CVVR), Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, United States
| | - Cordelia Manickam
- Center for Virology and Vaccine Research (CVVR), Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, United States
| | - Brady Hueber
- Center for Virology and Vaccine Research (CVVR), Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, United States
| | - Hannah L. Itell
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States
| | - Sallie R. Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States
| | - Valerie Varner
- Center for Virology and Vaccine Research (CVVR), Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, United States
| | - R. Keith Reeves
- Center for Virology and Vaccine Research (CVVR), Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, United States
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, Massachusetts, United States
- * E-mail:
| |
Collapse
|
45
|
Kared H, Martelli S, Tan SW, Simoni Y, Chong ML, Yap SH, Newell EW, Pender SLF, Kamarulzaman A, Rajasuriar R, Larbi A. Adaptive NKG2C +CD57 + Natural Killer Cell and Tim-3 Expression During Viral Infections. Front Immunol 2018; 9:686. [PMID: 29731749 PMCID: PMC5919961 DOI: 10.3389/fimmu.2018.00686] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/20/2018] [Indexed: 12/13/2022] Open
Abstract
Repetitive stimulation by persistent pathogens such as human cytomegalovirus (HCMV) or human immunodeficiency virus (HIV) induces the differentiation of natural killer (NK) cells. This maturation pathway is characterized by the acquisition of phenotypic markers, CD2, CD57, and NKG2C, and effector functions—a process regulated by Tim-3 and orchestrated by a complex network of transcriptional factors, involving T-bet, Eomes, Zeb2, promyelocytic leukemia zinc finger protein, and Foxo3. Here, we show that persistent immune activation during chronic viral co-infections (HCMV, hepatitis C virus, and HIV) interferes with the functional phenotype of NK cells by modulating the Tim-3 pathway; a decrease in Tim-3 expression combined with the acquisition of inhibitory receptors skewed NK cells toward an exhausted and cytotoxic phenotype in an inflammatory environment during chronic HIV infection. A better understanding of the mechanisms underlying NK cell differentiation could aid the identification of new immunological targets for checkpoint blockade therapies in a manner that is relevant to chronic infection and cancer.
Collapse
Affiliation(s)
- Hassen Kared
- Singapore Immunology Network (SIgN), Aging and Immunity Program, Agency for Science Technology and Research (ASTAR), Singapore, Singapore
| | - Serena Martelli
- Singapore Immunology Network (SIgN), Aging and Immunity Program, Agency for Science Technology and Research (ASTAR), Singapore, Singapore.,Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Shu Wen Tan
- Singapore Immunology Network (SIgN), Aging and Immunity Program, Agency for Science Technology and Research (ASTAR), Singapore, Singapore
| | - Yannick Simoni
- Singapore Immunology Network (SIgN), Aging and Immunity Program, Agency for Science Technology and Research (ASTAR), Singapore, Singapore
| | - Meng Li Chong
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
| | - Siew Hwei Yap
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
| | - Evan W Newell
- Singapore Immunology Network (SIgN), Aging and Immunity Program, Agency for Science Technology and Research (ASTAR), Singapore, Singapore
| | - Sylvia L F Pender
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Adeeba Kamarulzaman
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia.,Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Reena Rajasuriar
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia.,Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Aging and Immunity Program, Agency for Science Technology and Research (ASTAR), Singapore, Singapore.,Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
46
|
Terrén I, Mikelez I, Odriozola I, Gredilla A, González J, Orrantia A, Vitallé J, Zenarruzabeitia O, Borrego F. Implication of Interleukin-12/15/18 and Ruxolitinib in the Phenotype, Proliferation, and Polyfunctionality of Human Cytokine-Preactivated Natural Killer Cells. Front Immunol 2018; 9:737. [PMID: 29713323 PMCID: PMC5911648 DOI: 10.3389/fimmu.2018.00737] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/26/2018] [Indexed: 12/30/2022] Open
Abstract
A brief in vitro stimulation of natural killer (NK) cells with interleukin (IL)-12, IL-15, and IL-18 endow them a memory-like behavior, characterized by higher effector responses when they are restimulated after a resting period of time. These preactivated NK cells, also known as cytokine-induced memory-like (CIML) NK cells, have several properties that make them a promising tool in cancer immunotherapy. In the present study, we have described the effect that different combinations of IL-12, IL-15, and IL-18 have on the generation of human CIML NK cells. Our data points to a major contribution of IL-15 to CIML NK cell-mediated cytotoxicity against target cells. However, the synergistic effect of the three cytokines grant them the best polyfunctional profile, that is, cells that simultaneously degranulate (CD107a) and produce multiple cytokines and chemokines such as interferon γ, tumor necrosis factor α, and C-C motif chemokine ligand 3. We have also analyzed the involvement of each cytokine and their combinations in the expression of homing receptors CXCR4 and CD62L, as well as the expression of CD25 and IL-2-induced proliferation. Furthermore, we have tested the effects of the Jak1/2 inhibitor ruxolitinib in the generation of CIML NK cells. We found that ruxolitinib-treated CIML NK cells expressed lower levels of CD25 than non-treated CIML NK cells, but exhibited similar proliferation in response to IL-2. In addition, we have also found that ruxolitinib-treated NK cells displayed reduced effector functions after the preactivation, which can be recovered after a 4 days expansion phase in the presence of low doses of IL-2. Altogether, our results describe the impact that each cytokine and the Jak1/2 pathway have in the phenotype, IL-2-induced proliferation, and effector functions of human CIML NK cells.
Collapse
Affiliation(s)
- Iñigo Terrén
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Idoia Mikelez
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
- CIC biomaGUNE, Donostia-San Sebastián, Spain
| | - Irati Odriozola
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Andrea Gredilla
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Javier González
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Ane Orrantia
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | - Joana Vitallé
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
| | | | - Francisco Borrego
- Immunopathology Group, BioCruces Health Research Institute, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Basque Center for Transfusion and Human Tissues, Galdakao, Spain
| |
Collapse
|
47
|
Wagstaffe HR, Nielsen CM, Riley EM, Goodier MR. IL-15 Promotes Polyfunctional NK Cell Responses to Influenza by Boosting IL-12 Production. THE JOURNAL OF IMMUNOLOGY 2018; 200:2738-2747. [PMID: 29491009 PMCID: PMC5890538 DOI: 10.4049/jimmunol.1701614] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/31/2018] [Indexed: 11/19/2022]
Abstract
IL-15 is a key regulator of NK cell maintenance and proliferation and synergizes with other myeloid cell–derived cytokines to enhance NK cell effector function. At low concentrations, trans-presentation of IL-15 by dendritic cells can activate NK cells, whereas at higher concentrations it can act directly on NK cells, independently of accessory cells. In this study, we investigate the potential for IL-15 to boost responses to influenza virus by promoting accessory cell function. We find that coculture of human PBMCs with inactivated whole influenza virus (A/Victoria/361/2011) in the presence of very low concentrations of IL-15 results in increased production of myeloid cell–derived cytokines, including IL-12, IFN-α2, GM-CSF, and IL-1β, and an increased frequency of polyfunctional NK cells (defined by the expression of two or more of CD107a, IFN-γ, and CD25). Neutralization experiments demonstrate that IL-15–mediated enhancement of NK cell responses is primarily dependent on IL-12 and partially dependent on IFN-αβR1 signaling. Critically, IL-15 boosted the production of IL-12 in influenza-stimulated blood myeloid dendritic cells. IL-15 costimulation also restored the ability of less-differentiated NK cells from human CMV-seropositive individuals to respond to influenza virus. These data suggest that very low concentrations of IL-15 play an important role in boosting accessory cell function to support NK cell effector functions.
Collapse
Affiliation(s)
- Helen R Wagstaffe
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Carolyn M Nielsen
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom.,Jenner Institute, University of Oxford, Oxford OX3 7DQ, United Kingdom; and
| | - Eleanor M Riley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom.,The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom
| | - Martin R Goodier
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom;
| |
Collapse
|
48
|
Wagstaffe HR, Mooney JP, Riley EM, Goodier MR. Vaccinating for natural killer cell effector functions. Clin Transl Immunology 2018; 7:e1010. [PMID: 29484187 PMCID: PMC5822400 DOI: 10.1002/cti2.1010] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 12/19/2017] [Accepted: 12/29/2017] [Indexed: 12/21/2022] Open
Abstract
Vaccination has proved to be highly effective in reducing global mortality and eliminating infectious diseases. Building on this success will depend on the development of new and improved vaccines, new methods to determine efficacy and optimum dosing and new or refined adjuvant systems. NK cells are innate lymphoid cells that respond rapidly during primary infection but also have adaptive characteristics enabling them to integrate innate and acquired immune responses. NK cells are activated after vaccination against pathogens including influenza, yellow fever and tuberculosis, and their subsequent maturation, proliferation and effector function is dependent on myeloid accessory cell-derived cytokines such as IL-12, IL-18 and type I interferons. Activation of antigen-presenting cells by live attenuated or whole inactivated vaccines, or by the use of adjuvants, leads to enhanced and sustained NK cell activity, which in turn contributes to T cell recruitment and memory cell formation. This review explores the role of cytokine-activated NK cells as vaccine-induced effector cells and in recall responses and their potential contribution to vaccine and adjuvant development.
Collapse
Affiliation(s)
- Helen R Wagstaffe
- Department of Immunology and InfectionLondon School of Hygiene and Tropical MedicineLondonUK
| | - Jason P Mooney
- Department of Immunology and InfectionLondon School of Hygiene and Tropical MedicineLondonUK
- The Roslin Institute and Royal (Dick) School of Veterinary StudiesUniversity of EdinburghMidlothianUK
| | - Eleanor M Riley
- Department of Immunology and InfectionLondon School of Hygiene and Tropical MedicineLondonUK
- The Roslin Institute and Royal (Dick) School of Veterinary StudiesUniversity of EdinburghMidlothianUK
| | - Martin R Goodier
- Department of Immunology and InfectionLondon School of Hygiene and Tropical MedicineLondonUK
| |
Collapse
|
49
|
Freud AG, Mundy-Bosse BL, Yu J, Caligiuri MA. The Broad Spectrum of Human Natural Killer Cell Diversity. Immunity 2017; 47:820-833. [PMID: 29166586 DOI: 10.1016/j.immuni.2017.10.008] [Citation(s) in RCA: 445] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/07/2017] [Accepted: 10/16/2017] [Indexed: 11/17/2022]
Abstract
Natural killer (NK) cells provide protection against infectious pathogens and cancer. For decades it has been appreciated that two major NK cell subsets (CD56bright and CD56dim) exist in humans and have distinct anatomical localization patterns, phenotypes, and functions in immunity. In light of this traditional NK cell dichotomy, it is now clear that the spectrum of human NK cell diversity is much broader than originally appreciated as a result of variegated surface receptor, intracellular signaling molecule, and transcription factor expression; tissue-specific imprinting; and foreign antigen exposure. The recent discoveries of tissue-resident NK cell developmental intermediates, non-NK innate lymphoid cells, and the capacity for NK cells to adapt and differentiate into long-lived memory cells has added further complexity to this field. Here we review our current understanding of the breadth and generation of human NK cell diversity.
Collapse
Affiliation(s)
- Aharon G Freud
- Department of Pathology, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| | - Bethany L Mundy-Bosse
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Michael A Caligiuri
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
50
|
Goodier MR, Jonjić S, Riley EM, Juranić Lisnić V. CMV and natural killer cells: shaping the response to vaccination. Eur J Immunol 2017; 48:50-65. [PMID: 28960320 DOI: 10.1002/eji.201646762] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/14/2017] [Accepted: 09/22/2017] [Indexed: 12/14/2022]
Abstract
Cytomegaloviruses (CMVs) are highly prevalent, persistent human pathogens that not only evade but also shape our immune responses. Natural killer (NK) cells play an important role in the control of CMV and CMVs have in turn developed a plethora of immunoevasion mechanisms targeting NK cells. This complex interplay can leave a long-lasting imprint on the immune system in general and affect responses toward other pathogens and vaccines. This review aims to provide an overview of NK cell biology and development, the manipulation of NK cells by CMVs and the potential impact of these evasion strategies on responses to vaccination.
Collapse
Affiliation(s)
- Martin R Goodier
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Stipan Jonjić
- Department for Histology and Embryology and Center for Proteomics, Faculty of Medicine, University of Rijeka, Croatia
| | - Eleanor M Riley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Vanda Juranić Lisnić
- Department for Histology and Embryology and Center for Proteomics, Faculty of Medicine, University of Rijeka, Croatia
| |
Collapse
|