1
|
Song Y, Lu J, Qin P, Chen H, Chen L. Interferon-I modulation and natural products: Unraveling mechanisms and therapeutic potential in severe COVID-19. Cytokine Growth Factor Rev 2025; 82:18-30. [PMID: 39261232 DOI: 10.1016/j.cytogfr.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to pose a significant global public health threat, particularly to older adults, pregnant women, and individuals with underlying chronic conditions. Dysregulated immune responses to SARS-CoV-2 infection are believed to contribute to the progression of COVID-19 in severe cases. Previous studies indicates that a deficiency in type I interferon (IFN-I) immunity accounts for approximately 15 %-20 % of patients with severe pneumonia caused by COVID-19, highlighting the potential therapeutic importance of modulating IFN-I signals. Natural products and their derivatives, due to their structural diversity and novel scaffolds, play a crucial role in drug discovery. Some of these natural products targeting IFN-I have demonstrated applications in infectious diseases and inflammatory conditions. However, the immunomodulatory potential of IFN-I in critical COVID-19 pneumonia and the natural compounds regulating the related signal pathway remain not fully understood. In this review, we offer a comprehensive assessment of the association between IFN-I and severe COVID-19, exploring its mechanisms and integrating information on natural compounds effective for IFN-I regulation. Focusing on the primary targets of IFN-I, we also summarize the regulatory mechanisms of natural products, their impact on IFNs, and their therapeutic roles in viral infections. Collectively, by synthesizing these findings, our goal is to provide a valuable reference for future research and to inspire innovative treatment strategies for COVID-19.
Collapse
Affiliation(s)
- Yuheng Song
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiani Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pengcheng Qin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Henan University, Kaifeng 475001, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 200032, China
| | - Lili Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
2
|
Laeremans T, Janssens A, Aerts JL. From natural defenders to therapeutic warriors: NK cells in HIV immunotherapy. Immunotherapy 2025; 17:133-145. [PMID: 39905963 PMCID: PMC11901454 DOI: 10.1080/1750743x.2025.2460965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
Cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells both play essential roles in controlling viral infections by eliminating virus-infected cells. Unlike CTLs, which require priming and activation by antigen-presenting cells, NK cells possess a remarkable capacity to mount a rapid antiviral immune response immediately after infection. Additionally, they can bolster the adaptive immune system by secreting cytokines and directly interacting with other immune cells. However, during chronic human immunodeficiency virus (HIV) infection, various immune cells, including NK cells, experience functional impairments. This has led to the exploration of NK cell-based immunotherapy as a promising strategy to reverse these dysfunctions and contribute to the pursuit of a functional cure for HIV. Building on the success of NK cell therapies in cancer treatment, these approaches offer significant potential for transforming the HIV cure field. This review provides a comprehensive overview of the latest advances in NK cell-based immunotherapy for HIV, outlining the progress made and the key challenges that must be overcome to achieve a functional cure for people living with HIV.
Collapse
Affiliation(s)
- Thessa Laeremans
- Neuro-Aging and Viro-Immunotherapy (NAVI) Research Group, Faculty of Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Amber Janssens
- Neuro-Aging and Viro-Immunotherapy (NAVI) Research Group, Faculty of Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Joeri L. Aerts
- Neuro-Aging and Viro-Immunotherapy (NAVI) Research Group, Faculty of Pharmacy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
3
|
Xu ZR, Xi L, Wu J, Ni JW, Luo FH, Zhang MY. COVID-19 infection and inactivated vaccination: Impacts on clinical and immunological profiles in Chinese children with type 1 diabetes. World J Diabetes 2024; 15:2276-2284. [PMID: 39676798 PMCID: PMC11580597 DOI: 10.4239/wjd.v15.i12.2276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/27/2024] [Accepted: 10/22/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has been linked to an increased incidence of diabetes and diabetic ketoacidosis (DKA). However, the relationship between COVID-19 infection and progression to type 1 diabetes (T1D) in children has not been well defined. AIM To evaluate the influence of COVID-19 infection and inactivated vaccine administration on the progression of T1D among Chinese children. METHODS A total of 197 newly diagnosed patients with T1D were retrospectively enrolled from Children's Hospital of Fudan University between September 2020 and December 2023. The patients were divided into three groups based on their history of COVID-19 infection and vaccination: the infection group, the vaccination-only group, and the non-infection/non-vaccination group. Comprehensive clinical assessments and detailed immunological evaluations were performed to delineate the characteristics and immune responses of these groups. RESULTS The incidence of DKA was significantly higher in the COVID-19 infection group (70.2%) compared to the non-infection/non-vaccination group (62.5%) and vacscination-only group (45.6%; P = 0.015). Prior COVID-19 infection was correlated with increased DKA risk (OR: 1.981, 95%CI: 1.026-3.825, P = 0.042), while vaccination was associated with a reduced risk (OR: 0.558, 95%CI: 0.312-0.998, P = 0.049). COVID-19 infection mildly altered immune profiles, with modest differences in autoantibody positivity, lymphocyte distribution, and immunoglobulin levels. Notably, HLA-DR3 positive children with a history of COVID-19 infection had an earlier T1D onset and lower fasting C-peptide levels than the HLA-DR3 negative children with a history of infection (both P < 0.05). CONCLUSION COVID-19 infection predisposes children to severe T1D, characterized by enhanced DKA risk. Inactivated vaccination significantly lowers DKA incidence at T1D onset. These findings are valuable for guiding future vaccination and T1D risk surveillance strategies in epidemic scenarios in the general pediatric population.
Collapse
Affiliation(s)
- Zhen-Ran Xu
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, National Children’s Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Li Xi
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, National Children’s Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jing Wu
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, National Children’s Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jin-Wen Ni
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, National Children’s Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Fei-Hong Luo
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, National Children’s Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Miao-Ying Zhang
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, National Children’s Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| |
Collapse
|
4
|
Pedroso RB, Torres L, Ventura LA, Camatta GC, Mota C, Mendes AC, Ribeiro F, Guimarães HC, Barbuto RC, Caixeta F, Nascimento LS, Oliveira MA, Martins VD, Silveira-Nunes G, Tupinambás U, Teixeira-Carvalho A, Graça L, Faria AMC. Rapid progression of CD8 and CD4 T cells to cellular exhaustion and senescence during SARS-CoV2 infection. J Leukoc Biol 2024; 116:1385-1397. [PMID: 39298288 DOI: 10.1093/jleuko/qiae180] [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: 11/24/2023] [Revised: 05/19/2024] [Indexed: 09/21/2024] Open
Abstract
Risk factors for the development of severe COVID-19 include several comorbidities, but age was the most striking one since elderly people were disproportionately affected by SARS-CoV-2 infection. Among the reasons for this markedly unfavorable response in the elderly, immunosenescence and inflammaging appear as major drivers of this outcome. A finding that was also notable was that hospitalized patients with severe COVID-19 have an accumulation of senescent T cells, suggesting that immunosenescence may be aggravated by SARS-CoV-2 infection. The present work was designed to examine whether these immunosenescence changes are characteristic of COVID-19 and whether it is dependent on disease severity using cross-sectional and longitudinal studies. Our cross-sectional data show that COVID-19, but not other respiratory infections, rapidly increased cellular senescence and exhaustion in CD4 and CD8 T cells during early infection. In addition, longitudinal analyses with patients from Brazil and Portugal provided evidence of increased frequencies of senescent and exhausted T cells over a 7-d period in patients with mild/moderate and severe COVID-19. Altogether, the study suggests that accelerated immunosenescence in CD4 and especially CD8 T-cell compartments may represent a common and unique outcome of SARS-CoV2 infection.
Collapse
Affiliation(s)
- Rodrigo Balsinha Pedroso
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649, 1649-028, Lisboa, Portugal
| | - Lícia Torres
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Lucas Araújo Ventura
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Giovanna Caliman Camatta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Catarina Mota
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649, 1649-028, Lisboa, Portugal
| | - Ana Catarina Mendes
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649, 1649-028, Lisboa, Portugal
| | - Filipa Ribeiro
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649, 1649-028, Lisboa, Portugal
| | | | - Rafael Calvão Barbuto
- Hospital Risoleta Tolentino Neves, R. das Gabirobas, 1, 31744-012, Belo Horizonte, MG, Brazil
| | - Felipe Caixeta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Leandro Souza Nascimento
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Mariana Almeida Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Vinícius Dantas Martins
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| | - Gabriela Silveira-Nunes
- Departamento de Medicina, Universidade Federal de Juiz de Fora (UFJF), Av. Doutor Raimundo Monteiro Resende, Governador Valadares, 35010-177, MG, Brazil
| | - Unaí Tupinambás
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190, Belo Horizonte, 30130-100, MG, Brazil
| | - Andrea Teixeira-Carvalho
- Laboratório de Biomarcadores, Instituto de Pesquisa René Rachou, Fundação Oswaldo Cruz, FIOCRUZ-MG, Av. Augusto de Lima, 1715, Belo Horizonte, 30190-002, MG, Brazil
| | - Luis Graça
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649, 1649-028, Lisboa, Portugal
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Av. Antonio Carlos, 6627, 31270-901, Belo Horizonte, MG, Brazil
| |
Collapse
|
5
|
Andrejkovits ÁV, Huțanu A, Manu DR, Dobreanu M, Văsieșiu AM. Dynamic Changes in Lymphocyte Populations and Their Relationship with Disease Severity and Outcome in COVID-19. Int J Mol Sci 2024; 25:11921. [PMID: 39595989 PMCID: PMC11593669 DOI: 10.3390/ijms252211921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/26/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Studies suggest that the dynamic changes in cellular response might correlate with disease severity and outcomes in SARS-CoV-2 patients. The study aimed to investigate the dynamic changes of lymphocyte subsets in patients with COVID-19. In this regard, 53 patients with COVID-19 were prospectively included, classified as mild, moderate, and severe. The peripheral lymphocyte profiles (LyT, LyB, and NK cells), as well as CD4+/CD8+, CD3+/CD19+, CD3+/NK and CD19+/NK ratios, and their dynamic changes during hospitalization and correlation with disease severity and outcome were assessed. We found significant differences in CD3+ lymphocytes between severity groups (p < 0.0001), with significantly decreased CD3+CD4+ and CD3+CD8+ in patients with severe disease (p < 0.0001 and p = 0.048, respectively). Lower CD3+/CD19+ and CD3+/NK ratios among patients with severe disease (p = 0.019 and p = 0.010, respectively) were found. The dynamic changes of lymphocyte subsets showed a significant reduction in NK cells (%) and a significant increase in CD3+CD4+ and CD3+CD8+ cells in patients with moderate and severe disease. The ROC analysis on the relationship between CD3+ cells and fatal outcome yielded an AUC of 0.723 (95% CI 0.583-0.837; p = 0.007), while after addition of age and SpO2, ferritin and NLR, the AUC significantly improved to 0.927 (95%CI 0.811-0.983), p < 0.001 with a sensitivity of 90.9% (95% CI 58.7-99.8%) and specificity of 85.7% (95% CI 69.7-95.2%). The absolute number of CD3+ lymphocytes might independently predict fatal outcomes in COVID-19 patients and T-lymphocyte subset evaluation in high-risk patients might be useful in estimating disease progression.
Collapse
Affiliation(s)
- Ákos Vince Andrejkovits
- Department of Infectious Disease, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540139 Târgu Mureș, Romania; (Á.V.A.); (A.M.V.)
- Doctoral School of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540139 Târgu Mureș, Romania
| | - Adina Huțanu
- Department of Laboratory Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540139 Târgu Mureș, Romania;
- Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540139 Târgu Mureș, Romania;
| | - Doina Ramona Manu
- Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540139 Târgu Mureș, Romania;
| | - Minodora Dobreanu
- Department of Laboratory Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540139 Târgu Mureș, Romania;
- Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540139 Târgu Mureș, Romania;
| | - Anca Meda Văsieșiu
- Department of Infectious Disease, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540139 Târgu Mureș, Romania; (Á.V.A.); (A.M.V.)
- 1st Infectious Disease Clinic of County Clinical Hospital, 540146 Târgu Mureș, Romania
| |
Collapse
|
6
|
Chen Y, Deng H, Zhou R, Jiang X, Wang H, Xin S, Mo W, Wang S, Liu Y. Comprehensive mapping of immune perturbations associated with secondary hemophagocytic lymphohistiocytosis. J Leukoc Biol 2024; 116:1109-1126. [PMID: 38973235 DOI: 10.1093/jleuko/qiae138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/02/2024] [Accepted: 06/13/2024] [Indexed: 07/09/2024] Open
Abstract
Secondary hemophagocytic lymphohistiocytosis (sHLH) is a hyperinflammatory syndrome characterized by immune disorders. It is imperative to elucidate the immunophenotypic panorama and the interactions among these cells in patients. Human peripheral blood mononuclear cells were collected from healthy donors and sHLH patients and tested using multicolor flow cytometry. We used FlowSOM to explore and visualize the immunophenotypic characteristics of sHLH. By demonstrating the phenotypes of immune cells, we discovered that sHLH patients had significantly higher levels of CD56+ monocytes, higher levels of myeloid-derived suppressor cells, low-density neutrophil-to-T cell ratio, and higher heterogeneous T cell activation than healthy donors. However, natural killer cell cytotoxicity and function were impaired. We then assessed the correlations among 30 immune cell types and evaluated metabolic analysis. Our findings demonstrated polymorphonuclear myeloid-derived suppressor cells, CD56+ monocytes, and neutrophil-to-T cell ratio were elevated abnormally in sHLH patients, which may indicate an association with immune overactivation and inflammatory response. We are expected to confirm that they are involved in the occurrence of the disease through further in-depth research.
Collapse
Affiliation(s)
- Yinchun Chen
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Guangzhou 510180, China
- Department of Hematology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou 510180, China
| | - Haimei Deng
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628, Zhenyuan Road, Xinhuling Street, Shenzhen 518118, China
| | - Ruiqing Zhou
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Guangzhou 510180, China
- Department of Hematology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou 510180, China
| | - Xiaotao Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 16 Airport Road, Guangzhou 510400, China
| | - Huijuan Wang
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, No. 10 Huan Yu Second Road, Guangzhou 510180, China
| | - Songqing Xin
- Changan Hospital of Dongguan, No. 171 Changqing South Road, Dongguan 523850, China
| | - Wenjian Mo
- Department of Hematology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou 510180, China
| | - Shunqing Wang
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Guangzhou 510180, China
- Department of Hematology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou 510180, China
| | - Yufeng Liu
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Guangzhou 510180, China
- Department of Hematology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou 510180, China
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, No. 10 Huan Yu Second Road, Guangzhou 510180, China
| |
Collapse
|
7
|
Ustiuzhanina MO, Boyko AA, Vavilova JD, Siniavin AE, Streltsova MA, Astrakhantseva IV, Drutskaya MS, Chudakov DM, Kovalenko EI. The Antigen-Specific Response of NK Cells to SARS-CoV-2 Correlates With KIR2DS4 Expression. J Med Virol 2024; 96:e70057. [PMID: 39540437 DOI: 10.1002/jmv.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/03/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Natural killer (NK) cells play a pivotal role in the immune response against viral infections, including SARS-CoV-2. However, our understanding of memory NK cell responses in the context of SARS-CoV-2 remains limited. To address this, we investigated the memory-like response of NK cells to SARS-CoV-2 peptides, presented by autologous cells. Blood samples from 45 donors underwent analysis for SARS-CoV-2 IgG antibodies, categorizing them into four groups based on the antibody kind and level. NK cells from SARS-CoV-2-experienced donors demonstrated enhanced degranulation and activation levels, IFNγ production and proliferative potential in response to SARS-CoV-2 peptides. Investigation of highly proliferating NK cells demonstrated the formation of distinct clusters depending on the SARS-CoV-2 peptide supplementation and the donor group. RNA sequencing revealed differential gene expression patterns, highlighting metabolism, protein transport, and immune response genes. Notably, KIR2DS4 expression correlated with enhanced IFNγ production, degranulation and proliferation levels, suggesting a role in SARS-CoV-2 recognition. Collectively, these findings provide detailed insights into antigen-specific NK cell responses to SARS-CoV-2 peptides, indicating potential mechanisms underlying NK cell activation in antiviral immunity.
Collapse
Affiliation(s)
- M O Ustiuzhanina
- Department of Genomics of Adaptive Immunity, Department of immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - A A Boyko
- Department of Genomics of Adaptive Immunity, Department of immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - J D Vavilova
- Department of Genomics of Adaptive Immunity, Department of immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - A E Siniavin
- Department of Genomics of Adaptive Immunity, Department of immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Arbovirus and Experimental Production Department, Federal State Budget Institution "National Research Centre for Epidemiology and Microbiology Named After Honorary Academician N. F. Gamaleya" of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - M A Streltsova
- Department of Genomics of Adaptive Immunity, Department of immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - I V Astrakhantseva
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Russia
| | - M S Drutskaya
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - D M Chudakov
- Department of Genomics of Adaptive Immunity, Department of immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department Bioinformatics, Abu Dhabi Stem Cell Center, Al Muntazah, Abu Dhabi, United Arab Emirates
| | - E I Kovalenko
- Department of Genomics of Adaptive Immunity, Department of immunology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
8
|
de Los Rios Kobara I, Jayewickreme R, Lee MJ, Wilk AJ, Blomkalns AL, Nadeau KC, Yang S, Rogers AJ, Blish CA. Interferon-mediated NK cell activation is associated with limited neutralization breadth during SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619639. [PMID: 39484382 PMCID: PMC11527016 DOI: 10.1101/2024.10.22.619639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Best known for their ability to kill infected or malignant cells, natural killer (NK) cells are also underappreciated regulators of the antibody response to viral infection. In mice, NK cells can kill T follicular helper (Tfh) cells, decreasing somatic hypermutation and vaccine responses. Although human NK cell activation correlates with poor vaccine response, the mechanisms of human NK cell regulation of adaptive immunity are poorly understood. We found that in human ancestral SARS-CoV-2 infection, broad neutralizers, who were capable of neutralizing Alpha, Beta, and Delta, had fewer NK cells that expressed inhibitory and immaturity markers whereas NK cells from narrow neutralizers were highly activated and expressed interferon-stimulated genes (ISGs). ISG-mediated activation in NK cells from healthy donors increased cytotoxicity and functional responses to induced Tfh-like cells. This work reveals that NK cell activation and dysregulated inflammation may play a role in poor antibody response to SARS-CoV-2 and opens exciting avenues for designing improved vaccines and adjuvants to target emerging pathogens.
Collapse
|
9
|
Syrimi E, Khan N, Murray P, Willcox C, Haigh T, Willcox B, Masand N, Bowen C, Dimakou DB, Zuo J, Barone SM, Irish JM, Kearns P, Taylor GS. Defects in NK cell immunity of pediatric cancer patients revealed by deep immune profiling. iScience 2024; 27:110837. [PMID: 39310750 PMCID: PMC11416690 DOI: 10.1016/j.isci.2024.110837] [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: 07/06/2023] [Revised: 12/21/2023] [Accepted: 08/24/2024] [Indexed: 09/25/2024] Open
Abstract
Systemic immunity plays an important role in cancer immune surveillance and response to therapy, but little is known about the immune status of children with solid cancers. We performed a high-dimensional single-cell analysis of systemic immunity in 50 treatment-naive pediatric cancer patients, comparing them to age-matched healthy children. Children with cancer had a lower frequency of peripheral NK cells, which was not due to tumor sequestration, had lower surface levels of activating receptors and increased levels of the inhibitory NKG2A receptor. Furthermore, the natural killer (NK) cells of pediatric cancer patients were less mature and less cytotoxic when tested in vitro. Culture of these NK cells with interleukin-2 restored their cytotoxicity. Collectively, our data show that NK cells in pediatric cancer patients are impaired through multiple mechanisms and identify rational strategies to restore their functionality.
Collapse
Affiliation(s)
- Eleni Syrimi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Naeem Khan
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Paul Murray
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Health research Institute, University of Limerick, Limerick, Ireland
| | - Carrie Willcox
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Tracey Haigh
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Benjamin Willcox
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Navta Masand
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Claire Bowen
- Pathology department, Birmingham Children’s Hospital, Birmingham, UK
| | - Danai B. Dimakou
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Sierra M. Barone
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan M. Irish
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pamela Kearns
- Cancer Research UK Clinical Trials Unit, National Institute for Health Research Birmingham Biomedical Research Centre, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Graham S. Taylor
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| |
Collapse
|
10
|
Silva-Junior AL, Oliveira LS, Dias S, Costa TCC, Xabregas LA, Alves-Hanna FS, Abrahim CMM, Neves WLL, Crispim MAE, Toro DM, Silva-Neto PV, Aponte DCM, Oliveira TC, Silva MCC, Matos MMM, Carvalho MPSS, Tarragô AM, Fraiji NA, Faccioli LH, Sorgi CA, Sabino EC, Teixeira-Carvalho A, Martins-Filho OA, Costa AG, Malheiro A. Immunologic mediators profile in COVID-19 convalescence. Sci Rep 2024; 14:20930. [PMID: 39251702 PMCID: PMC11384766 DOI: 10.1038/s41598-024-71419-x] [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: 04/13/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024] Open
Abstract
SARS-CoV-2 caused the pandemic situation experienced since the beginning of 2020, and many countries faced the rapid spread and severe form of the disease. Mechanisms of interaction between the virus and the host were observed during acute phase, but few data are available when related to immunity dynamics in convalescents. We conducted a longitudinal study, with 51 healthy donors and 62 COVID-19 convalescent patients, which these had a 2-month follow-up after symptoms recovery. Venous blood sample was obtained from all participants to measure blood count, subpopulations of monocytes, lymphocytes, natural killer cells and dendritic cells. Serum was used to measure cytokines, chemokines, growth factors, anti-N IgG and anti-S IgG/IgM antibodies. Statistic was performed by Kruskal-Wallis test, and linear regression with days post symptoms and antibody titers. All analysis had confidence interval of 95%. Less than 35% of convalescents were anti-S IgM+, while more than 80% were IgG+ in D30. Anti-N IgG decreased along time, with loss of seroreactivity of 13%. Eosinophil count played a distinct role on both antibodies during all study, and the convalescence was orchestrated by higher neutrophil-to-lymphocyte ratio and IL-15, but initial stages were marked by increase in myeloid DCs, B1 lymphocytes, inflammatory and patrolling monocytes, G-CSF and IL-2. Later convalescence seemed to change to cytotoxicity mediated by T lymphocytes, plasmacytoid DCs, VEGF, IL-9 and CXCL10. Anti-S IgG antibodies showed the longest perseverance and may be a better option for diagnosis. The inflammatory pattern is yet present on initial stage of convalescence, but quickly shifts to a reparative dynamic. Meanwhile eosinophils seem to play a role on anti-N levels in convalescence, although may not be the major causative agent. We must highlight the importance of immunological markers on acute clinical outcomes, but their comprehension to potentialize adaptive system must be explored to improve immunizations and further preventive policies.
Collapse
Affiliation(s)
- Alexander Leonardo Silva-Junior
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Lucas Silva Oliveira
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Stephanny Dias
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil
| | - Thaina Cristina Cardoso Costa
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Lilyane Amorim Xabregas
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Fabíola Silva Alves-Hanna
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | - Cláudia Maria Moura Abrahim
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Walter Luiz Lima Neves
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Myuki Alfaia Esashika Crispim
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Diana Mota Toro
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | - Pedro Vieira Silva-Neto
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | | | | | | | | | | | - Andrea Monteiro Tarragô
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil
- Rede Genômica em Saúde do Estado do Amazonas (REGESAM), Manaus, AM, Brazil
| | - Nelson Abrahim Fraiji
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Lúcia Helena Faccioli
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Carlos Artério Sorgi
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | | | - Andrea Teixeira-Carvalho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil
| | - Allyson Guimarães Costa
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil.
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil.
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil.
- Rede Genômica em Saúde do Estado do Amazonas (REGESAM), Manaus, AM, Brazil.
| | - Adriana Malheiro
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil.
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil.
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil.
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil.
- Rede Genômica em Saúde do Estado do Amazonas (REGESAM), Manaus, AM, Brazil.
| |
Collapse
|
11
|
Yang X, Mann KK, Wu H, Ding J. scCross: a deep generative model for unifying single-cell multi-omics with seamless integration, cross-modal generation, and in silico exploration. Genome Biol 2024; 25:198. [PMID: 39075536 PMCID: PMC11285326 DOI: 10.1186/s13059-024-03338-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 07/16/2024] [Indexed: 07/31/2024] Open
Abstract
Single-cell multi-omics data reveal complex cellular states, providing significant insights into cellular dynamics and disease. Yet, integration of multi-omics data presents challenges. Some modalities have not reached the robustness or clarity of established transcriptomics. Coupled with data scarcity for less established modalities and integration intricacies, these challenges limit our ability to maximize single-cell omics benefits. We introduce scCross, a tool leveraging variational autoencoders, generative adversarial networks, and the mutual nearest neighbors (MNN) technique for modality alignment. By enabling single-cell cross-modal data generation, multi-omics data simulation, and in silico cellular perturbations, scCross enhances the utility of single-cell multi-omics studies.
Collapse
Affiliation(s)
- Xiuhui Yang
- School of Software, Shandong University, 1500 Shunhua, Jinan, 250101, Shandong, China
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, H4A 3J1, QC, Canada
- Quantitative Life Sciences, Faculty of Medicine & Health Sciences, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Koren K Mann
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Hao Wu
- School of Software, Shandong University, 1500 Shunhua, Jinan, 250101, Shandong, China.
| | - Jun Ding
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, H4A 3J1, QC, Canada.
- Quantitative Life Sciences, Faculty of Medicine & Health Sciences, McGill University, Montreal, QC, H3G 1Y6, Canada.
- Mila-Quebec AI Institute, Montreal, QC, H2S 3H1, Canada.
| |
Collapse
|
12
|
Lee MJ, de los Rios Kobara I, Barnard TR, Vales Torres X, Tobin NH, Ferbas KG, Rimoin AW, Yang OO, Aldrovandi GM, Wilk AJ, Fulcher JA, Blish CA. NK Cell-Monocyte Cross-talk Underlies NK Cell Activation in Severe COVID-19. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1693-1705. [PMID: 38578283 PMCID: PMC11102029 DOI: 10.4049/jimmunol.2300731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/13/2024] [Indexed: 04/06/2024]
Abstract
NK cells in the peripheral blood of severe COVID-19 patients exhibit a unique profile characterized by activation and dysfunction. Previous studies have identified soluble factors, including type I IFN and TGF-β, that underlie this dysregulation. However, the role of cell-cell interactions in modulating NK cell function during COVID-19 remains unclear. To address this question, we combined cell-cell communication analysis on existing single-cell RNA sequencing data with in vitro primary cell coculture experiments to dissect the mechanisms underlying NK cell dysfunction in COVID-19. We found that NK cells are predicted to interact most strongly with monocytes and that this occurs via both soluble factors and direct interactions. To validate these findings, we performed in vitro cocultures in which NK cells from healthy human donors were incubated with monocytes from COVID-19+ or healthy donors. Coculture of healthy NK cells with monocytes from COVID-19 patients recapitulated aspects of the NK cell phenotype observed in severe COVID-19, including decreased expression of NKG2D, increased expression of activation markers, and increased proliferation. When these experiments were performed in a Transwell setting, we found that only CD56bright CD16- NK cells were activated in the presence of severe COVID-19 patient monocytes. O-link analysis of supernatants from Transwell cocultures revealed that cultures containing severe COVID-19 patient monocytes had significantly elevated levels of proinflammatory cytokines and chemokines, as well as TGF-β. Collectively, these results demonstrate that interactions between NK cells and monocytes in the peripheral blood of COVID-19 patients contribute to NK cell activation and dysfunction in severe COVID-19.
Collapse
Affiliation(s)
- Madeline J. Lee
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
- Stanford Immunology Program, Stanford University School of Medicine, Palo Alto, CA
| | - Izumi de los Rios Kobara
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
- Stanford Immunology Program, Stanford University School of Medicine, Palo Alto, CA
| | - Trisha R. Barnard
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - Xariana Vales Torres
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
- Stanford Immunology Program, Stanford University School of Medicine, Palo Alto, CA
| | - Nicole H. Tobin
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Kathie G. Ferbas
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Anne W. Rimoin
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA
| | - Otto O. Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Grace M. Aldrovandi
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Aaron J. Wilk
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Palo Alto, CA
| | - Jennifer A. Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Catherine A. Blish
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
- Chan Zuckerberg Biohub, San Francisco, CA
| |
Collapse
|
13
|
Zhang N, Wang Z, Li H, Chen K, Wang HS, Shao JB, Jiang SY, Zhai XW, Jiang H. Immunophenotype of lymphocytes and real-world outcome of COVID-19 infection in children with hematology and oncology. BMC Cancer 2024; 24:538. [PMID: 38678181 PMCID: PMC11056048 DOI: 10.1186/s12885-024-12262-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Patients with immunocompromise were suspected to encounter a high risk for severe coronavirus disease 2019 (COVID-19) infection on early period; however, data is lacking nowadays and immune response remain unclear. METHODS In this retrospective study, internet questionnaire survey and medical records were acquired in pediatric hematology oncology patients. Clinical severity, immunological characteristics, and outcomes were analyzed from December 1, 2022 to January 31, 2023 at the 3rd year of pandemic in China. RESULTS A total of 306 patients were included, with 21 patients (6.9%) asymptomatic, 262 (85.6%) mild severity, 17 (5.6%) moderate severity, 5 (1.6%) severe severity, and 1 (0.3%) critical severity. Seventy-eight (25.5%) patients were on intensive chemotherapy, and 32.0% children were on maintenance chemotherapy. Delays in cancer therapy occurred in 86.7% patients. Univariable analysis revealed active chemotherapy (P < 0.0001), long duration of symptom (P < 0.0001), low lymphocytes count (P = 0.095), low CD3 + and CD8 + T cell count (P = 0.013, P = 0.022), high percentage of CD4 + TCM (P = 0.016), and low percentage of transitional B cells (P = 0.045) were high risk factors for severe COVID-19 infection. Cox regression model showed that the absolute lymphocytes count (P = 0.027) and long duration of symptom (P = 0.002) were the independent factors for severity. Patients with CD8 + dominant and B cell depletion subtype wasn't related with severity, but had higher percentage of CD8 + effector memory T cells (TEM) and terminally differentiated effector memory T cells (TEMRA) (P < 0.001, P < 0.001), and a longer COVID-19 duration (P = 0.045). CONCLUSION The severity was relatively mild in children with immunodeficiencies in the third year of COVID-19 pandemic. Low lymphocyte count and long duration of symptom were the independent risk factors with COVID-19 severity. Delays in cancer care remain a major concern and the long outcome is pending.
Collapse
Affiliation(s)
- Na Zhang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Zhen Wang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Hong Li
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Kai Chen
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Hong-Sheng Wang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Jing-Bo Shao
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Sha-Yi Jiang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China
| | - Xiao-Wen Zhai
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Hui Jiang
- Department of Hematology and Oncology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, 200040, Shanghai, China.
| |
Collapse
|
14
|
Wiśniewska A, Kijak A, Nowak K, Lulek M, Skwarek A, Małecka-Giełdowska M, Śmiarowski M, Wąsik S, Ciepiela O. Organ-Dysfunction Markers in Mild-to-Moderate COVID-19 Convalescents. J Clin Med 2024; 13:2241. [PMID: 38673514 PMCID: PMC11050795 DOI: 10.3390/jcm13082241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/01/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Background: A coronavirus disease 2019 (COVID-19) outbreak led to a worldwide pandemic. COVID-19 not only caused acute symptoms during the severe phase of the disease, but also induced long-term side effects on the functioning of many organs and systems. Symptoms that were associated with the disease and present at least 3 months after recovery were named long COVID. The aim of this study was to assess if mild-to-moderate COVID-19 may lead to the dysfunction of respiratory, cardiovascular, neural, and renal systems in healthy blood donors who recovered from the disease at least 6 months earlier. Methods: Here, we examined 294 adults among volunteer blood donors divided into convalescents (n = 215) and healthy controls (n = 79). Concentrations of soluble CD163, TGF beta, Lp-PLA2, NCAM-1, S100, NGAL, and creatinine were measured either by ELISA or automated methods. The probability value p < 0.05 was considered as statistically significant. Results: We found significant differences in Lp-PLA2, S100, and NCAM-1 between convalescents and never-infected subjects. Lp-PLA2 and NCAM-1 were lower, and S100 higher, in convalescents than in the control group. Conclusion: Mild-to-moderate COVID-19 convalescents are at a low risk of developing lung fibrosis or chronic kidney disease. However, they should regularly carry out their prophylaxis examinations for early detection of possible negative outcomes of COVID-19.
Collapse
Affiliation(s)
- Aleksandra Wiśniewska
- Students Scientific Group of Laboratory Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland (M.L.); (A.S.); (M.Ś.); (S.W.)
| | - Aleksandra Kijak
- Students Scientific Group of Laboratory Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland (M.L.); (A.S.); (M.Ś.); (S.W.)
| | - Karolina Nowak
- Students Scientific Group of Laboratory Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland (M.L.); (A.S.); (M.Ś.); (S.W.)
| | - Michalina Lulek
- Students Scientific Group of Laboratory Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland (M.L.); (A.S.); (M.Ś.); (S.W.)
- Clinical Laboratory of Central Teaching Hospital, University Clinical Center of Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Agata Skwarek
- Students Scientific Group of Laboratory Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland (M.L.); (A.S.); (M.Ś.); (S.W.)
| | - Milena Małecka-Giełdowska
- Clinical Laboratory of Central Teaching Hospital, University Clinical Center of Medical University of Warsaw, 02-097 Warsaw, Poland;
- Department of Laboratory Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Marcin Śmiarowski
- Students Scientific Group of Laboratory Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland (M.L.); (A.S.); (M.Ś.); (S.W.)
| | - Szczepan Wąsik
- Students Scientific Group of Laboratory Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland (M.L.); (A.S.); (M.Ś.); (S.W.)
| | - Olga Ciepiela
- Clinical Laboratory of Central Teaching Hospital, University Clinical Center of Medical University of Warsaw, 02-097 Warsaw, Poland;
- Department of Laboratory Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
15
|
Fernández-Soto D, García-Jiménez ÁF, Casasnovas JM, Valés-Gómez M, Reyburn HT. Elevated levels of cell-free NKG2D-ligands modulate NKG2D surface expression and compromise NK cell function in severe COVID-19 disease. Front Immunol 2024; 15:1273942. [PMID: 38410511 PMCID: PMC10895954 DOI: 10.3389/fimmu.2024.1273942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/22/2024] [Indexed: 02/28/2024] Open
Abstract
Introduction It is now clear that coronavirus disease 19 (COVID-19) severity is associated with a dysregulated immune response, but the relative contributions of different immune cells is still not fully understood. SARS CoV-2 infection triggers marked changes in NK cell populations, but there are contradictory reports as to whether these effector lymphocytes play a protective or pathogenic role in immunity to SARS-CoV-2. Methods To address this question we have analysed differences in the phenotype and function of NK cells in SARS-CoV-2 infected individuals who developed either very mild, or life-threatening COVID-19 disease. Results Although NK cells from patients with severe disease appeared more activated and the frequency of adaptive NK cells was increased, they were less potent mediators of ADCC than NK cells from patients with mild disease. Further analysis of peripheral blood NK cells in these patients revealed that a population of NK cells that had lost expression of the activating receptor NKG2D were a feature of patients with severe disease and this correlated with elevated levels of cell free NKG2D ligands, especially ULBP2 and ULBP3 in the plasma of critically ill patients. In vitro, culture in NKG2DL containing patient sera reduced the ADCC function of healthy donor NK cells and this could be blocked by NKG2DL-specific antibodies. Discussion These observations of reduced NK function in severe disease are consistent with the hypothesis that defects in immune surveillance by NK cells permit higher levels of viral replication, rather than that aberrant NK cell function contributes to immune system dysregulation and immunopathogenicity.
Collapse
Affiliation(s)
- Daniel Fernández-Soto
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB), Spanish National Research Council (CSIC), Madrid, Spain
| | - Álvaro F. García-Jiménez
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB), Spanish National Research Council (CSIC), Madrid, Spain
| | - José M. Casasnovas
- Department of Macromolecular Structures, National Centre for Biotechnology (CNB), Spanish National Research Council (CSIC), Madrid, Spain
| | - Mar Valés-Gómez
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB), Spanish National Research Council (CSIC), Madrid, Spain
| | - Hugh T. Reyburn
- Department of Immunology and Oncology, National Centre for Biotechnology (CNB), Spanish National Research Council (CSIC), Madrid, Spain
| |
Collapse
|
16
|
Farias TD, Brugiapaglia S, Croci S, Magistroni P, Curcio C, Zguro K, Fallerini C, Fava F, Pettini F, Kichula KM, Pollock NR, Font-Porterias N, Palmer WH, Marin WM, Baldassarri M, Bruttini M, Hollenbach JA, Hendricks AE, Meloni I, Novelli F, Renieri A, Furini S, Norman PJ, Amoroso A. HLA-DPB1*13:01 associates with enhanced, and KIR2DS4*001 with diminished protection from developing severe COVID-19. HLA 2024; 103:e15251. [PMID: 37850268 PMCID: PMC10873037 DOI: 10.1111/tan.15251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 08/22/2023] [Accepted: 09/26/2023] [Indexed: 10/19/2023]
Abstract
Extreme polymorphism of HLA and killer-cell immunoglobulin-like receptors (KIR) differentiates immune responses across individuals. Additional to T cell receptor interactions, subsets of HLA class I act as ligands for inhibitory and activating KIR, allowing natural killer (NK) cells to detect and kill infected cells. We investigated the impact of HLA and KIR polymorphism on the severity of COVID-19. High resolution HLA class I and II and KIR genotypes were determined from 403 non-hospitalized and 1575 hospitalized SARS-CoV-2 infected patients from Italy collected in 2020. We observed that possession of the activating KIR2DS4*001 allotype is associated with severe disease, requiring hospitalization (OR = 1.48, 95% CI 1.20-1.85, pc = 0.017), and this effect is greater in individuals homozygous for KIR2DS4*001 (OR = 3.74, 95% CI 1.75-9.29, pc = 0.003). We also observed the HLA class II allotype, HLA-DPB1*13:01 protects SARS-CoV-2 infected patients from severe disease (OR = 0.49, 95% CI 0.33-0.74, pc = 0.019). These association analyses were replicated using logistic regression with sex and age as covariates. Autoantibodies against IFN-α associated with COVID-19 severity were detected in 26% of 156 hospitalized patients tested. HLA-C*08:02 was more frequent in patients with IFN-α autoantibodies than those without, and KIR3DL1*01502 was only present in patients lacking IFN-α antibodies. These findings suggest that KIR and HLA polymorphism is integral in determining the clinical outcome following SARS-CoV-2 infection, by influencing the course both of innate and adaptive immunity.
Collapse
Affiliation(s)
- Ticiana D.J. Farias
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Silvia Brugiapaglia
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Laboratory of Tumor Immunology Center for Experimental Research and Medical Studies, Città della Salute e della Scienza di Torino, Turin, 10126, Italy
| | - Susanna Croci
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Paola Magistroni
- Immunogenetics and Transplant Biology, Azienda Ospedaliera Universitaria, Città della Salute e della Scienza di Torino, Turin, 10126, Italy
| | - Claudia Curcio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Laboratory of Tumor Immunology Center for Experimental Research and Medical Studies, Città della Salute e della Scienza di Torino, Turin, 10126, Italy
| | - Kristina Zguro
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Chiara Fallerini
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Francesca Fava
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, 53100, Italy
| | - Francesco Pettini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, 53100, Italy
| | - Katherine M. Kichula
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Nicholas R. Pollock
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Neus Font-Porterias
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - William H. Palmer
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Wesley M. Marin
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Margherita Baldassarri
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Mirella Bruttini
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, 53100, Italy
| | - Jill A. Hollenbach
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Audrey E. Hendricks
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Mathematical and Statistical Sciences, and Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Ilaria Meloni
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Laboratory of Tumor Immunology Center for Experimental Research and Medical Studies, Città della Salute e della Scienza di Torino, Turin, 10126, Italy
- Molecular Biotechnology Center, University of Turin, Turin, 10126, Italy
| | | | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, 53100, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, 53100, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, 53100, Italy
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, 53100, Italy
| | - Paul J. Norman
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, 80045, USA
| | - Antonio Amoroso
- Immunogenetics and Transplant Biology, Azienda Ospedaliera Universitaria, Città della Salute e della Scienza di Torino, Turin, 10126, Italy
- Department of Medical Sciences, University of Turin, Turin, 10126, Italy
| |
Collapse
|
17
|
Bjorgen JC, Dick JK, Cromarty R, Hart GT, Rhein J. NK cell subsets and dysfunction during viral infection: a new avenue for therapeutics? Front Immunol 2023; 14:1267774. [PMID: 37928543 PMCID: PMC10620977 DOI: 10.3389/fimmu.2023.1267774] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
In the setting of viral challenge, natural killer (NK) cells play an important role as an early immune responder against infection. During this response, significant changes in the NK cell population occur, particularly in terms of their frequency, location, and subtype prevalence. In this review, changes in the NK cell repertoire associated with several pathogenic viral infections are summarized, with a particular focus placed on changes that contribute to NK cell dysregulation in these settings. This dysregulation, in turn, can contribute to host pathology either by causing NK cells to be hyperresponsive or hyporesponsive. Hyperresponsive NK cells mediate significant host cell death and contribute to generating a hyperinflammatory environment. Hyporesponsive NK cell populations shift toward exhaustion and often fail to limit viral pathogenesis, possibly enabling viral persistence. Several emerging therapeutic approaches aimed at addressing NK cell dysregulation have arisen in the last three decades in the setting of cancer and may prove to hold promise in treating viral diseases. However, the application of such therapeutics to treat viral infections remains critically underexplored. This review briefly explores several therapeutic approaches, including the administration of TGF-β inhibitors, immune checkpoint inhibitors, adoptive NK cell therapies, CAR NK cells, and NK cell engagers among other therapeutics.
Collapse
Affiliation(s)
- Jacob C. Bjorgen
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Jenna K. Dick
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Ross Cromarty
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Geoffrey T. Hart
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Joshua Rhein
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
18
|
Rutkowska E, Kwiecień I, Pietruszka-Wałęka E, Więsik-Szewczyk E, Rzepecki P, Jahnz-Różyk K. Analysis of Leukocyte Subpopulations by Flow Cytometry during Hospitalization Depending on the Severity of COVID-19 Course. Biomedicines 2023; 11:2728. [PMID: 37893102 PMCID: PMC10604221 DOI: 10.3390/biomedicines11102728] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
The mechanisms underlying the immune response to coronavirus disease 2019 (COVID-19) and the recovery process have not been fully elucidated. The aim of the study was to analyze leukocyte subpopulations in patients at significant time points (at diagnosis, and 3 and 6 months after infection) selected according to the analysis of changes in the lungs by the CT classification system, considering the severity of the disease. The study groups consisted of severe and non-severe COVID-19 patients. There was a significant decrease in CD8+ T cells, NK and eosinophils, with an increasing percentage of neutrophils during hospitalization. We noticed lower levels of CD4 and CD8 T lymphocytes, eosinophils, basophils, and CD16+ monocytes and elevated neutrophil levels in severe patients relative to non-severe patients. Three months after infection, we observed higher levels of basophils, and after 6 months, higher CD4/CD8 ratios and T cell levels in the severe compared to non-severe group. Non-severe patients showed significant changes in the leukocyte populations studied at time of hospitalization and both within 3 months and 6 months of onset. The CT CSS classification with parameters of the flow cytometry method used for COVID-19 patients determined changes that proved useful in the initial evaluation of patients.
Collapse
Affiliation(s)
- Elżbieta Rutkowska
- Laboratory of Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland;
| | - Iwona Kwiecień
- Laboratory of Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland;
| | - Ewa Pietruszka-Wałęka
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland; (E.P.-W.); (E.W.-S.); (K.J.-R.)
| | - Ewa Więsik-Szewczyk
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland; (E.P.-W.); (E.W.-S.); (K.J.-R.)
| | - Piotr Rzepecki
- Department of Internal Medicine and Hematology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland;
| | - Karina Jahnz-Różyk
- Department of Internal Medicine, Pulmonology, Allergology and Clinical Immunology, Military Institute of Medicine Warsaw—National Research Institute, 04-141 Warsaw, Poland; (E.P.-W.); (E.W.-S.); (K.J.-R.)
| |
Collapse
|
19
|
Muyayalo KP, Gong GS, Kiyonga Aimeé K, Liao AH. Impaired immune response against SARS-CoV-2 infection is the major factor indirectly altering reproductive function in COVID-19 patients: a narrative review. HUM FERTIL 2023; 26:778-796. [PMID: 37811836 DOI: 10.1080/14647273.2023.2262757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/25/2023] [Indexed: 10/10/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease affecting multiple systems and organs, including the reproductive system. SARS-CoV-2, the virus that causes COVID-19, can damage reproductive organs through direct (angiotensin converting enzyme-2, ACE-2) and indirect mechanisms. The immune system plays an essential role in the homeostasis and function of the male and female reproductive systems. Therefore, an altered immune response related to infectious and inflammatory diseases can affect reproductive function and fertility in both males and females. This narrative review discussed the dysregulation of innate and adaptive systems induced by SARS-CoV-2 infection. We reviewed the evidence showing that this altered immune response in COVID-19 patients is the major indirect mechanism leading to adverse reproduction outcomes in these patients. We summarized studies reporting the long-term effect of SARS-CoV-2 infection on women's reproductive function and proposed the chronic inflammation and chronic autoimmunity characterizing long COVID as potential underlying mechanisms. Further studies are needed to clarify the role of autoimmunity and chronic inflammation (long COVID) in altered female reproduction function in COVID-19.
Collapse
Affiliation(s)
- Kahindo P Muyayalo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Department of Obstetrics and Gynecology, University of Kinshasa, Kinshasa, D. R. Congo
| | - Guang-Shun Gong
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Kahindo Kiyonga Aimeé
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, People's Republic of China
- Department of Tropical Medicine Infectious and Parasitic Diseases, University of Kinshasa, Kinshasa, D. R. Congo
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| |
Collapse
|
20
|
Lee MJ, Blish CA. Defining the role of natural killer cells in COVID-19. Nat Immunol 2023; 24:1628-1638. [PMID: 37460639 PMCID: PMC10538371 DOI: 10.1038/s41590-023-01560-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/08/2023] [Indexed: 09/20/2023]
Abstract
Natural killer (NK) cells are critical effectors of antiviral immunity. Researchers have therefore sought to characterize the NK cell response to coronavirus disease 2019 (COVID-19) and the virus that causes it, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The NK cells of patients with severe COVID-19 undergo extensive phenotypic and functional changes. For example, the NK cells from critically ill patients with COVID-19 are highly activated and exhausted, with poor cytotoxic function and cytokine production upon stimulation. The NK cell response to SARS-CoV-2 is also modulated by changes induced in virally infected cells, including the ability of a viral peptide to bind HLA-E, preventing NK cells from receiving inhibitory signals, and the downregulation of major histocompatibility complex class I and ligands for the activating receptor NKG2D. These changes have important implications for the ability of infected cells to escape NK cell killing. The implications of these findings for antibody-dependent NK cell activity in COVID-19 are also reviewed. Despite these advances in the understanding of the NK cell response to SARS-CoV-2, there remain critical gaps in our current understanding and a wealth of avenues for future research on this topic.
Collapse
Affiliation(s)
- Madeline J Lee
- Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
21
|
Kisiolek JN, Flores VA, Ramani A, Butler B, Haughian JM, Stewart LK. Eight Weeks of Daily Cannabidiol Supplementation Improves Sleep Quality and Immune Cell Cytotoxicity. Nutrients 2023; 15:4173. [PMID: 37836465 PMCID: PMC10574483 DOI: 10.3390/nu15194173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND The endocannabinoid system is active in nervous and immune cells and involves the expression of two cannabinoid receptor genes (CB1 and CB2), along with endogenous endocannabinoid ligands, 2-arachidonoyl glycerol (2-AG) and arachidonoyl ethanolamide (anandamide), and their synthetic enzymes. Cannabidiol (CBD) is a non-intoxicating exogenous cannabinoid agonist derived from plants that, at high doses, has received FDA approval as an anticonvulsant for epileptic seizures, and at low doses is marketed as a food-grade supplement for improved mental health, sleep quality, and immunological function. At present, the predominance of published CBD clinical research has focused on ameliorative or disease-specific intervention, with few trials investigating CBD effects in healthy populations. METHODS This clinical study aimed to investigate the effects of 8 weeks of 50 mg oral CBD on mental health, sleep quantity and quality, and immune cell function in healthy, college-aged individuals. Twenty-eight participants (average age 25.9 ± 6.1 y) were randomized to receive either daily oral capsules of 50 mg of CBD (CB, n = 14) or a calorie-matched placebo (CN, n = 14). Participants completed pre- and post-intervention assessments, including anthropometric measurements, mental health surveys, sleep analysis, and immunological function assessments. RESULTS After completing the 8-week intervention, there were no significant changes in body weight and BMI (CN: 1.09 ± 0.89%: CB: 1.41 ± 1.07%), or body fat percentage (CN: 9.01 ± 7.51%: CB: 8.57 ± 7.81%), respectively (values are % change pre to post, p > 0.05). There were also no significant differences between CB and CN groups with respect to mental health measures, sleep quantity, or circulating immunophenotype as a result of the intervention. However, the CB group experienced significant improvements in sleep quality measured objectively using a sleep questionnaire (p = 0.0023) and enhanced Natural Killer (NK) immune cell function assessed in situ (p = 0.0125). CONCLUSIONS Eight weeks of daily 50 mg CBD may improve sleep quality, and NK immunosurveillance in healthy, younger adults.
Collapse
Affiliation(s)
- Jacob N. Kisiolek
- Department of Kinesiology, Nutrition, and Dietetics, University of Northern Colorado, Greeley, CO 80639, USA; (V.A.F.); (A.R.); (B.B.)
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Victoria A. Flores
- Department of Kinesiology, Nutrition, and Dietetics, University of Northern Colorado, Greeley, CO 80639, USA; (V.A.F.); (A.R.); (B.B.)
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Arjun Ramani
- Department of Kinesiology, Nutrition, and Dietetics, University of Northern Colorado, Greeley, CO 80639, USA; (V.A.F.); (A.R.); (B.B.)
| | - Blake Butler
- Department of Kinesiology, Nutrition, and Dietetics, University of Northern Colorado, Greeley, CO 80639, USA; (V.A.F.); (A.R.); (B.B.)
| | - James M. Haughian
- Department of Biological Sciences, College of Natural and Health Sciences, University of Northern Colorado, Greeley, CO 80639, USA;
| | - Laura K. Stewart
- Department of Kinesiology, Nutrition, and Dietetics, University of Northern Colorado, Greeley, CO 80639, USA; (V.A.F.); (A.R.); (B.B.)
| |
Collapse
|
22
|
Lee G, Schauner R, Burke J, Borocz J, Vasana S, Sobieraj L, Giraudo M, Jackson Z, Ansari Q, Navas M, Sakr H, Wald D. NK cells from COVID-19 positive patients exhibit enhanced cytotoxic activity upon NKG2A and KIR2DL1 blockade. Front Immunol 2023; 14:1022890. [PMID: 37483595 PMCID: PMC10360118 DOI: 10.3389/fimmu.2023.1022890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
SARS CoV-2 has caused a global pandemic leading to significant morbidity and mortality. There is a need to elucidate and further understand the implications of COVID-19 disease on the immune system to develop improved therapeutic strategies. In particular, Natural Killer (NK) cells play an essential role in mediating the innate immune response against viral infections. To better understand the role of innate immunity in COVID-19, we characterized the phenotype of circulating NK cells from 74 COVID-19 patients and 25 controls. Through evaluating the protein expression of activating and inhibitory NK cell surface molecules using dimension reduction analysis and clustering, we identified 4 specific clusters of NK cells specific to disease state (COVID-19 positive or COVID-19 negative) and characterized COVID-19 positive NK cells as: NGK2A+KIR2DL1+NKG2C-. Utilizing blocking antibodies specific for receptors NKG2A and KIR2DL1, we found that both NKG2A and KIR2DL1 blockade markedly enhances the ability of NK cells from COVID-19 positive patients to lyse SARS-Cov-2 infected cells. Overall, this study reveals new insights into NK cell phenotypes during SARS-CoV-2 infection and suggests a therapeutic approach worthy of further investigation to enhance NK cell-mediated responses against the virus.
Collapse
Affiliation(s)
- Grace Lee
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Robert Schauner
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Juanita Burke
- Department of Pathology, Louis Stokes Cleveland Veteran Affairs (VA) Medical Center, Cleveland, OH, United States
| | - Jade Borocz
- Department of Pathology, Louis Stokes Cleveland Veteran Affairs (VA) Medical Center, Cleveland, OH, United States
| | - Smitha Vasana
- Department of Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Lukasz Sobieraj
- Midwestern University Chicago College of Osteopathic Medicine, Downers Grove, IL, United States
| | - Maria Giraudo
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Zachary Jackson
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Qasim Ansari
- Department of Pathology, Louis Stokes Cleveland Veteran Affairs (VA) Medical Center, Cleveland, OH, United States
| | - Maria Navas
- Department of Pathology, Louis Stokes Cleveland Veteran Affairs (VA) Medical Center, Cleveland, OH, United States
| | - Hany Sakr
- Department of Pathology, Louis Stokes Cleveland Veteran Affairs (VA) Medical Center, Cleveland, OH, United States
| | - David Wald
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
- Department of Pathology, Louis Stokes Cleveland Veteran Affairs (VA) Medical Center, Cleveland, OH, United States
| |
Collapse
|
23
|
Silva-Junior AL, Oliveira LDS, Belezia NCT, Tarragô AM, Costa AGD, Malheiro A. Immune Dynamics Involved in Acute and Convalescent COVID-19 Patients. IMMUNO 2023; 3:86-111. [DOI: 10.3390/immuno3010007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
COVID-19 is a viral disease that has caused millions of deaths around the world since 2020. Many strategies have been developed to manage patients in critical conditions; however, comprehension of the immune system is a key factor in viral clearance, tissue repairment, and adaptive immunity stimulus. Participation of immunity has been identified as a major factor, along with biomarkers, prediction of clinical outcomes, and antibody production after infection. Immune cells have been proposed not only as a hallmark of severity, but also as a predictor of clinical outcomes, while dynamics of inflammatory molecules can also induce worse consequences for acute patients. For convalescent patients, mild disease was related to higher antibody production, although the factors related to the specific antibodies based on a diversity of antigens were not clear. COVID-19 was explored over time; however, the study of immunological predictors of outcomes is still lacking discussion, especially in convalescent patients. Here, we propose a review using previously published studies to identify immunological markers of COVID-19 outcomes and their relation to antibody production to further contribute to the clinical and laboratorial management of patients.
Collapse
Affiliation(s)
- Alexander Leonardo Silva-Junior
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus 69050-001, AM, Brazil
- Centro Universitário do Norte (UNINORTE), Manaus 69020-031, AM, Brazil
| | - Lucas da Silva Oliveira
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus 69050-001, AM, Brazil
- Centro Universitário do Norte (UNINORTE), Manaus 69020-031, AM, Brazil
| | - Nara Caroline Toledo Belezia
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus 69050-001, AM, Brazil
- Centro Universitário do Norte (UNINORTE), Manaus 69020-031, AM, Brazil
| | - Andréa Monteiro Tarragô
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus 69050-001, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69065-001, AM, Brazil
| | - Allyson Guimarães da Costa
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus 69050-001, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69065-001, AM, Brazil
- Programa de Pós-Graduação em Imunologia, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| | - Adriana Malheiro
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus 69050-001, AM, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69065-001, AM, Brazil
- Programa de Pós-Graduação em Imunologia, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| |
Collapse
|
24
|
Islam MS, Wang Z, Abdel-Mohsen M, Chen X, Montaner LJ. Tissue injury and leukocyte changes in post-acute sequelae of SARS-CoV-2: review of 2833 post-acute patient outcomes per immune dysregulation and microbial translocation in long COVID. J Leukoc Biol 2023; 113:236-254. [PMID: 36807444 DOI: 10.1093/jleuko/qiac001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Indexed: 01/18/2023] Open
Abstract
A significant number of persons with coronavirus disease 2019 (COVID-19) experience persistent, recurrent, or new symptoms several months after the acute stage of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This phenomenon, termed post-acute sequelae of SARS-CoV-2 (PASC) or long COVID, is associated with high viral titers during acute infection, a persistently hyperactivated immune system, tissue injury by NETosis-induced micro-thrombofibrosis (NETinjury), microbial translocation, complement deposition, fibrotic macrophages, the presence of autoantibodies, and lymphopenic immune environments. Here, we review the current literature on the immunological imbalances that occur during PASC. Specifically, we focus on data supporting common immunopathogenesis and tissue injury mechanisms shared across this highly heterogenous disorder, including NETosis, coagulopathy, and fibrosis. Mechanisms include changes in leukocyte subsets/functions, fibroblast activation, cytokine imbalances, lower cortisol, autoantibodies, co-pathogen reactivation, and residual immune activation driven by persistent viral antigens and/or microbial translocation. Taken together, we develop the premise that SARS-CoV-2 infection results in PASC as a consequence of acute and/or persistent single or multiple organ injury mediated by PASC determinants to include the degree of host responses (inflammation, NETinjury), residual viral antigen (persistent antigen), and exogenous factors (microbial translocation). Determinants of PASC may be amplified by comorbidities, age, and sex.
Collapse
Affiliation(s)
- Md Sahidul Islam
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, Avenida da Universidade, Taipa 999078, University of Macau, Macau S.A.R., China
| | - Zhaoxiong Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, Avenida da Universidade, Taipa 999078, University of Macau, Macau S.A.R., China
| | - Mohamed Abdel-Mohsen
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, United States
| | - Xin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, Avenida da Universidade, Taipa 999078, University of Macau, Macau S.A.R., China.,Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa 999078, Macau S.A.R., China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa 999078, Macau S.A.R., China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Research Building N22, University of Macau, Avenida da Universidade, Taipa 999078, Macau S.A.R., China
| | - Luis J Montaner
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, United States
| |
Collapse
|
25
|
Fadlallah MM, Salman SM, Fadlallah MM, Rahal H. Hemophagocytic Syndrome and COVID-19: A Comprehensive Review. Cureus 2023; 15:e36140. [PMID: 37065291 PMCID: PMC10101193 DOI: 10.7759/cureus.36140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 03/17/2023] Open
Abstract
Hemophagocytic lymphohistiocytosis (HLH), a hyperinflammatory hyperferritinemic syndrome, is triggered by various etiologies and diseases and can lead to multiorgan dysfunction and death. There are two types of HLH: primary and secondary. Primary HLH (pHLH) is caused by a genetic mutation resulting in dysfunction in cytotoxic T lymphocytes (CTLs), natural killer (NK) cells, hyperactivated immune cells, and hypercytokinemia. In secondary HLH (sHLH), an underlying etiology is the cause of the disease. Infections, malignancy, and autoimmune diseases are well-known triggers for sHLH. Infectious triggers for sHLH are most frequently viruses, where different mechanisms, including dysregulated CTLs and NK cell activity and persistent immune system stimulation, have been reported. Similarly, in severe coronavirus disease 2019 (COVID-19) patients, a hyperinflammatory mechanism leading to hypercytokinemia and hyperferritinemia has been demonstrated. A similar dysfunction in CTLs and NK cells, persistent immune system stimulation with increased cytokines production, and severe end-organ damage have been reported. Therefore, a significant overlap is present between the clinical and laboratory features seen in COVID-19 and sHLH. However, SARS-CoV-2, similar to other viruses, can trigger sHLH. Hence, a diagnostic approach is needed in severe COVID-19 patients presenting with multiorgan failure, in whom sHLH should be considered.
Collapse
Affiliation(s)
- Mahdi M Fadlallah
- Department of Laboratory Medicine, Faculty of Medical Sciences, Lebanese University, Beirut, LBN
| | - Sarah M Salman
- Department of Laboratory Medicine, Al-Zahraa Hospital University Medical Center, Beirut, LBN
- Department of Laboratory Medicine, Faculty of Medical Sciences, Lebanese University, Beirut, LBN
| | | | - Hassan Rahal
- Department of Infectious Diseases, Bahman Hospital, Beirut, LBN
| |
Collapse
|
26
|
Saini P, Adeniji OS, Bordoloi D, Kinslow J, Martinson J, Parent DM, Hong KY, Koshy J, Kulkarni AJ, Zilberstein NF, Balk RA, Moy JN, Giron LB, Tracy RP, Keshavarzian A, Muthumani K, Landay A, Weiner DB, Abdel-Mohsen M. Siglec-9 Restrains Antibody-Dependent Natural Killer Cell Cytotoxicity against SARS-CoV-2. mBio 2023; 14:e0339322. [PMID: 36728420 PMCID: PMC9973332 DOI: 10.1128/mbio.03393-22] [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: 12/03/2022] [Accepted: 12/23/2022] [Indexed: 02/03/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection alters the immunological profiles of natural killer (NK) cells. However, whether NK antiviral functions are impaired during severe coronavirus disease 2019 (COVID-19) and what host factors modulate these functions remain unclear. We found that NK cells from hospitalized COVID-19 patients degranulate less against SARS-CoV-2 antigen-expressing cells (in direct cytolytic and antibody-dependent cell cytotoxicity [ADCC] assays) than NK cells from mild COVID-19 patients or negative controls. The lower NK degranulation was associated with higher plasma levels of SARS-CoV-2 nucleocapsid antigen. Phenotypic and functional analyses showed that NK cells expressing the glyco-immune checkpoint Siglec-9 elicited higher ADCC than Siglec-9- NK cells. Consistently, Siglec-9+ NK cells exhibit an activated and mature phenotype with higher expression of CD16 (FcγRIII; mediator of ADCC), CD57 (maturation marker), and NKG2C (activating receptor), along with lower expression of the inhibitory receptor NKG2A, than Siglec-9- CD56dim NK cells. These data are consistent with the concept that the NK cell subpopulation expressing Siglec-9 is highly activated and cytotoxic. However, the Siglec-9 molecule itself is an inhibitory receptor that restrains NK cytotoxicity during cancer and other viral infections. Indeed, blocking Siglec-9 significantly enhanced the ADCC-mediated NK degranulation and lysis of SARS-CoV-2-antigen-positive target cells. These data support a model in which the Siglec-9+ CD56dim NK subpopulation is cytotoxic even while it is restrained by the inhibitory effects of Siglec-9. Alleviating the Siglec-9-mediated restriction on NK cytotoxicity may further improve NK immune surveillance and presents an opportunity to develop novel immunotherapeutic tools against SARS-CoV-2 infected cells. IMPORTANCE One mechanism that cancer cells use to evade natural killer cell immune surveillance is by expressing high levels of sialoglycans, which bind to Siglec-9, a glyco-immune checkpoint molecule on NK cells. This binding inhibits NK cell cytotoxicity. Several viruses, such as hepatitis B virus (HBV) and HIV, also use a similar mechanism to evade NK surveillance. We found that NK cells from SARS-CoV-2-hospitalized patients are less able to function against cells expressing SARS-CoV-2 Spike protein than NK cells from SARS-CoV-2 mild patients or uninfected controls. We also found that the cytotoxicity of the Siglec-9+ NK subpopulation is indeed restrained by the inhibitory nature of the Siglec-9 molecule and that blocking Siglec-9 can enhance the ability of NK cells to target cells expressing SARS-CoV-2 antigens. Our results suggest that a targetable glyco-immune checkpoint mechanism, Siglec-9/sialoglycan interaction, may contribute to the ability of SARS-CoV-2 to evade NK immune surveillance.
Collapse
Affiliation(s)
- Pratima Saini
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | - Kai Ying Hong
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Jane Koshy
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | - Kar Muthumani
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | | | | |
Collapse
|
27
|
Ruffieux H, Hanson AL, Lodge S, Lawler NG, Whiley L, Gray N, Nolan TH, Bergamaschi L, Mescia F, Turner L, de Sa A, Pelly VS, Kotagiri P, Kingston N, Bradley JR, Holmes E, Wist J, Nicholson JK, Lyons PA, Smith KGC, Richardson S, Bantug GR, Hess C. A patient-centric modeling framework captures recovery from SARS-CoV-2 infection. Nat Immunol 2023; 24:349-358. [PMID: 36717723 PMCID: PMC9892000 DOI: 10.1038/s41590-022-01380-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 11/03/2022] [Indexed: 02/01/2023]
Abstract
The biology driving individual patient responses to severe acute respiratory syndrome coronavirus 2 infection remains ill understood. Here, we developed a patient-centric framework leveraging detailed longitudinal phenotyping data and covering a year after disease onset, from 215 infected individuals with differing disease severities. Our analyses revealed distinct 'systemic recovery' profiles, with specific progression and resolution of the inflammatory, immune cell, metabolic and clinical responses. In particular, we found a strong inter-patient and intra-patient temporal covariation of innate immune cell numbers, kynurenine metabolites and lipid metabolites, which highlighted candidate immunologic and metabolic pathways influencing the restoration of homeostasis, the risk of death and that of long COVID. Based on these data, we identified a composite signature predictive of systemic recovery, using a joint model on cellular and molecular parameters measured soon after disease onset. New predictions can be generated using the online tool http://shiny.mrc-bsu.cam.ac.uk/apps/covid-19-systemic-recovery-prediction-app , designed to test our findings prospectively.
Collapse
Affiliation(s)
- Hélène Ruffieux
- MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| | - Aimee L Hanson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Samantha Lodge
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Nathan G Lawler
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Luke Whiley
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| | - Nicola Gray
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Tui H Nolan
- MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Federica Mescia
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Lorinda Turner
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Aloka de Sa
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Victoria S Pelly
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Prasanti Kotagiri
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Nathalie Kingston
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - John R Bradley
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Elaine Holmes
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Julien Wist
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Chemistry Department, Universidad del Valle, Cali, Colombia
| | - Jeremy K Nicholson
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Center for Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
- Institute of Global Health Innovation, Imperial College London, London, UK
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Sylvia Richardson
- MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Glenn R Bantug
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Botnar Research Centre for Child Health (BRCCH) University Basel & ETH Zurich, Basel, Switzerland
| | - Christoph Hess
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland.
- Botnar Research Centre for Child Health (BRCCH) University Basel & ETH Zurich, Basel, Switzerland.
| |
Collapse
|
28
|
Nasrollahi H, Talepoor AG, Saleh Z, Eshkevar Vakili M, Heydarinezhad P, Karami N, Noroozi M, Meri S, Kalantar K. Immune responses in mildly versus critically ill COVID-19 patients. Front Immunol 2023; 14:1077236. [PMID: 36793739 PMCID: PMC9923185 DOI: 10.3389/fimmu.2023.1077236] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
The current coronavirus pandemic (COVID-19), caused by SARS-CoV-2, has had devastating effects on the global health and economic system. The cellular and molecular mediators of both the innate and adaptive immune systems are critical in controlling SARS-CoV-2 infections. However, dysregulated inflammatory responses and imbalanced adaptive immunity may contribute to tissue destruction and pathogenesis of the disease. Important mechanisms in severe forms of COVID-19 include overproduction of inflammatory cytokines, impairment of type I IFN response, overactivation of neutrophils and macrophages, decreased frequencies of DC cells, NK cells and ILCs, complement activation, lymphopenia, Th1 and Treg hypoactivation, Th2 and Th17 hyperactivation, as well as decreased clonal diversity and dysregulated B lymphocyte function. Given the relationship between disease severity and an imbalanced immune system, scientists have been led to manipulate the immune system as a therapeutic approach. For example, anti-cytokine, cell, and IVIG therapies have received attention in the treatment of severe COVID-19. In this review, the role of immunity in the development and progression of COVID-19 is discussed, focusing on molecular and cellular aspects of the immune system in mild vs. severe forms of the disease. Moreover, some immune- based therapeutic approaches to COVID-19 are being investigated. Understanding key processes involved in the disease progression is critical in developing therapeutic agents and optimizing related strategies.
Collapse
Affiliation(s)
- Hamid Nasrollahi
- Radio-Oncology Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atefe Ghamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Saleh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Eshkevar Vakili
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paria Heydarinezhad
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Karami
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Noroozi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seppo Meri
- Department of Bacteriology and Immunology, University of Helsinki and Diagnostic Center of the Helsinki University Hospital, Helsinki, Finland
| | - Kurosh Kalantar
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
29
|
Lee MJ, Leong MW, Rustagi A, Beck A, Zeng L, Holmes S, Qi LS, Blish CA. SARS-CoV-2 escapes direct NK cell killing through Nsp1-mediated downregulation of ligands for NKG2D. Cell Rep 2022; 41:111892. [PMID: 36543165 PMCID: PMC9742201 DOI: 10.1016/j.celrep.2022.111892] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/09/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells are cytotoxic effector cells that target and lyse virally infected cells; many viruses therefore encode mechanisms to escape such NK cell killing. Here, we interrogate the ability of SARS-CoV-2 to modulate NK cell recognition and lysis of infected cells. We find that NK cells exhibit poor cytotoxic responses against SARS-CoV-2-infected targets, preferentially killing uninfected bystander cells. We demonstrate that this escape is driven by downregulation of ligands for the activating receptor NKG2D (NKG2D-L). Indeed, early in viral infection, prior to NKG2D-L downregulation, NK cells are able to target and kill infected cells; however, this ability is lost as viral proteins are expressed. Finally, we find that SARS-CoV-2 non-structural protein 1 (Nsp1) mediates downregulation of NKG2D-L and that Nsp1 alone is sufficient to confer resistance to NK cell killing. Collectively, our work demonstrates that SARS-CoV-2 evades direct NK cell cytotoxicity and describes a mechanism by which this occurs.
Collapse
Affiliation(s)
- Madeline J Lee
- Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle W Leong
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arjun Rustagi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aimee Beck
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leiping Zeng
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Susan Holmes
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Sarafan Chem-H, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94157, USA
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94157, USA; Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
30
|
Ruenjaiman V, Sodsai P, Kueanjinda P, Bunrasmee W, Klinchanhom S, Reantragoon R, Tunvirachaisakul C, Manothummetha K, Mejun N, Liengswangwong K, Torvorapanit P, Paitoonpong L, Putcharoen O, Palaga T, Hirankarn N, Mittrakulkij C, Chiewbangyang F, Kaewsrihawong J, Sanpakit J, Kulkiatprasert K, Munkong K, Keawthawon N, Wattanakul N, Limchanachon N, Roopsuwankun N, Chaosuwannakij N, Larpanekanan P, Pitakkitnukun P, Homswad P, Ratanapraisorn S, Atchariyapakorn S, Vongphanich S, Jessadapornchai S, Avihingsanon T, Piyasathapornpong T. Impact of SARS-CoV-2 infection on the profiles and responses of innate immune cells after recovery. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022; 55:993-1004. [PMID: 36220753 PMCID: PMC9519362 DOI: 10.1016/j.jmii.2022.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/30/2022] [Accepted: 09/24/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUNDS SARS-CoV-2 infection results in a broad spectrum of clinical outcomes, ranging from asymptomatic to severe symptoms and death. Most COVID-19 pathogenesis is associated with hyperinflammatory conditions driven primarily by myeloid cell lineages. The long-term effects of SARS-CoV-2 infection post recovery include various symptoms. METHODS We performed a longitudinal study of the innate immune profiles 1 and 3 months after recovery in the Thai cohort by comparing patients with mild, moderate, and severe clinical symptoms using peripheral blood mononuclear cells (n = 62). RESULTS Significant increases in the frequencies of monocytes compared to controls and NK cells compared to mild and moderate patients were observed in severe patients 1-3 months post recovery. Increased polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were observed in all recovered patients, even after 3 months. Increased IL-6 and TNFα levels in monocytes were observed 1 month after recovery in response to lipopolysaccharide (LPS) stimulation, while decreased CD86 and HLA-DR levels were observed regardless of stimulation. A multiplex analysis of serum cytokines performed at 1 month revealed that most innate cytokines, except for TNFα, IL4/IL-13 (Th2) and IFNγ (Th1), were elevated in recovered patients in a severity-dependent manner. Finally, the myelopoiesis cytokines G-CSF and GM-CSF were higher in all patient groups. Increased monocytes and IL-6- and TNFα-producing cells were significantly associated with long COVID-19 symptoms. CONCLUSIONS These results reveal that COVID-19 infection influences the frequencies and functions of innate immune cells for up to 3 months after recovery, which may potentially lead to some of the long COVID symptoms.
Collapse
Affiliation(s)
- Vichaya Ruenjaiman
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pimpayao Sodsai
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | - Patipark Kueanjinda
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | - Worawan Bunrasmee
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | - Siriwan Klinchanhom
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | - Rangsima Reantragoon
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | | | - Kasama Manothummetha
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nuthchaya Mejun
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kaewkwan Liengswangwong
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pattama Torvorapanit
- Thai Red Cross Emerging Infectious Diseases Clinical Centre, King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand,Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Leilani Paitoonpong
- Thai Red Cross Emerging Infectious Diseases Clinical Centre, King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand,Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Opass Putcharoen
- Thai Red Cross Emerging Infectious Diseases Clinical Centre, King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand,Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Tanapat Palaga
- Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand,Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand,Corresponding author. Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nattiya Hirankarn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand,Center of Excellence in Immunology and Immune-Mediated Diseases Chulalongkorn University, Bangkok, 10330, Thailand
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Fiorino S, Carusi A, Hong W, Cernuschi P, Gallo CG, Ferrara E, Maloberti T, Visani M, Lari F, de Biase D, Zippi M. SARS-CoV-2 vaccines: What we know, what we can do to improve them and what we could learn from other well-known viruses. AIMS Microbiol 2022; 8:422-453. [PMID: 36694588 PMCID: PMC9834075 DOI: 10.3934/microbiol.2022029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/24/2022] [Accepted: 11/06/2022] [Indexed: 11/18/2022] Open
Abstract
In recent weeks, the rate of SARS-CoV-2 infections has been progressively increasing all over the globe, even in countries where vaccination programs have been strongly implemented. In these regions in 2021, a reduction in the number of hospitalizations and deaths compared to 2020 was observed. This decrease is certainly associated with the introduction of vaccination measures. The process of the development of effective vaccines represents an important challenge. Overall, the breakthrough infections occurring in vaccinated subjects are in most cases less severe than those observed in unvaccinated individuals. This review examines the factors affecting the immunogenicity of vaccines against SARS-CoV-2 and the possible role of nutrients in modulating the response of distinct immune cells to the vaccination.
Collapse
Affiliation(s)
- Sirio Fiorino
- Internal Medicine Unit, Budrio Hospital, Budrio (Bologna), Azienda USL, Bologna, Italy
| | - Andrea Carusi
- Internal Medicine Unit, Budrio Hospital, Budrio (Bologna), Azienda USL, Bologna, Italy
| | - Wandong Hong
- Department of Gastroenterology and Hepatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang, The People's Republic of China
| | - Paolo Cernuschi
- Internal Medicine Unit, Quisana Private Hospital, Ferrara, Italy
| | | | | | - Thais Maloberti
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna-Molecular Diagnostic Unit, Azienda USL di Bologna, Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Michela Visani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna-Molecular Diagnostic Unit, Azienda USL di Bologna, Bologna, Italy
| | - Federico Lari
- Internal Medicine Unit, Budrio Hospital, Budrio (Bologna), Azienda USL, Bologna, Italy
| | - Dario de Biase
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Maddalena Zippi
- Unit of Gastroenterology and Digestive Endoscopy, Sandro Pertini Hospital, Rome, Italy
| |
Collapse
|
32
|
Jasim SA, Mahdi RS, Bokov DO, Najm MAA, Sobirova GN, Bafoyeva ZO, Taifi A, Alkadir OKA, Mustafa YF, Mirzaei R, Karampoor S. The deciphering of the immune cells and marker signature in COVID-19 pathogenesis: An update. J Med Virol 2022; 94:5128-5148. [PMID: 35835586 PMCID: PMC9350195 DOI: 10.1002/jmv.28000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/28/2022] [Accepted: 07/13/2022] [Indexed: 12/15/2022]
Abstract
The precise interaction between the immune system and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is critical in deciphering the pathogenesis of coronavirus disease 2019 (COVID-19) and is also vital for developing novel therapeutic tools, including monoclonal antibodies, antivirals drugs, and vaccines. Viral infections need innate and adaptive immune reactions since the various immune components, such as neutrophils, macrophages, CD4+ T, CD8+ T, and B lymphocytes, play different roles in various infections. Consequently, the characterization of innate and adaptive immune reactions toward SARS-CoV-2 is crucial for defining the pathogenicity of COVID-19. In this study, we explain what is currently understood concerning the conventional immune reactions to SARS-CoV-2 infection to shed light on the protective and pathogenic role of immune response in this case. Also, in particular, we investigate the in-depth roles of other immune mediators, including neutrophil elastase, serum amyloid A, and syndecan, in the immunopathogenesis of COVID-19.
Collapse
Affiliation(s)
| | - Roaa Salih Mahdi
- Department of Pathology, College of MedicineUniversity of BabylonHillaIraq
| | - Dmitry Olegovich Bokov
- Institute of PharmacySechenov First Moscow State Medical UniversityMoscowRussian Federation
- Laboratory of Food ChemistryFederal Research Center of Nutrition, Biotechnology and Food SafetyMoscowRussian Federation
| | - Mazin A. A. Najm
- Pharmaceutical Chemistry Department, College of PharmacyAl‐Ayen UniversityThi‐QarIraq
| | - Guzal N. Sobirova
- Department of Rehabilitation, Folk Medicine and Physical EducationTashkent Medical AcademyTashkentUzbekistan
| | - Zarnigor O. Bafoyeva
- Department of Rehabilitation, Folk Medicine and Physical EducationTashkent Medical AcademyTashkentUzbekistan
| | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of PharmacyUniversity of MosulMosulIraq
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research CenterPasteur Institute of IranTehranIran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research CenterIran University of Medical SciencesTehranIran
| |
Collapse
|
33
|
Mitsuyama Y, Yamakawa K, Kayano K, Maruyama M, Umemura Y, Wada T, Fujimi S. Residual persistence of cytotoxicity lymphocytes and regulatory T cells in patients with severe coronavirus disease 2019 over a 1-year recovery process. Acute Med Surg 2022; 9:e803. [PMID: 36311179 PMCID: PMC9597380 DOI: 10.1002/ams2.803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022] Open
Abstract
Aim To clarify the immune cellular changes in critically ill patients recovering from coronavirus disease 2019 (COVID‐19). Methods The immune response of peripheral blood mononuclear cells from patients with severe COVID‐19 in different stages of recovery (3, 6, and 12 months from hospitalization) was evaluated by single‐cell mass cytometry. Immunological changes in patients were compared with those in age‐matched healthy donors. Results Three patients with severe COVID‐19 were compared with four healthy donors. In the patients, there was an increase in the cell density of CD4‐ and CD8‐positive T lymphocytes, and B cells, over the course of the recovery period. CD4‐ and CD8‐positive T lymphocytes expressing T‐bet and granzyme B (Gzm B) in patients were abundant during all recovery periods. The level of regulatory T cells remained high throughout the year. The levels of natural killer (NK) cells in patients were higher than in those in the healthy donors, and the frequency of CD16+ NK cells expressing Gzm B increased throughout the year. Conclusion Patients recovering from severe COVID‐19 showed persistence of cytotoxic lymphocytes, NK cells, and regulatory T cells throughout the posthospitalization year of recovery.
Collapse
Affiliation(s)
- Yumi Mitsuyama
- Division of Trauma and Surgical Critical CareOsaka General Medical CenterOsakaJapan,Department of Traumatology and Acute Critical MedicineOsaka University Graduate School of MedicineOsakaJapan
| | - Kazuma Yamakawa
- Department of Emergency and Critical Care MedicineOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Katsuhide Kayano
- Department of Emergency and Critical Care MedicineOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Miho Maruyama
- Division of Trauma and Surgical Critical CareOsaka General Medical CenterOsakaJapan
| | - Yutaka Umemura
- Division of Trauma and Surgical Critical CareOsaka General Medical CenterOsakaJapan
| | - Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care MedicineHokkaido University Faculty of MedicineSapporoJapan
| | - Satoshi Fujimi
- Division of Trauma and Surgical Critical CareOsaka General Medical CenterOsakaJapan
| |
Collapse
|
34
|
Malengier-Devlies B, Filtjens J, Ahmadzadeh K, Boeckx B, Vandenhaute J, De Visscher A, Bernaerts E, Mitera T, Jacobs C, Vanderbeke L, Van Mol P, Van Herck Y, Hermans G, Meersseman P, Wilmer A, Gouwy M, Garg AD, Humblet-Baron S, De Smet F, Martinod K, Wauters E, Proost P, Wouters C, Leclercq G, Lambrechts D, Wauters J, Matthys P. Severe COVID-19 patients display hyper-activated NK cells and NK cell-platelet aggregates. Front Immunol 2022; 13:861251. [PMID: 36275702 PMCID: PMC9581751 DOI: 10.3389/fimmu.2022.861251] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 08/15/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is characterised by a broad spectrum of clinical and pathological features. Natural killer (NK) cells play an important role in innate immune responses to viral infections. Here, we analysed the phenotype and activity of NK cells in the blood of COVID-19 patients using flow cytometry, single-cell RNA-sequencing (scRNA-seq), and a cytotoxic killing assay. In the plasma of patients, we quantified the main cytokines and chemokines. Our cohort comprises COVID-19 patients hospitalised in a low-care ward unit (WARD), patients with severe COVID-19 disease symptoms hospitalised in intensive care units (ICU), and post-COVID-19 patients, who were discharged from hospital six weeks earlier. NK cells from hospitalised COVID-19 patients displayed an activated phenotype with substantial differences between WARD and ICU patients and the timing when samples were taken post-onset of symptoms. While NK cells from COVID-19 patients at an early stage of infection showed increased expression of the cytotoxic molecules perforin and granzyme A and B, NK cells from patients at later stages of COVID-19 presented enhanced levels of IFN-γ and TNF-α which were measured ex vivo in the absence of usual in vitro stimulation. These activated NK cells were phenotyped as CD49a+CD69a+CD107a+ cells, and their emergence in patients correlated to the number of neutrophils, and plasma IL-15, a key cytokine in NK cell activation. Despite lower amounts of cytotoxic molecules in NK cells of patients with severe symptoms, majority of COVID-19 patients displayed a normal cytotoxic killing of Raji tumour target cells. In vitro stimulation of patients blood cells by IL-12+IL-18 revealed a defective IFN-γ production in NK cells of ICU patients only, indicative of an exhausted phenotype. ScRNA-seq revealed, predominantly in patients with severe COVID-19 disease symptoms, the emergence of an NK cell subset with a platelet gene signature that we identified by flow and imaging cytometry as aggregates of NK cells with CD42a+CD62P+ activated platelets. Post-COVID-19 patients show slow recovery of NK cell frequencies and phenotype. Our study points to substantial changes in NK cell phenotype during COVID-19 disease and forms a basis to explore the contribution of platelet-NK cell aggregates to antiviral immunity against SARS-CoV-2 and disease pathology.
Collapse
Affiliation(s)
- Bert Malengier-Devlies
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Jessica Filtjens
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Kourosh Ahmadzadeh
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Bram Boeckx
- Laboratory of Translational Genetics, Department of Human Genetics, VIB-KU Leuven, Leuven, Belgium
| | - Jessica Vandenhaute
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Amber De Visscher
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Eline Bernaerts
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Tania Mitera
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Cato Jacobs
- Laboratory for Clinical Infectious and Inflammatory Disorders, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Lore Vanderbeke
- Laboratory of Clinical Bacteriology and Mycology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Pierre Van Mol
- Laboratory of Translational Genetics, Department of Human Genetics, VIB-KU Leuven, Leuven, Belgium
| | - Yannick Van Herck
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Greet Hermans
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Philippe Meersseman
- Laboratory for Clinical Infectious and Inflammatory Disorders, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Alexander Wilmer
- Laboratory for Clinical Infectious and Inflammatory Disorders, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Abhishek D. Garg
- Laboratory for Cell Stress & Immunity (CSI), Department of Cellular and Molecular Medicine (CMM), KU Leuven, Leuven, Belgium
| | - Stephanie Humblet-Baron
- Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Els Wauters
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Carine Wouters
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Georges Leclercq
- Laboratory of Experimental Immunology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Diether Lambrechts
- Laboratory of Translational Genetics, Department of Human Genetics, VIB-KU Leuven, Leuven, Belgium
| | - Joost Wauters
- Laboratory for Clinical Infectious and Inflammatory Disorders, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
35
|
Hejazian SS, Hejazian SM, Farnood F, Abedi Azar S. Dysregulation of immunity in COVID-19 and SLE. Inflammopharmacology 2022; 30:1517-1531. [PMID: 36028612 PMCID: PMC9417079 DOI: 10.1007/s10787-022-01047-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/30/2022] [Indexed: 12/15/2022]
Abstract
The immune response plays a crucial role in preventing diseases, such as infections. There are two types of immune responses, specific and innate immunity, each of which consists of two components: cellular immunity and humoral immunity. Dysfunction in any immune system component increases the risk of developing certain diseases. Systemic lupus erythematosus (SLE), an autoimmune disease in the human body, develops an immune response against its own components. In these patients, due to underlying immune system disorders and receipt of immunosuppressive drugs, the susceptibility to infections is higher than in the general population and is the single largest cause of mortality in this group. COVID-19 infection, which first appeared in late 2019, has caused several concerns in patients with SLE. However, there is no strong proof of additional risk of developing COVID-19 in patients with SLE, and in some cases, studies have shown less severity of the disease in these individuals. This review paper discusses the immune disorders in SLE and COVID-19.
Collapse
Affiliation(s)
- Seyyed Sina Hejazian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farahnoosh Farnood
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sima Abedi Azar
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
36
|
Radandish M, Esmaeil N, Khorvash F, Andalib A. Diagnostic Value of Natural Killer Cells, CD56+ CD16+ Natural Killer Cells, NLRP3, and Lactate Dehydrogenase in Severe/Critical COVID-19: A Prospective Longitudinal Study According to the Severe/Critical COVID-19 Definitions. Viral Immunol 2022; 35:616-628. [PMID: 36099205 DOI: 10.1089/vim.2022.0060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Innate immunity, as the first line of defense of our immune system, plays a crucial role in defending against SARS-CoV-2 infection and also its immunopathogenesis. We aim to investigate the immune status of natural killer (NK) cells, natural killer T (NKT) cells, and NLRP3 gene expression in COVID-19 patient blood samples. The immunophenotype of NK cell subsets and NKT cells was detected by flow cytometry and the expression of NLRP3 gene assessed by reverse transcriptase real-time polymerase chain reaction in 44 COVID-19 patients and 20 healthy individuals. The percentage of most of NK cell subpopulation and NKT cells was significantly decreased in COVID-19 patients. The percentage of CD56dim CD16- NK cell subsets, and NLRP3 gene expression increased. The percentage of total NK cells, CD56+ CD16+ NK cells, and NLRP3 gene expression had acceptable sensitivity and specificity for assisting diagnosis of severe/critical COVID-19. O2 saturation% and lactate dehydrogenase levels showed valuable diagnostic value to identify critical cases. The declined NK and NKT cells in COVID-19 patients and enhanced NLRP3 gene expression were associated with disease severity. Total NK cells, CD56+ CD16+ NK cells, and NLRP3 gene expression might be used as meaningful indicators for assisting diagnosis of severe/critical COVID-19.
Collapse
Affiliation(s)
- Maedeh Radandish
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farzin Khorvash
- Department of Infectious Diseases, Faculty of Medicine, Nosocomial Infections Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Andalib
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
37
|
Safety and Efficacy of Allogeneic Natural Killer Cells in Combination with Pembrolizumab in Patients with Chemotherapy-Refractory Biliary Tract Cancer: A Multicenter Open-Label Phase 1/2a Trial. Cancers (Basel) 2022; 14:cancers14174229. [PMID: 36077766 PMCID: PMC9454779 DOI: 10.3390/cancers14174229] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: This study investigated the administration of combination therapy, allogeneic natural killer (NK) cells and pembrolizumab in the treatment of advanced biliary tract cancer to determine the safety and tolerability (phase 1) and the efficacy and safety (phase 2a). Methods: Forty patients (phase 1, n = 6; phase 2a, n = 34) were enrolled between December 2019 and June 2021. The patients received highly activated allogeneic NK cells (“SMT-NK”) on weeks 1 and 2 and pembrolizumab on week 1. This 3-week schedule (one cycle) was repeated until confirmed disease progression, intolerable adverse events (AEs), patient withdrawal, or finishing the maximum treatment schedule. The tumor response was evaluated after every three cycles. Results: In phase 1, four patients (66.7%) experienced seven AEs, but no severe AE was observed. In phase 2a, 126 AEs occurred in 29 patients (85.3%). Severe AEs (≥grade 3) were reported in 16 patients (47.1%). The overall response rate (ORR) was 17.4% in the full analysis set and 50.0% in the per-protocol set. Conclusions: SMT-NKs plus pembrolizumab resulted in no severe AEs directly related to the drug combination. The combination therapy also exerted antitumor activity with improved efficacy compared to the recent monotherapy with pembrolizumab in patients with advanced biliary tract cancer.
Collapse
|
38
|
Di Vito C, Calcaterra F, Coianiz N, Terzoli S, Voza A, Mikulak J, Della Bella S, Mavilio D. Natural Killer Cells in SARS-CoV-2 Infection: Pathophysiology and Therapeutic Implications. Front Immunol 2022; 13:888248. [PMID: 35844604 PMCID: PMC9279859 DOI: 10.3389/fimmu.2022.888248] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/01/2022] [Indexed: 12/23/2022] Open
Abstract
Natural Killer (NK) cells are lymphocytes of the innate immunity that play a crucial role in the control of viral infections in the absence of a prior antigen sensitization. Indeed, they display rapid effector functions against target cells with the capability of direct cell killing and antibody-dependent cell-mediated cytotoxicity. Furthermore, NK cells are endowed with immune-modulatory functions innate and adaptive immune responses via the secretion of chemokines/cytokines and by undertaking synergic crosstalks with other innate immune cells, including monocyte/macrophages, dendritic cells and neutrophils. Recently, the Coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread globally. Although the specific role of NK cells in COVID-19 pathophysiology still need to be explored, mounting evidence indicates that NK cell tissue distribution and effector functions could be affected by SARS-CoV-2 infection and that a prompt NK cell response could determine a good clinical outcome in COVID-19 patients. In this review, we give a comprehensive overview of how SARS-CoV-2 infection interferes with NK cell antiviral effectiveness and their crosstalk with other innate immune cells. We also provide a detailed characterization of the specific NK cell subsets in relation to COVID-19 patient severity generated from publicly available single cell RNA sequencing datasets. Finally, we summarize the possible NK cell-based therapeutic approaches against SARS-CoV-2 infection and the ongoing clinical trials updated at the time of submission of this review. We will also discuss how a deep understanding of NK cell responses could open new possibilities for the treatment and prevention of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Clara Di Vito
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- *Correspondence: Domenico Mavilio, ; Clara Di Vito,
| | - Francesca Calcaterra
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra) , University of Milan, Milan, Italy
| | - Nicolò Coianiz
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Sara Terzoli
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Antonio Voza
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Emergency Medicine Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Joanna Mikulak
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Silvia Della Bella
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra) , University of Milan, Milan, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra) , University of Milan, Milan, Italy
- *Correspondence: Domenico Mavilio, ; Clara Di Vito,
| |
Collapse
|
39
|
Wang M, Wu D, Liu CH, Li Y, Hu J, Wang W, Jiang W, Zhang Q, Huang Z, Bai L, Tang H. Predicting progression to severe COVID-19 using the PAINT score. BMC Infect Dis 2022; 22:498. [PMID: 35619076 PMCID: PMC9134988 DOI: 10.1186/s12879-022-07466-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 05/10/2022] [Indexed: 02/08/2023] Open
Abstract
Objectives One of the major challenges in treating patients with coronavirus disease 2019 (COVID-19) is predicting the severity of disease. We aimed to develop a new score for predicting progression from mild/moderate to severe COVID-19. Methods A total of 239 hospitalized patients with COVID-19 from two medical centers in China between February 6 and April 6, 2020 were retrospectively included. The prognostic abilities of variables, including clinical data and laboratory findings from the electronic medical records of each hospital, were analysed using the Cox proportional hazards model and Kaplan–Meier methods. A prognostic score was developed to predict progression from mild/moderate to severe COVID-19. Results Among the 239 patients, 216 (90.38%) patients had mild/moderate disease, and 23 (9.62%) progressed to severe disease. After adjusting for multiple confounding factors, pulmonary disease, age > 75, IgM, CD16+/CD56+ NK cells and aspartate aminotransferase were independent predictors of progression to severe COVID-19. Based on these five factors, a new predictive score (the ‘PAINT score’) was established and showed a high predictive value (C-index = 0.91, 0.902 ± 0.021, p < 0.001). The PAINT score was validated using a nomogram, bootstrap analysis, calibration curves, decision curves and clinical impact curves, all of which confirmed its high predictive value. Conclusions The PAINT score for progression from mild/moderate to severe COVID-19 may be helpful in identifying patients at high risk of progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07466-4.
Collapse
Affiliation(s)
- Ming Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, Sichuan Province, 610041, People's Republic of China.,COVID-19 Medical Team (Hubei) of West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, Sichuan Province, 610041, People's Republic of China.,COVID-19 Medical Team (Hubei) of West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Chang-Hai Liu
- Center of Infectious Diseases, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, Sichuan Province, 610041, People's Republic of China
| | - Yan Li
- The People's Hospital of Qianxi, Qianxi, 551500, People's Republic of China
| | - Jianghong Hu
- The People's Hospital of Duyun, Duyun, 558000, People's Republic of China
| | - Wei Wang
- COVID-19 Medical Team (Hubei) of West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.,Emergency Department, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Wei Jiang
- Center of Infectious Diseases, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, Sichuan Province, 610041, People's Republic of China
| | - Qifan Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Zhixin Huang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, Sichuan Province, 610041, People's Republic of China. .,COVID-19 Medical Team (Hubei) of West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Hong Tang
- COVID-19 Medical Team (Hubei) of West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
40
|
Soleimanian S, Alyasin S, Sepahi N, Ghahramani Z, Kanannejad Z, Yaghobi R, Karimi MH. An Update on Protective Effectiveness of Immune Responses After Recovery From COVID-19. Front Immunol 2022; 13:884879. [PMID: 35669767 PMCID: PMC9163347 DOI: 10.3389/fimmu.2022.884879] [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: 02/27/2022] [Accepted: 04/20/2022] [Indexed: 12/22/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exhibits variable immunity responses among hosts based on symptom severity. Whether immunity in recovered individuals is effective for avoiding reinfection is poorly understood. Determination of immune memory status against SARS-CoV-2 helps identify reinfection risk and vaccine efficacy. Hence, after recovery from COVID-19, evaluation of protective effectiveness and durable immunity of prior disease could be significant. Recent reports described the dynamics of SARS-CoV-2 -specific humoral and cellular responses for more than six months in convalescent SARS-CoV-2 individuals. Given the current evidence, NK cell subpopulations, especially the memory-like NK cell subset, indicate a significant role in determining COVID-19 severity. Still, the information on the long-term NK cell immunity conferred by SARS-CoV-2 infection is scant. The evidence from vaccine clinical trials and observational studies indicates that hybrid natural/vaccine immunity to SARS-CoV-2 seems to be notably potent protection. We suggested the combination of plasma therapy from recovered donors and vaccination could be effective. This focused review aims to update the current information regarding immune correlates of COVID-19 recovery to understand better the probability of reinfection in COVID-19 infected cases that may serve as guides for ongoing vaccine strategy improvement.
Collapse
Affiliation(s)
- Saeede Soleimanian
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheila Alyasin
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Allergy and Clinical Immunology, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Sepahi
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Ghahramani
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Kanannejad
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ramin Yaghobi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
41
|
Wang Y, Wu M, Li Y, Yuen HH, He ML. The effects of SARS-CoV-2 infection on modulating innate immunity and strategies of combating inflammatory response for COVID-19 therapy. J Biomed Sci 2022; 29:27. [PMID: 35505345 PMCID: PMC9063252 DOI: 10.1186/s12929-022-00811-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/27/2022] [Indexed: 12/15/2022] Open
Abstract
The global pandemic of COVID-19 has caused huge causality and unquantifiable loss of social wealth. The innate immune response is the first line of defense against SARS-CoV-2 infection. However, strong inflammatory response associated with dysregulation of innate immunity causes severe acute respiratory syndrome (SARS) and death. In this review, we update the current knowledge on how SARS-CoV-2 modulates the host innate immune response for its evasion from host defense and its corresponding pathogenesis caused by cytokine storm. We emphasize Type I interferon response and the strategies of evading innate immune defense used by SARS-CoV-2. We also extensively discuss the cells and their function involved in the innate immune response and inflammatory response, as well as the promises and challenges of drugs targeting excessive inflammation for antiviral treatment. This review would help us to figure out the current challenge questions of SARS-CoV-2 infection on innate immunity and directions for future studies.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Mandi Wu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Yichen Li
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Ho Him Yuen
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China. .,CityU Shenzhen Research Institute, Nanshan, Shenzhen, China.
| |
Collapse
|
42
|
Exploring the Utility of NK Cells in COVID-19. Biomedicines 2022; 10:biomedicines10051002. [PMID: 35625739 PMCID: PMC9138257 DOI: 10.3390/biomedicines10051002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) can manifest as acute respiratory distress syndrome and is associated with substantial morbidity and mortality. Extensive data now indicate that immune responses to SARS-CoV-2 infection determine the COVID-19 disease course. A wide range of immunomodulatory agents have been tested for the treatment of COVID-19. Natural killer (NK) cells play an important role in antiviral innate immunity, and anti-SARS-CoV-2 activity and antifibrotic activity are particularly critical for COVID-19 control. Notably, SARS-CoV-2 clearance rate, antibody response, and disease progression in COVID-19 correlate with NK cell status, and NK cell dysfunction is linked with increased SARS-CoV-2 susceptibility. Thus, NK cells function as the key element in the switch from effective to harmful immune responses in COVID-19. However, dysregulation of NK cells has been observed in COVID-19 patients, exhibiting depletion and dysfunction, which correlate with COVID-19 severity; this dysregulation perhaps contributes to disease progression. Given these findings, NK-cell-based therapies with anti-SARS-CoV-2 activity, antifibrotic activity, and strong safety profiles for cancers may encourage the rapid application of functional NK cells as a potential therapeutic strategy to eliminate SARS-CoV-2-infected cells at an early stage, facilitate immune–immune cell interactions, and favor inflammatory processes that prevent and/or reverse over-inflammation and inhibit fibrosis progression, thereby helping in the fight against COVID-19. However, our understanding of the role of NK cells in COVID-19 remains incomplete, and further research on the involvement of NK cells in the pathogenesis of COVID-19 is needed. The rationale of NK-cell-based therapies for COVID-19 has to be based on the timing of therapeutic interventions and disease severity, which may be determined by the balance between beneficial antiviral and potential detrimental pathologic actions. NK cells would be more effective early in SARS-CoV-2 infection and prevent the progression of COVID-19. Immunomodulation by NK cells towards regulatory functions could be useful as an adjunct therapy to prevent the progression of COVID-19.
Collapse
|
43
|
Huot N, Planchais C, Contreras V, Jacquelin B, Petitdemange C, Lazzerini M, Rosenbaum P, Rey F, Reeves RK, Le Grand R, Mouquet H, Müller-Trutwin M. Adaptive MHC-E restricted tissue-resident NK cells are associated with persistent low antigen load in alveolar macrophages after SARS-CoV-2 infection. RESEARCH SQUARE 2022:rs.3.rs-1561222. [PMID: 35547853 PMCID: PMC9094104 DOI: 10.21203/rs.3.rs-1561222/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Natural killer (NK) cells are innate lymphocytes with potent activity against a wide range of viruses. In SARS-CoV-2 infection, NK cell activity might be of particular importance within lung tissues. Here, we investigated whether NK cells with activity against Spike+ cells are induced during SARS-CoV-2 infection and have a role in modulating viral persistence beyond primary clearance from nasopharyngeal and tracheal tissues. We performed an integrated analysis of NK cells and macrophages in blood and bronchoalveolar lavage fluids (BALF) of COVID-19 convalescent non-human primates in comparison to uninfected control animals. SARS-CoV-2 protein expression was detected for at least 9-18 months post-infection in alveolar macrophages. Convalescent animals segregated into two groups based on cellular phenotypes and viral persistence profiles in BALF. The animals with lower persistent antigen displayed macrophages with a regulatory phenotype and enhanced MHC-E restricted NK cell activity toward cells presenting peptides derived from the SARS-CoV-2 Spike protein leader sequence, while NK cell activity from the other convalescent animals, control animals and healthy humans were strongly inhibited by these Spike peptides. The adaptive NK cell activity was not detected in blood but in tissue-resident NK cells, and cross-reacted against MERS-CoV and SARS-CoV Spike-derived peptides.
Collapse
|
44
|
Mace EM. Profiling natural killers in COVID-19. J Allergy Clin Immunol 2022; 149:1223-1224. [PMID: 35051507 PMCID: PMC8761581 DOI: 10.1016/j.jaci.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Emily M Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York NY USA.
| |
Collapse
|
45
|
Casado JL, Moraga E, Vizcarra P, Velasco H, Martín-Hondarza A, Haemmerle J, Gómez S, Quereda C, Vallejo A. Expansion of CD56 dimCD16 neg NK Cell Subset and Increased Inhibitory KIRs in Hospitalized COVID-19 Patients. Viruses 2021; 14:v14010046. [PMID: 35062250 PMCID: PMC8780522 DOI: 10.3390/v14010046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 01/08/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2) infection induces elevated levels of inflammatory cytokines, which are mainly produced by the innate response to the virus. The role of NK cells, which are potent producers of IFN-γ and cytotoxicity, has not been sufficiently studied in the setting of SARS-CoV-2 infection. We confirmed a different distribution of NK cell subsets in hospitalized COVID-19 patients despite their NK cell deficiency. The impairment of this innate defense is mainly focused on the cytotoxic capacity of the CD56dim NK cells. On the one hand, we found an expansion of the CD56dimCD16neg NK subset, lower cytotoxic capacities, and high frequencies of inhibitory 2DL1 and 2DL1/S1 KIR receptors in COVID-19 patients. On the other hand, the depletion of CD56dimCD16dim/bright NK cell subsets, high cytotoxic capacities, and high frequencies of inhibitory 2DL1 KIR receptors were found in COVID-19 patients. In contrast, no differences in the distribution of CD56bright NK cell subsets were found in this study. These alterations in the distribution and phenotype of NK cells might enhance the impairment of this crucial innate line of defense during COVID-19 infection.
Collapse
Affiliation(s)
- José L. Casado
- Department of Infectious Diseases, Ramón y Cajal Institute for Health Research (IRyCIS), University Hospital Ramón y Cajal, 28034 Madrid, Spain; (E.M.); (P.V.); (H.V.); (A.M.-H.); (S.G.); (C.Q.)
- Correspondence: (J.L.C.); (A.V.)
| | - Elisa Moraga
- Department of Infectious Diseases, Ramón y Cajal Institute for Health Research (IRyCIS), University Hospital Ramón y Cajal, 28034 Madrid, Spain; (E.M.); (P.V.); (H.V.); (A.M.-H.); (S.G.); (C.Q.)
- Laboratory of Immunovirology, Ramón y Cajal Institute for Health Research, University Hospital Ramón y Cajal, 28034 Madrid, Spain
| | - Pilar Vizcarra
- Department of Infectious Diseases, Ramón y Cajal Institute for Health Research (IRyCIS), University Hospital Ramón y Cajal, 28034 Madrid, Spain; (E.M.); (P.V.); (H.V.); (A.M.-H.); (S.G.); (C.Q.)
| | - Héctor Velasco
- Department of Infectious Diseases, Ramón y Cajal Institute for Health Research (IRyCIS), University Hospital Ramón y Cajal, 28034 Madrid, Spain; (E.M.); (P.V.); (H.V.); (A.M.-H.); (S.G.); (C.Q.)
- Laboratory of Immunovirology, Ramón y Cajal Institute for Health Research, University Hospital Ramón y Cajal, 28034 Madrid, Spain
| | - Adrián Martín-Hondarza
- Department of Infectious Diseases, Ramón y Cajal Institute for Health Research (IRyCIS), University Hospital Ramón y Cajal, 28034 Madrid, Spain; (E.M.); (P.V.); (H.V.); (A.M.-H.); (S.G.); (C.Q.)
- Laboratory of Immunovirology, Ramón y Cajal Institute for Health Research, University Hospital Ramón y Cajal, 28034 Madrid, Spain
| | - Johannes Haemmerle
- Department of Prevention of Occupational Risks, University Hospital Ramón y Cajal, 28034 Madrid, Spain;
| | - Sandra Gómez
- Department of Infectious Diseases, Ramón y Cajal Institute for Health Research (IRyCIS), University Hospital Ramón y Cajal, 28034 Madrid, Spain; (E.M.); (P.V.); (H.V.); (A.M.-H.); (S.G.); (C.Q.)
| | - Carmen Quereda
- Department of Infectious Diseases, Ramón y Cajal Institute for Health Research (IRyCIS), University Hospital Ramón y Cajal, 28034 Madrid, Spain; (E.M.); (P.V.); (H.V.); (A.M.-H.); (S.G.); (C.Q.)
| | - Alejandro Vallejo
- Department of Infectious Diseases, Ramón y Cajal Institute for Health Research (IRyCIS), University Hospital Ramón y Cajal, 28034 Madrid, Spain; (E.M.); (P.V.); (H.V.); (A.M.-H.); (S.G.); (C.Q.)
- Laboratory of Immunovirology, Ramón y Cajal Institute for Health Research, University Hospital Ramón y Cajal, 28034 Madrid, Spain
- Correspondence: (J.L.C.); (A.V.)
| |
Collapse
|
46
|
Mitsuyama Y, Yamakawa K, Kayano K, Maruyama M, Wada T, Fujimi S. Prolonged enhancement of cytotoxic T lymphocytes in the post-recovery state of severe COVID-19. J Intensive Care 2021; 9:76. [PMID: 34930468 PMCID: PMC8685802 DOI: 10.1186/s40560-021-00591-3] [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: 09/18/2021] [Accepted: 12/07/2021] [Indexed: 11/22/2022] Open
Abstract
We evaluated the peripheral blood immune responses of lymphocytes in severe Coronavirus disease 2019 (COVID-19) patients in different stages of recovery using single-cell mass cytometry. The patients with prolonged hospitalization did not show recovery of B lymphocyte counts and CD4-positive T lymphocyte counts but did show abundant CD8-positive T lymphocytes. CD4 and CD8 T cells expressing high levels of T-bet and Granzyme B were more abundant in post-recovery patients. This study showed that cytotoxic Th1 and CD8 T cells are recruited to the peripheral blood long after recovery from COVID-19.
Collapse
Affiliation(s)
- Yumi Mitsuyama
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | - Kazuma Yamakawa
- Department of Emergency Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan.
| | - Katsuhide Kayano
- Department of Emergency Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan
| | - Miho Maruyama
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | - Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Fujimi
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| |
Collapse
|