1
|
Jermakow N, Brodaczewska K, Kot J, Lubas A, Kłos K, Siewiera J. Bayesian Modeling of the Impact of HBOT on the Reduction in Cytokine Storms. J Clin Med 2025; 14:1180. [PMID: 40004710 PMCID: PMC11856955 DOI: 10.3390/jcm14041180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Since the initial identification of SARS-CoV-2 infections, numerous clinical challenges have arisen, revealing both acute and long-term effects associated with COVID-19. These effects impact various systems within the body, including the respiratory, cardiovascular, and nervous systems. Background/Objectives: This study aimed to investigate the immunological and inflammatory parameters in patients with severe COVID-19 and evaluate the effects of hyperbaric oxygen therapy (HBOT) on these parameters. Methods: This study enrolled thirty patients from the Military Medical Institute-National Research Institute in Warsaw, who were hospitalized for SARS-CoV-2 infection. Patients were screened for eligibility based on pre-defined inclusion criteria. The subjects were randomly assigned to one of two groups: hyperbaric oxygen therapy (HBOT) or a control group. Immune profiling was performed, measuring cytokine concentrations and leukocyte subpopulations in serum samples. Outcomes were assessed using Bayesian modeling. Results: Bayesian regression analysis confirmed previous findings, indicating that HBOT may reduce inflammatory cytokine levels while improving oxygen saturation (SpO2) in patients with moderate and severe COVID-19. Moreover, the analysis suggested a higher probability of HBOT success in modulating the immune response and reducing inflammatory parameters, particularly in T lymphocyte subpopulations. Conclusions: Hyperbaric oxygen therapy (HBOT) may serve as an effective adjunctive treatment for patients with COVID-19 by enhancing oxygen saturation and modulating the immune response. Further studies are needed to elucidate the underlying mechanisms of HBOT on inflammatory and immunological parameters in COVID-19 patients.
Collapse
Affiliation(s)
- Natalia Jermakow
- Department of Hyperbaric Medicine, Military Institute of Medicine, National Science Institute, Szaserów 128, 04-141 Warsaw, Poland;
| | - Klaudia Brodaczewska
- The Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, National Science Institute, Szaserów 128, 04-141 Warsaw, Poland;
| | - Jacek Kot
- National Centre for Hyperbaric Medicine, Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Powstania Styczniowego 9B, 81-519 Gdynia, Poland;
| | - Arkadiusz Lubas
- Department of Internal Diseases Nephrology and Dialysis, Military Institute of Medicine, National Science Institute, Szaserów 128, 04-141 Warsaw, Poland;
| | - Krzysztof Kłos
- Department of Infectious Diseases and Allergology, Military Institute of Medicine, National Science Institute, Szaserów 128, 04-141 Warsaw, Poland;
| | - Jacek Siewiera
- Department of Hyperbaric Medicine, Military Institute of Medicine, National Science Institute, Szaserów 128, 04-141 Warsaw, Poland;
| |
Collapse
|
2
|
Flores-Gonzalez J, Buendia-Roldan I, Téllez-Quijada F, Peña-Bates C, Ramón-Luing LA, Castorena-Maldonado A, Falfán-Valencia R, Pérez-Rubio G, Selman M, Chavez-Galan L, Chávez-Galán L. Altered immune surveillance of B and T cells in patients with persistent residual lung abnormalities 12 months after severe COVID-19. Respir Res 2025; 26:22. [PMID: 39827348 PMCID: PMC11742501 DOI: 10.1186/s12931-025-03102-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/05/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Post-COVID-19 respiratory sequelae often involve lung damage, which is called residual lung abnormalities, and potentially lead to chronic respiratory issues. The adaptive immune response, involving T-cells and B-cells, plays a critical role in pathogen control, inflammation, and tissue repair. However, the link between immune dysregulation and the development of residual lung abnormalities remains unclear. METHODS 109 patients discharged with residual lung abnormalities after a critical COVID-19 were followed for 12 months and divided as full recovery patients (FRG, n = 88) and persistent lung abnormalities (PLAG, n = 21). Cell profiling analysis was done using flow cytometry at 24 h of not antigen-specific in vitro stimulation. Plasma or supernatant levels of IFN-g, IL-4, IL-10, IgM, and IgG were assessed, and 10 patients (5 FRG, 5 PLAG) were randomly selected for detailed immune cell phenotyping and functional analysis of peripheral blood mononuclear cells using flow cytometry. RESULTS Compared to the FRG group, PLAG exhibited an increase of unswitched (p = 0.0159) and decreased double-negative activated B-cells (p = 0.0317), systemic IL-10 levels were lower, displayed reduced frequency of total B-cells, and impaired spontaneous IgM (p = 0.0357) and IgG (p = 0.0079) release in culture. Regarding T-cells, PLAG patients showed a reduction in effector memory CD4 + cells (p = 0.0159) and an increase in CD4 + TEMRA cells (p = 0.0079) following in vitro stimulation. Notably, the PLAG group also exhibited higher frequencies of central memory CD4 + Th2 (GATA3+) T-cells in response to activation than the FRG group (p = 0.0079). CONCLUSIONS Patients with residual lung abnormalities 12 months post-critical COVID-19 exhibit impaired B-cell function, increased unswitched B-cells, and higher frequencies of CD4 + TEMRA T-cells following in vitro activation. These immune imbalances may contribute to ongoing lung dysfunction and warrant further investigation as a potential mechanism in residual lung abnormalities. Larger studies are necessary to confirm these findings.
Collapse
Affiliation(s)
- Julio Flores-Gonzalez
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Ivette Buendia-Roldan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Fernanda Téllez-Quijada
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Carlos Peña-Bates
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Lucero A Ramón-Luing
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | | | - Ramcés Falfán-Valencia
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Gloria Pérez-Rubio
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| | - Leslie Chavez-Galan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico.
| | - Leslie Chávez-Galán
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, 14080, Mexico
| |
Collapse
|
3
|
Long H, Tai Y, Fan J, Ou X, Yan L, Fan Y, Feng W, Chen J, Li Y. Characteristics of peripheral lymphocyte subsets and antibodies in COVID-19-infected kidney transplantation recipients. Int Immunopharmacol 2025; 145:113755. [PMID: 39672021 DOI: 10.1016/j.intimp.2024.113755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Peripheral lymphocyte subsets play vital roles in various disease conditions. However, the roles of kidney transplant recipients (KTRs) with novel coronavirus pneumonia (COVID-19) are still unclear. In this research, we investigated the predictive value of peripheral blood lymphocyte subsets on the severity of KTRs with COVID-19 and the correlation between antibodies and lymphocyte levels. METHODS 84 patients with kidney transplantation combined with COVID-19 admitted from December 2022 to February 2023 were included. On the basis of the severity of COVID-19, they were categorized into a mild (n = 49) and a severe group (n = 35). The logistic regression method was utilized to build the critical risk prediction model for KTRs complicated with COVID-19. The receiver operator characteristic curve (ROC), calibration plot and clinical decision curve analysis (DCA) were applied to assess the discrimination, calibration and clinical application value of this model. The Spearman correlation test was applied to examine the connection between antibodies and various immune indexes. RESULTS Except for the increase of CD4+HLA-DR+ T cells, the number of CD3+, CD4+, and CD8+ T cell subsets in severe was lower than that in mild (P < 0.05). IL-6 in severe was higher than mild (P < 0.05). After screening variables, we established a regression equation prediction model, logit (P) = 4.965+ (-0.038) × (CD3+/lymphocytes (%)) + 0.064× (CD4+HLA-DR+/ CD4+ T cells (%)) + (-0.040) × (CD14+HLA-DR+/monocytes (%)). The area under the ROC curve (AUC) of the prediction model was 0.779 (95 % CI 0.679-0.879, P = 0.001). The cut-off value was 0.308, with a prediction sensitivity of 0.829 (95 % CI 0.657-0.928) and a specificity of 0.653 (95 % CI 0.503-0.779). Logistic regression analysis showed the increase in the percentage of CD4+HLA-DR+ T cells among CD4+ T cells was a risk factor for COVID-19 severity among kidney transplant recipients, while the increase in the percentage of CD3+ T cells among lymphocytes and CD14+HLA-DR+ monocytes among CD14+ monocytes acted as protective factors. COVID-19 antibodies were negatively correlated with CD8+CD45RA+CD27- (Terminally Differentiated Effector Memory T Cells, TEMRA), CD8+CD28-, CD8+CD38+ and CD4+CD38+ T cells, while positively correlated with CD8+CD45RA-CD27- (Effector Memory T cells, T8EM), CD8+CD45RA-CD27+ (Central Memory T cells, T8CM) and CD8+CD28+ T cells. CONCLUSION A predictive model was developed and validated for predicting the severe form of COVID-19 in KTRs. The model showed good predictive ability, concordance, and potential clinical utility.
Collapse
Affiliation(s)
- Honghui Long
- Department of Transfusion Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yunze Tai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jiwen Fan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xiaoqi Ou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Lin Yan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yu Fan
- Department of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu 610041, China
| | - Weihua Feng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jie Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China.
| |
Collapse
|
4
|
Wang X, Chen J, Wu W, Fan J, Huang L, Sun W, He K, Li S, Li C. Circulating memory T cells and TCF1 + T cells aid in diagnosis and monitor disease activity in vitiligo. J Pharm Anal 2024; 14:100998. [PMID: 39698313 PMCID: PMC11652857 DOI: 10.1016/j.jpha.2024.100998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/28/2024] [Accepted: 05/05/2024] [Indexed: 12/20/2024] Open
Abstract
Vitiligo is an immune memory skin disease. T-cell factor 1 (TCF1) is essential for maintaining the memory T-cell pool. There is an urgent need to investigate the characteristics of peripheral memory T-cell profile and TCF1+ T-cell frequencies in patients with vitiligo. In this study, 31 patients with active vitiligo (AV), 22 with stable vitiligo (SV), and 30 healthy controls (HCs) were included. We measured circulating memory and TCF1+ T-cell frequencies using flow cytometry. The Spearman's rank test was used to evaluate the correlation between cell frequencies and disease characteristics. Receiver operating characteristic curves (ROC) were constructed to investigate the discriminative power of the cell subpopulations. Circulating CD4+ and CD8+ terminally differentiated effector memory T-cell (TEMRA) frequencies were significantly higher in the AV group than in HCs (P < 0.05). TCF1+ T-cell subpopulations were widespread increased in patients with vitiligo (P < 0.05). After adjusting for potential confounders, CD8+ and CD4+ central memory (TCM) cells, and CD8+ TEMRA were correlated with disease activity (P < 0.05). The combined diagnostic value of the four (naïve, effector memory, TCM, and TEMRA) CD8+TCF1+ T-cell subsets was relatively high (area under the ROC curve (AUC) = 0.804, sensitivity = 71.70%, specificity = 83.34%), and the CD8+ T-cell subsets combination performed well in discriminating disease activity (AUC = 0.849, sensitivity = 70.97%, specificity = 90.91%). We demonstrated an altered circulating memory T-cell profile and increased TCF1+ T-cell percentage in patients with vitiligo. T-cell subpopulations had a strong value for vitiligo diagnosis and activity evaluation. This evidence presents a potential new pharmacological target for inhibiting autoimmunity that leads to vitiligo.
Collapse
Affiliation(s)
| | | | | | - Jinrong Fan
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Luling Huang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Weiwei Sun
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Kaiqiao He
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shuli Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
5
|
Oliván-Blázquez B, Bona-Otal M, Méndez-López F, Lerma-Irureta D, García-Izuel P, Ibáñez-Ruiz J, Montolío A, Ruiz-Herreros M, Godino J, Jimeno-Beltran B, Encabo-Berzosa MDM, Arenaz I, Medel-Martínez A, Casado-Vicente V, Coiras M, Tellería-Orriols C, Schoorlemmer J, Magallón-Botaya R. Characterization model of the post COVID-19 condition based on immunological, biochemical, and cytokine markers. iScience 2024; 27:110839. [PMID: 39318534 PMCID: PMC11420445 DOI: 10.1016/j.isci.2024.110839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 06/02/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Post-coronavirus disease condition (PCC) continues to affect many people globally, yet there remains a lack of diagnostic biomarkers to distinguish PCC from those recovered from acute COVID-19. This study compared biomarkers between two age- and gender-matched groups: PCC individuals and those recovered within three months of acute COVID-19 in 2020 (n = 85 each). Biomarkers were assessed 12-24 months after initial diagnosis, examining biochemical profiles, blood cell counts, coagulation status, antibody serology, lymphocyte populations, and cytokine levels. PCC individuals exhibited significant alterations in 49 of 167 markers, including K+ levels, αGAD antibodies, antithrombin III, insulin-like growth factor-binding protein 3 (IGFBP3), and interleukin-10 (IL-10). A panel of αGAD, IL-10, potassium levels, and CD16brightCD56- cell presence distinguished PCC individuals from recovered patients with >88% accuracy and <92% precision.
Collapse
Affiliation(s)
- Bárbara Oliván-Blázquez
- Department of Psychology and Sociology, University of Zaragoza, Zaragoza, Spain
- Aragonese Primary Care Research Group (GAIAP), Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Carlos III Health Institute, Madrid, Spain
| | | | - Fátima Méndez-López
- Aragonese Primary Care Research Group (GAIAP), Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Carlos III Health Institute, Madrid, Spain
| | - David Lerma-Irureta
- Aragonese Primary Care Research Group (GAIAP), Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
| | - Paula García-Izuel
- Department of Medicine, Psychiatry and Dermatology, University of Zaragoza, Zaragoza, Spain
| | - Jesús Ibáñez-Ruiz
- Biocomputing Unit, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Alberto Montolío
- Biocomputing Unit, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - María Ruiz-Herreros
- Tarazona Primary Health Care Center, Aragonese Healthcare Service (SALUD), Zaragoza, Spain
| | - Javier Godino
- Cell Separation and Flow Cytometry Core, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Beatriz Jimeno-Beltran
- Cell Separation and Flow Cytometry Core, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | | | - Izaskun Arenaz
- Biobank of the Aragon Health System, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Ana Medel-Martínez
- Placental Pathophysiology & Fetal Programming Research Group, Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
| | - Verónica Casado-Vicente
- Parquesol University Health Center, Castilla y León Health Service (SACYL), University Teaching Unit of Family and Community Medicine, University of Valladolid, Valladolid, Spain
| | - Mayte Coiras
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Jon Schoorlemmer
- Endogenous Retroviruses (ERVs) in Development and Disease Group, Instituto Aragonés deCiencias de la Salud (IACS), Zaragoza, Spain; and ARAID Foundation, Zaragoza, Spain
| | - Rosa Magallón-Botaya
- Aragonese Primary Care Research Group (GAIAP), Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Carlos III Health Institute, Madrid, Spain
- Department of Medicine, Psychiatry and Dermatology, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
6
|
Kumagai Y. BootCellNet, a resampling-based procedure, promotes unsupervised identification of cell populations via robust inference of gene regulatory networks. PLoS Comput Biol 2024; 20:e1012480. [PMID: 39348410 PMCID: PMC11466406 DOI: 10.1371/journal.pcbi.1012480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 10/10/2024] [Accepted: 09/11/2024] [Indexed: 10/02/2024] Open
Abstract
Recent advances in measurement technologies, particularly single-cell RNA sequencing (scRNA-seq), have revolutionized our ability to acquire large amounts of omics-level data on cellular states. As measurement techniques evolve, there has been an increasing need for data analysis methodologies, especially those focused on cell-type identification and inference of gene regulatory networks (GRNs). We have developed a new method named BootCellNet, which employs smoothing and resampling to infer GRNs. Using the inferred GRNs, BootCellNet further infers the minimum dominating set (MDS), a set of genes that determines the dynamics of the entire network. We have demonstrated that BootCellNet robustly infers GRNs and their MDSs from scRNA-seq data and facilitates unsupervised identification of cell clusters using scRNA-seq datasets of peripheral blood mononuclear cells and hematopoiesis. It has also identified COVID-19 patient-specific cells and their potential regulatory transcription factors. BootCellNet not only identifies cell types in an unsupervised and explainable way but also provides insights into the characteristics of identified cell types through the inference of GRNs and MDS.
Collapse
Affiliation(s)
- Yutaro Kumagai
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Higashi, Tsukuba, Ibaraki, Japan
| |
Collapse
|
7
|
Gay L, Rouviere MS, Mezouar S, Richaud M, Gorvel L, Foucher E, La Scola B, Menard A, Allardet-Servent J, Halfon P, Frohna P, Cano C, Mege JL, Olive D. Vγ9Vδ2 T-cells Are Potent Inhibitors of SARS-CoV-2 Replication and Represent Effector Phenotypes in Patients With COVID-19. J Infect Dis 2024; 229:1759-1769. [PMID: 38557809 DOI: 10.1093/infdis/jiae169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/11/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
Vγ9Vδ2 T cells play a key role in the innate immune response to viral infections through butyrophilin 3A (BTN3A). Here, we report blood Vγ9Vδ2 T cells decreased in clinically mild COVID-19 compared to healthy volunteers, and this was maintained up to 28 days and in the recovery period. Terminally differentiated Vγ9Vδ2 T cells tended to be enriched on the day of diagnosis, 28 days after, and during the recovery period. These cells showed cytotoxic and inflammatory activities following anti-BTN3A activation. BTN3A upregulation and Vγ9Vδ2 T-cell infiltration were observed in a lung biopsy from a fatal SARS-CoV-2 infection. In vitro, SARS-CoV-2 infection increased BTN3A expression in macrophages and lung cells that enhanced the anti-SARS-CoV-2 Vγ9Vδ2 T-cell cytotoxicity and interferon-γ and tumor necrosis factor-α. Increasing concentrations of anti-BTN3A lead to viral replication inhibition. Altogether, we report Vγ9Vδ2 T cells are important in the immune response against SARS-CoV-2 infection and activation by anti-BTN3A antibody may enhance their response. Clinical Trials Registration. NCT04816760.
Collapse
Affiliation(s)
- Laetitia Gay
- Institut de recherche pour le developpement (IRD), Assistance-Publique Hopitaux de Marseille (APHM), Microbes Evolution Phylogénie et Infections (MEPHI), Aix-Marseille University, Marseille, France
- ImCheck Therapeutics, Marseille, France
| | - Marie-Sarah Rouviere
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm UMR1068, CNRS UMR7258, Institut Paoli Calmettes, Marseille, France
| | - Soraya Mezouar
- Etablissement Français du Sang, Centre National de la Recherche Scientifique, Anthropologie Bio-Culturelle, Droit, Éthique et Santé, "Biologie des Groupes Sanguins," Aix-Marseille University, Marseille, France
| | - Manon Richaud
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm UMR1068, CNRS UMR7258, Institut Paoli Calmettes, Marseille, France
| | - Laurent Gorvel
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm UMR1068, CNRS UMR7258, Institut Paoli Calmettes, Marseille, France
| | | | - Bernard La Scola
- Institut de recherche pour le developpement (IRD), Assistance-Publique Hopitaux de Marseille (APHM), Microbes Evolution Phylogénie et Infections (MEPHI), Aix-Marseille University, Marseille, France
| | - Amélie Menard
- Unité COVID-Long, Service de Médecine Interne, Centre Hospitalo-Universitaire Nord (CHU NORD), Assistance-Publique Hopitaux de Marseille (APHM), Marseille, France
| | | | - Philippe Halfon
- Département de Médecine Interne et Maladies Infectieuses, Hôpital Européen-Laboratoire Alphabio-Biogroup, Marseille, France
| | | | | | - Jean-Louis Mege
- Institut de recherche pour le developpement (IRD), Assistance-Publique Hopitaux de Marseille (APHM), Microbes Evolution Phylogénie et Infections (MEPHI), Aix-Marseille University, Marseille, France
- Assistance-Publique Hopitaux de Marseille (APHM), Hôpital de la Conception, Laboratoire d'Immunologie, Aix-Marseille University, Marseille, France
| | - Daniel Olive
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm UMR1068, CNRS UMR7258, Institut Paoli Calmettes, Marseille, France
| |
Collapse
|
8
|
Andreu-Ballester JC, Galindo-Regal L, Cuéllar C, López-Chuliá F, García-Ballesteros C, Fernández-Murga L, Llombart-Cussac A, Domínguez-Márquez MV. A Low Number of Baselines γδ T Cells Increases the Risk of SARS-CoV-2 Post-Vaccination Infection. Vaccines (Basel) 2024; 12:553. [PMID: 38793803 PMCID: PMC11125751 DOI: 10.3390/vaccines12050553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Background: The COVID-19 pandemic is the biggest global health problem in the last hundred years. The efficacy of the vaccine to protect against severe disease is estimated to be 70-95% according to the studies carried out, although there are aspects of the immune response to the vaccine that remain unclear. Methods: Humoral and cellular immunity after the administration of three doses of the Pfizer-BioNTech and Oxford AstraZeneca vaccines against SARS-CoV-2 over one year and the appearance of post-vaccination COVID-19 were studied. SARS-CoV-2 IgG and IgA antibodies, αβ and γδ T-cell subsets, and their differentiation stages and apoptosis were analyzed. Results: Anti-SARS-CoV-2 IgG and IgA antibodies showed a progressive increase throughout the duration of the study. This increase was the greatest after the third dose. The highest levels were observed in subjects who had anti-SARS-CoV-2 antibodies prior to vaccination. There was an increase in CD4+ αβ, CD8+ γδ and TEM CD8+ γδ T cells, and a decrease in apoptosis in CD4+ CD8+ and CD56+ αβ and γδ T cells. Post-vaccination SARS-CoV-2 infection was greater than 60%. The symptoms of COVID-19 were very mild and were related to a γδ T cell deficit, specifically CD8+ TEMRA and CD56+ γδ TEM, as well as lower pre-vaccine apoptosis levels. Conclusions: The results unveil the important role of γδ T cells in SARS-CoV-2-vaccine-mediated protection from the disease.
Collapse
Affiliation(s)
- Juan Carlos Andreu-Ballester
- FISABIO Foundation, 46020 Valencia, Spain; (L.G.-R.); (F.L.-C.)
- Parasitic Immunobiology and Immunomodulation Research Group (INMUNOPAR), Complutense University of Madrid, 28040 Madrid, Spain;
| | - Lorena Galindo-Regal
- FISABIO Foundation, 46020 Valencia, Spain; (L.G.-R.); (F.L.-C.)
- Laboratory of Molecular Biology and Research Department, Arnau de Vilanova University Hospital, FISABIO Foundation, 46015 Valencia, Spain;
| | - Carmen Cuéllar
- Parasitic Immunobiology and Immunomodulation Research Group (INMUNOPAR), Complutense University of Madrid, 28040 Madrid, Spain;
- Microbiology and Parasitology Department, Complutense University, 28040 Madrid, Spain
| | - Francisca López-Chuliá
- FISABIO Foundation, 46020 Valencia, Spain; (L.G.-R.); (F.L.-C.)
- Hematology Department, Arnau de Vilanova Hospital, 46015 Valencia, Spain
- Medicine Department, Cardenal Herrera University, 46115 Valencia, Spain
| | - Carlos García-Ballesteros
- Laboratory of Molecular Biology and Research Department, Arnau de Vilanova University Hospital, FISABIO Foundation, 46015 Valencia, Spain;
- Hematology Department, Arnau de Vilanova Hospital, 46015 Valencia, Spain
| | | | | | | |
Collapse
|
9
|
Casado-Fernández G, Cantón J, Nasarre L, Ramos-Martín F, Manzanares M, Sánchez-Menéndez C, Fuertes D, Mateos E, Murciano-Antón MA, Pérez-Olmeda M, Cervero M, Torres M, Rodríguez-Rosado R, Coiras M. Pre-existing cell populations with cytotoxic activity against SARS-CoV-2 in people with HIV and normal CD4/CD8 ratio previously unexposed to the virus. Front Immunol 2024; 15:1362621. [PMID: 38812512 PMCID: PMC11133563 DOI: 10.3389/fimmu.2024.1362621] [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: 12/28/2023] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
Introduction HIV-1 infection may produce a detrimental effect on the immune response. Early start of antiretroviral therapy (ART) is recommended to preserve the integrity of the immune system. In fact, people with HIV (PWH) and normal CD4/CD8 ratio appear not to be more susceptible to severe forms of COVID-19 than the general population and they usually present a good seroconversion rate in response to vaccination against SARS-CoV-2. However, few studies have fully characterized the development of cytotoxic immune populations in response to COVID-19 vaccination in these individuals. Methods In this study, we recruited PWH with median time of HIV-1 infection of 6 years, median CD4/CD8 ratio of 1.0, good adherence to ART, persistently undetectable viral load, and negative serology against SARS-CoV-2, who then received the complete vaccination schedule against COVID-19. Blood samples were taken before vaccination against COVID-19 and one month after receiving the complete vaccination schedule. Results PWH produced high levels of IgG against SARS-CoV-2 in response to vaccination that were comparable to healthy donors, with a significantly higher neutralization capacity. Interestingly, the cytotoxic activity of PBMCs from PWH against SARS-CoV-2-infected cells was higher than healthy donors before receiving the vaccination schedule, pointing out the pre-existence of activated cell populations with likely unspecific antiviral activity. The characterization of these cytotoxic cell populations revealed high levels of Tgd cells with degranulation capacity against SARS-CoV-2-infected cells. In response to vaccination, the degranulation capacity of CD8+ T cells also increased in PWH but not in healthy donors. Discussion The full vaccination schedule against COVID-19 did not modify the ability to respond against HIV-1-infected cells in PWH and these individuals did not show more susceptibility to breakthrough infection with SARS-CoV-2 than healthy donors after 12 months of follow-up. These results revealed the development of protective cell populations with broad-spectrum antiviral activity in PWH with normal CD4/CD8 ratio and confirmed the importance of early ART and treatment adherence to avoid immune dysfunctions.
Collapse
Affiliation(s)
- Guiomar Casado-Fernández
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- PhD Program in Health Sciences, Faculty of Sciences, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Juan Cantón
- PhD Program in Health Sciences, Faculty of Sciences, Universidad de Alcalá, Alcalá de Henares, Spain
- Internal Medicine Service, Hospital Universitario Severo Ochoa, Leganés, Madrid, Spain
| | - Laura Nasarre
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Fernando Ramos-Martín
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Mario Manzanares
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- PhD Program in Biomedical Sciences and Public Health, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Clara Sánchez-Menéndez
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- PhD Program in Biomedical Sciences and Public Health, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Daniel Fuertes
- School of Telecommunications Engineering, Universidad Politécnica de Madrid, Madrid, Spain
| | - Elena Mateos
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases [Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFEC)], Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - María Aranzazu Murciano-Antón
- Family Medicine, Centro de Salud Doctor Pedro Laín Entralgo, Alcorcón, Madrid, Spain
- International PhD School, Universidad Rey Juan Carlos, Alcorcón, Madrid, Spain
| | - Mayte Pérez-Olmeda
- Biomedical Research Center Network in Infectious Diseases [Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFEC)], Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Serology Service, Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel Cervero
- Internal Medicine Service, Hospital Universitario Severo Ochoa, Leganés, Madrid, Spain
- School of Medicine, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Montserrat Torres
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases [Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFEC)], Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Rafael Rodríguez-Rosado
- Internal Medicine Service, Hospital Universitario Severo Ochoa, Leganés, Madrid, Spain
- School of Medicine, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Mayte Coiras
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Biomedical Research Center Network in Infectious Diseases [Centro de Investigación Biomédica en Red Enfermedades Infecciosas (CIBERINFEC)], Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| |
Collapse
|
10
|
Terzoli S, Marzano P, Cazzetta V, Piazza R, Sandrock I, Ravens S, Tan L, Prinz I, Balin S, Calvi M, Carletti A, Cancellara A, Coianiz N, Franzese S, Frigo A, Voza A, Calcaterra F, Di Vito C, Della Bella S, Mikulak J, Mavilio D. Expansion of memory Vδ2 T cells following SARS-CoV-2 vaccination revealed by temporal single-cell transcriptomics. NPJ Vaccines 2024; 9:63. [PMID: 38509155 PMCID: PMC10954735 DOI: 10.1038/s41541-024-00853-9] [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: 01/20/2023] [Accepted: 03/05/2024] [Indexed: 03/22/2024] Open
Abstract
γδ T cells provide rapid cellular immunity against pathogens. Here, we conducted matched single-cell RNA-sequencing and γδ-TCR-sequencing to delineate the molecular changes in γδ T cells during a longitudinal study following mRNA SARS-CoV-2 vaccination. While the first dose of vaccine primes Vδ2 T cells, it is the second administration that significantly boosts their immune response. Specifically, the second vaccination uncovers memory features of Vδ2 T cells, shaped by the induction of AP-1 family transcription factors and characterized by a convergent central memory signature, clonal expansion, and an enhanced effector potential. This temporally distinct effector response of Vδ2 T cells was also confirmed in vitro upon stimulation with SARS-CoV-2 spike-peptides. Indeed, the second challenge triggers a significantly higher production of IFNγ by Vδ2 T cells. Collectively, our findings suggest that mRNA SARS-CoV-2 vaccination might benefit from the establishment of long-lasting central memory Vδ2 T cells to confer protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Sara Terzoli
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
| | - Paolo Marzano
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Valentina Cazzetta
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School (MHH), Hannover, Germany
| | - Sarina Ravens
- Institute of Immunology, Hannover Medical School (MHH), Hannover, Germany
| | - Likai Tan
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School (MHH), Hannover, Germany
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Balin
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Michela Calvi
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Anna Carletti
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Assunta Cancellara
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Nicolò Coianiz
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Sara Franzese
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Alessandro Frigo
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Antonio Voza
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
- Department of Biomedical Unit, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Francesca Calcaterra
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Clara Di Vito
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Silvia Della Bella
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Joanna Mikulak
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy.
| | - Domenico Mavilio
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Rozzano, Italy.
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| |
Collapse
|
11
|
Cezar R, Kundura L, André S, Lozano C, Vincent T, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Loubet P, Sotto A, Tran TA, Estaquier J, Corbeau P. T4 apoptosis in the acute phase of SARS-CoV-2 infection predicts long COVID. Front Immunol 2024; 14:1335352. [PMID: 38235145 PMCID: PMC10791767 DOI: 10.3389/fimmu.2023.1335352] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024] Open
Abstract
Background As about 10% of patients with COVID-19 present sequelae, it is important to better understand the physiopathology of so-called long COVID. Method To this aim, we recruited 29 patients hospitalized for SARS-CoV-2 infection and, by Luminex®, quantified 19 soluble factors in their plasma and in the supernatant of their peripheral blood mononuclear cells, including inflammatory and anti-inflammatory cytokines and chemokines, Th1/Th2/Th17 cytokines, and endothelium activation markers. We also measured their T4, T8 and NK differentiation, activation, exhaustion and senescence, T cell apoptosis, and monocyte subpopulations by flow cytometry. We compared these markers between participants who developed long COVID or not one year later. Results None of these markers was predictive for sequelae, except programmed T4 cell death. T4 lymphocytes from participants who later presented long COVID were more apoptotic in culture than those of sequelae-free participants at Month 12 (36.9 ± 14.7 vs. 24.2 ± 9.0%, p = 0.016). Conclusions Our observation raises the hypothesis that T4 cell death during the acute phase of SARS-CoV-2 infection might pave the way for long COVID. Mechanistically, T4 lymphopenia might favor phenomena that could cause sequelae, including SARS-CoV-2 persistence, reactivation of other viruses, autoimmunity and immune dysregulation. In this scenario, inhibiting T cell apoptosis, for instance, by caspase inhibitors, could prevent long COVID.
Collapse
Affiliation(s)
- Renaud Cezar
- Immunology Department, Nîmes University Hospital, Nîmes, France
| | - Lucy Kundura
- Institute of Human Genetics, UMR9002, Centre National de la Recherche Scientifique (CNRS) and Montpellier University, Montpellier, France
| | - Sonia André
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1124, Université de Paris, Paris, France
| | - Claire Lozano
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Thierry Vincent
- Immunology Department, Montpellier University Hospital, Montpellier, France
| | - Laurent Muller
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Jean-Yves Lefrant
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Claire Roger
- Surgical Intensive Care Department, Nîmes University Hospital, Nîmes, France
| | - Pierre-Géraud Claret
- Medical and Surgical Emergency Department, Nîmes University Hospital, Nîmes, France
| | - Sandra Duvnjak
- Gerontology Department, Nîmes University Hospital, Nîmes, France
| | - Paul Loubet
- Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Albert Sotto
- Infectious Diseases Department, Nîmes University Hospital, Nîmes, France
| | - Tu-Ahn Tran
- Pediatrics Department, Nîmes University Hospital, Nîmes, France
| | - Jérôme Estaquier
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1124, Université de Paris, Paris, France
- Laval University Research Center, Quebec City, QC, Canada
| | - Pierre Corbeau
- Immunology Department, Nîmes University Hospital, Nîmes, France
- Institute of Human Genetics, UMR9002, Centre National de la Recherche Scientifique (CNRS) and Montpellier University, Montpellier, France
| |
Collapse
|
12
|
Reagin KL, Lee RL, Cocciolone L, Funk KE. Antigen non-specific CD8 + T cells accelerate cognitive decline in aged mice following respiratory coronavirus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.02.573675. [PMID: 38260669 PMCID: PMC10802364 DOI: 10.1101/2024.01.02.573675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Primarily a respiratory infection, numerous patients infected with SARS-CoV-2 present with neurologic symptoms, some continuing long after viral clearance as a persistent symptomatic phase termed "long COVID". Advanced age increases the risk of severe disease, as well as incidence of long COVID. We hypothesized that perturbations in the aged immune response predispose elderly individuals to severe coronavirus infection and post-infectious sequelae. Using a murine model of respiratory coronavirus, mouse hepatitis virus strain A59 (MHV-A59), we found that aging increased clinical illness and lethality to MHV infection, with aged animals harboring increased virus in the brain during acute infection. This was coupled with an unexpected increase in activated CD8+ T cells within the brains of aged animals but reduced antigen specificity of those CD8+ T cells. Aged animals demonstrated spatial learning impairment following MHV infection, which correlated with increased neuronal cell death and reduced neuronal regeneration in aged hippocampus. Using primary cell culture, we demonstrated that activated CD8+ T cells induce neuronal death, independent of antigen-specificity. Specifically, higher levels of CD8+ T cell-derived IFN-γ correlated with neuronal death. These results support the evidence that CD8+ T cells in the brain directly contribute to cognitive dysfunction following coronavirus infection in aged individuals.
Collapse
Affiliation(s)
- Katie L. Reagin
- Department of Biological Sciences, University of North Carolina at Charlotte
| | - Rae-Ling Lee
- Department of Biological Sciences, University of North Carolina at Charlotte
| | - Loren Cocciolone
- Department of Biological Sciences, University of North Carolina at Charlotte
| | - Kristen E. Funk
- Department of Biological Sciences, University of North Carolina at Charlotte
| |
Collapse
|
13
|
Hu Y, Hu Q, Li Y, Lu L, Xiang Z, Yin Z, Kabelitz D, Wu Y. γδ T cells: origin and fate, subsets, diseases and immunotherapy. Signal Transduct Target Ther 2023; 8:434. [PMID: 37989744 PMCID: PMC10663641 DOI: 10.1038/s41392-023-01653-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 11/23/2023] Open
Abstract
The intricacy of diseases, shaped by intrinsic processes like immune system exhaustion and hyperactivation, highlights the potential of immune renormalization as a promising strategy in disease treatment. In recent years, our primary focus has centered on γδ T cell-based immunotherapy, particularly pioneering the use of allogeneic Vδ2+ γδ T cells for treating late-stage solid tumors and tuberculosis patients. However, we recognize untapped potential and optimization opportunities to fully harness γδ T cell effector functions in immunotherapy. This review aims to thoroughly examine γδ T cell immunology and its role in diseases. Initially, we elucidate functional differences between γδ T cells and their αβ T cell counterparts. We also provide an overview of major milestones in γδ T cell research since their discovery in 1984. Furthermore, we delve into the intricate biological processes governing their origin, development, fate decisions, and T cell receptor (TCR) rearrangement within the thymus. By examining the mechanisms underlying the anti-tumor functions of distinct γδ T cell subtypes based on γδTCR structure or cytokine release, we emphasize the importance of accurate subtyping in understanding γδ T cell function. We also explore the microenvironment-dependent functions of γδ T cell subsets, particularly in infectious diseases, autoimmune conditions, hematological malignancies, and solid tumors. Finally, we propose future strategies for utilizing allogeneic γδ T cells in tumor immunotherapy. Through this comprehensive review, we aim to provide readers with a holistic understanding of the molecular fundamentals and translational research frontiers of γδ T cells, ultimately contributing to further advancements in harnessing the therapeutic potential of γδ T cells.
Collapse
Affiliation(s)
- Yi Hu
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Qinglin Hu
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China
| | - Zheng Xiang
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Zhinan Yin
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts-University Kiel, Kiel, Germany.
| | - Yangzhe Wu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China.
| |
Collapse
|
14
|
Uvarova AN, Stasevich EM, Ustiugova AS, Mitkin NA, Zheremyan EA, Sheetikov SA, Zornikova KV, Bogolyubova AV, Rubtsov MA, Kulakovskiy IV, Kuprash DV, Korneev KV, Schwartz AM. rs71327024 Associated with COVID-19 Hospitalization Reduces CXCR6 Promoter Activity in Human CD4 + T Cells via Disruption of c-Myb Binding. Int J Mol Sci 2023; 24:13790. [PMID: 37762093 PMCID: PMC10530726 DOI: 10.3390/ijms241813790] [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: 07/11/2023] [Revised: 08/11/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Single-nucleotide polymorphism rs71327024 located in the human 3p21.31 locus has been associated with an elevated risk of hospitalization upon SARS-CoV-2 infection. The 3p21.31 locus contains several genes encoding chemokine receptors potentially relevant to severe COVID-19. In particular, CXCR6, which is prominently expressed in T lymphocytes, NK, and NKT cells, has been shown to be involved in the recruitment of immune cells to non-lymphoid organs in chronic inflammatory and respiratory diseases. In COVID-19, CXCR6 expression is reduced in lung resident memory T cells from patients with severe disease as compared to the control cohort with moderate symptoms. We demonstrate here that rs71327024 is located within an active enhancer that augments the activity of the CXCR6 promoter in human CD4+ T lymphocytes. The common rs71327024(G) variant makes a functional binding site for the c-Myb transcription factor, while the risk rs71327024(T) variant disrupts c-Myb binding and reduces the enhancer activity. Concordantly, c-Myb knockdown in PMA-treated Jurkat cells negates rs71327024's allele-specific effect on CXCR6 promoter activity. We conclude that a disrupted c-Myb binding site may decrease CXCR6 expression in T helper cells of individuals carrying the minor rs71327024(T) allele and thus may promote the progression of severe COVID-19 and other inflammatory pathologies.
Collapse
Affiliation(s)
- Aksinya N. Uvarova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.M.S.); (A.S.U.); (N.A.M.); (E.A.Z.); (D.V.K.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (S.A.S.); (K.V.Z.); (M.A.R.)
| | - Ekaterina M. Stasevich
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.M.S.); (A.S.U.); (N.A.M.); (E.A.Z.); (D.V.K.)
| | - Alina S. Ustiugova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.M.S.); (A.S.U.); (N.A.M.); (E.A.Z.); (D.V.K.)
| | - Nikita A. Mitkin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.M.S.); (A.S.U.); (N.A.M.); (E.A.Z.); (D.V.K.)
| | - Elina A. Zheremyan
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.M.S.); (A.S.U.); (N.A.M.); (E.A.Z.); (D.V.K.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (S.A.S.); (K.V.Z.); (M.A.R.)
| | - Savely A. Sheetikov
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (S.A.S.); (K.V.Z.); (M.A.R.)
- National Research Center for Hematology, 125167 Moscow, Russia;
| | - Ksenia V. Zornikova
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (S.A.S.); (K.V.Z.); (M.A.R.)
- National Research Center for Hematology, 125167 Moscow, Russia;
| | | | - Mikhail A. Rubtsov
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (S.A.S.); (K.V.Z.); (M.A.R.)
| | | | - Dmitry V. Kuprash
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.M.S.); (A.S.U.); (N.A.M.); (E.A.Z.); (D.V.K.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia; (S.A.S.); (K.V.Z.); (M.A.R.)
| | - Kirill V. Korneev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.M.S.); (A.S.U.); (N.A.M.); (E.A.Z.); (D.V.K.)
- National Research Center for Hematology, 125167 Moscow, Russia;
| | - Anton M. Schwartz
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Abba Khoushy Avenue, Mount Carmel, Haifa 3498838, Israel;
| |
Collapse
|
15
|
Ivanova EN, Shwetar J, Devlin JC, Buus TB, Gray-Gaillard S, Koide A, Cornelius A, Samanovic MI, Herrera A, Mimitou EP, Zhang C, Karmacharya T, Desvignes L, Ødum N, Smibert P, Ulrich RJ, Mulligan MJ, Koide S, Ruggles KV, Herati RS, Koralov SB. mRNA COVID-19 vaccine elicits potent adaptive immune response without the persistent inflammation seen in SARS-CoV-2 infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2021.04.20.21255677. [PMID: 33907755 PMCID: PMC8077568 DOI: 10.1101/2021.04.20.21255677] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SARS-CoV-2 infection and vaccination elicit potent immune responses. Our study presents a comprehensive multimodal single-cell dataset of peripheral blood of patients with acute COVID-19 and of healthy volunteers before and after receiving the SARS-CoV-2 mRNA vaccine and booster. We compared host immune responses to the virus and vaccine using transcriptional profiling, coupled with B/T cell receptor repertoire reconstruction. COVID-19 patients displayed an enhanced interferon signature and cytotoxic gene upregulation, absent in vaccine recipients. These findings were validated in an independent dataset. Analysis of B and T cell repertoires revealed that, while the majority of clonal lymphocytes in COVID-19 patients were effector cells, clonal expansion was more evident among circulating memory cells in vaccine recipients. Furthermore, while clonal αβ T cell responses were observed in both COVID-19 patients and vaccine recipients, dramatic expansion of clonal γδT cells was found only in infected individuals. Our dataset enables comparative analyses of immune responses to infection versus vaccination, including clonal B and T cell responses. Integrating our data with publicly available datasets allowed us to validate our findings in larger cohorts. To our knowledge, this is the first dataset to include comprehensive profiling of longitudinal samples from healthy volunteers pre/post SARS-CoV-2 vaccine and booster.
Collapse
|
16
|
Luo Q, Dwaraka VB, Chen Q, Tong H, Zhu T, Seale K, Raffaele JM, Zheng SC, Mendez TL, Chen Y, Carreras N, Begum S, Mendez K, Voisin S, Eynon N, Lasky-Su JA, Smith R, Teschendorff AE. A meta-analysis of immune-cell fractions at high resolution reveals novel associations with common phenotypes and health outcomes. Genome Med 2023; 15:59. [PMID: 37525279 PMCID: PMC10388560 DOI: 10.1186/s13073-023-01211-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/10/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Changes in cell-type composition of tissues are associated with a wide range of diseases and environmental risk factors and may be causally implicated in disease development and progression. However, these shifts in cell-type fractions are often of a low magnitude, or involve similar cell subtypes, making their reliable identification challenging. DNA methylation profiling in a tissue like blood is a promising approach to discover shifts in cell-type abundance, yet studies have only been performed at a relatively low cellular resolution and in isolation, limiting their power to detect shifts in tissue composition. METHODS Here we derive a DNA methylation reference matrix for 12 immune-cell types in human blood and extensively validate it with flow-cytometric count data and in whole-genome bisulfite sequencing data of sorted cells. Using this reference matrix, we perform a directional Stouffer and fixed effects meta-analysis comprising 23,053 blood samples from 22 different cohorts, to comprehensively map associations between the 12 immune-cell fractions and common phenotypes. In a separate cohort of 4386 blood samples, we assess associations between immune-cell fractions and health outcomes. RESULTS Our meta-analysis reveals many associations of cell-type fractions with age, sex, smoking and obesity, many of which we validate with single-cell RNA sequencing. We discover that naïve and regulatory T-cell subsets are higher in women compared to men, while the reverse is true for monocyte, natural killer, basophil, and eosinophil fractions. Decreased natural killer counts associated with smoking, obesity, and stress levels, while an increased count correlates with exercise and sleep. Analysis of health outcomes revealed that increased naïve CD4 + T-cell and N-cell fractions associated with a reduced risk of all-cause mortality independently of all major epidemiological risk factors and baseline co-morbidity. A machine learning predictor built only with immune-cell fractions achieved a C-index value for all-cause mortality of 0.69 (95%CI 0.67-0.72), which increased to 0.83 (0.80-0.86) upon inclusion of epidemiological risk factors and baseline co-morbidity. CONCLUSIONS This work contributes an extensively validated high-resolution DNAm reference matrix for blood, which is made freely available, and uses it to generate a comprehensive map of associations between immune-cell fractions and common phenotypes, including health outcomes.
Collapse
Affiliation(s)
- Qi Luo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Varun B Dwaraka
- TruDiagnostics, 881 Corporate Dr., Lexington, KY, 40503, USA
| | - Qingwen Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Huige Tong
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Tianyu Zhu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Kirsten Seale
- Institute for Health and Sport (iHeS), Victoria University, Footscray, VIC, 3011, Australia
| | - Joseph M Raffaele
- PhysioAge LLC, 30 Central Park South / Suite 8A, New York, NY, 10019, USA
| | - Shijie C Zheng
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | - Tavis L Mendez
- TruDiagnostics, 881 Corporate Dr., Lexington, KY, 40503, USA
| | - Yulu Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | | | - Sofina Begum
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Kevin Mendez
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Sarah Voisin
- Institute for Health and Sport (iHeS), Victoria University, Footscray, VIC, 3011, Australia
| | - Nir Eynon
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| | - Ryan Smith
- TruDiagnostics, 881 Corporate Dr., Lexington, KY, 40503, USA.
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
| |
Collapse
|
17
|
Sweet DR, Freeman ML, Zidar DA. Immunohematologic Biomarkers in COVID-19: Insights into Pathogenesis, Prognosis, and Prevention. Pathog Immun 2023; 8:17-50. [PMID: 37427016 PMCID: PMC10324469 DOI: 10.20411/pai.v8i1.572] [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: 02/24/2023] [Accepted: 05/24/2023] [Indexed: 07/11/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has had profound effects on the health of individuals and on healthcare systems worldwide. While healthcare workers on the frontlines have fought to quell multiple waves of infection, the efforts of the larger research community have changed the arch of this pandemic as well. This review will focus on biomarker discovery and other efforts to identify features that predict outcomes, and in so doing, identify possible effector and passenger mechanisms of adverse outcomes. Identifying measurable soluble factors, cell-types, and clinical parameters that predict a patient's disease course will have a legacy for the study of immunologic responses, especially stimuli, which induce an overactive, yet ineffectual immune system. As prognostic biomarkers were identified, some have served to represent pathways of therapeutic interest in clinical trials. The pandemic conditions have created urgency for accelerated target identification and validation. Collectively, these COVID-19 studies of biomarkers, disease outcomes, and therapeutic efficacy have revealed that immunologic systems and responses to stimuli are more heterogeneous than previously assumed. Understanding the genetic and acquired features that mediate divergent immunologic outcomes in response to this global exposure is ongoing and will ultimately improve our preparedness for future pandemics, as well as impact preventive approaches to other immunologic diseases.
Collapse
Affiliation(s)
- David R. Sweet
- Case Western Reserve University School of Medicine, Cleveland, OH
| | - Michael L. Freeman
- Division of Infectious Diseases and HIV Medicine, Case Western Reserve University, Cleveland, OH
| | - David A. Zidar
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH
- Cardiology Section, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH
- Center for Global Health and Diseases, Department of Pathology, Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
18
|
Vazquez-Alejo E, Tarancon-Diez L, Espinar-Buitrago MDLS, Genebat M, Calderón A, Pérez-Cabeza G, Magro-Lopez E, Leal M, Muñoz-Fernández MÁ. Persistent Exhausted T-Cell Immunity after Severe COVID-19: 6-Month Evaluation in a Prospective Observational Study. J Clin Med 2023; 12:jcm12103539. [PMID: 37240647 DOI: 10.3390/jcm12103539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/03/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
INTRODUCTION Severe COVID-19 can result in a significant and irreversible impact on long-term recovery and subsequent immune protection. Understanding the complex immune reactions may be useful for establishing clinically relevant monitoring. METHODS Hospitalized adults with SARS-CoV-2 between March/October 2020 (n = 64) were selected. Cryopreserved peripheral blood mononuclear cells (PBMCs) and plasma samples were obtained at hospitalization (baseline) and 6 months after recovery. Immunological components' phenotyping and SARS-CoV-2-specific T-cell response were studied in PBMCs by flow cytometry. Up to 25 plasma pro/anti-inflammatory cytokines/chemokines were assessed by LEGENDplex immunoassays. The SARS-CoV-2 group was compared to matched healthy donors. RESULTS Biochemical altered parameters during infection were normalized at a follow-up time point in the SARS-CoV-2 group. Most of the cytokine/chemokine levels were increased at baseline in the SARS-CoV-2 group. This group showed increased Natural Killer cells (NK) activation and decreased CD16high NK subset, which normalized six months later. They also presented a higher intermediate and patrolling monocyte proportion at baseline. T cells showed an increased terminally differentiated (TemRA) and effector memory (EM) subsets distribution in the SARS-CoV-2 group at baseline and continued to increase six months later. Interestingly, T-cell activation (CD38) in this group decreased at the follow-up time point, contrary to exhaustion markers (TIM3/PD1). In addition, we observed the highest SARS-CoV-2-specific T-cell magnitude response in TemRA CD4 T-cell and EM CD8 T-cell subsets at the six-months time point. CONCLUSIONS The immunological activation in the SARS-CoV-2 group during hospitalization is reversed at the follow-up time point. However, the marked exhaustion pattern remains over time. This dysregulation could constitute a risk factor for reinfection and the development of other pathologies. Additionally, high SARS-CoV-2-specific T-cells response levels appear to be associated with infection severity.
Collapse
Affiliation(s)
- Elena Vazquez-Alejo
- Immunology Section, Molecular Immuno-Biology Laboratory, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Tarancon-Diez
- Immunology Section, Molecular Immuno-Biology Laboratory, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria de la Sierra Espinar-Buitrago
- Immunology Section, Molecular Immuno-Biology Laboratory, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel Genebat
- Internal Medicine Department, Hospital Fátima, 41012 Sevilla, Spain
| | - Alba Calderón
- Internal Medicine Department, Hospital Fátima, 41012 Sevilla, Spain
| | | | - Esmeralda Magro-Lopez
- Immunology Section, Molecular Immuno-Biology Laboratory, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manuel Leal
- Internal Medicine Department, Hospital Viamed Santa Ángela de la Cruz, 41013 Sevilla, Spain
| | - Mª Ángeles Muñoz-Fernández
- Immunology Section, Molecular Immuno-Biology Laboratory, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
19
|
De Rose DU, Pace PG, Ceccherini-Silberstein F, Dotta A, Andreoni M, Sarmati L, Iannetta M. T Lymphocyte Subset Counts and Interferon-Gamma Production in Adults and Children with COVID-19: A Narrative Review. J Pers Med 2023; 13:jpm13050755. [PMID: 37240926 DOI: 10.3390/jpm13050755] [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: 03/22/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Adults and children exhibit a broad range of clinical outcomes from SARS-CoV-2 infection, with minimal to mild symptoms, especially in the pediatric age. However, some children present with a severe hyperinflammatory post-infectious complication named multisystem inflammatory syndrome in children (MIS-C), mainly affecting previously healthy subjects. Understanding these differences is still an ongoing challenge, that can lead to new therapeutic strategies and avoid unfavorable outcomes. In this review, we discuss the different roles of T lymphocyte subsets and interferon-γ (IFN-γ) in the immune responses of adults and children. Lymphopenia can influence these responses and represent a good predictor for the outcome, as reported by most authors. The increased IFN-γ response exhibited by children could be the starting point for the activation of a broad response that leads to MIS-C, with a significantly higher risk than in adults, although a single IFN signature has not been identified. Multicenter studies with large cohorts in both age groups are still needed to study SARS-CoV-2 pathogenesis with new tools and to understand how is possible to better modulate immune responses.
Collapse
Affiliation(s)
- Domenico Umberto De Rose
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, 00165 Rome, Italy
- PhD Course in Microbiology, Immunology, Infectious Diseases, and Transplants (MIMIT), Faculty of Medicine and Surgery, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Pier Giorgio Pace
- Infectious Disease Unit, Department of System Medicine, "Tor Vergata" University and Hospital, 00133 Rome, Italy
| | | | - Andrea Dotta
- Neonatal Intensive Care Unit, "Bambino Gesù" Children's Hospital IRCCS, 00165 Rome, Italy
| | - Massimo Andreoni
- Infectious Disease Unit, Department of System Medicine, "Tor Vergata" University and Hospital, 00133 Rome, Italy
| | - Loredana Sarmati
- Infectious Disease Unit, Department of System Medicine, "Tor Vergata" University and Hospital, 00133 Rome, Italy
| | - Marco Iannetta
- Infectious Disease Unit, Department of System Medicine, "Tor Vergata" University and Hospital, 00133 Rome, Italy
| |
Collapse
|
20
|
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
|
21
|
Al-Akioui-Sanz K, Pascual-Miguel B, Díaz-Almirón M, Mestre-Durán C, Navarro-Zapata A, Clares-Villa L, Martín-Cortázar C, Vicario JL, Moreno MÁ, Balas A, De Paz R, Minguillón J, Pérez-Martínez A, Ferreras C. Donor selection for adoptive cell therapy with CD45RA - memory T cells for patients with coronavirus disease 2019, and dexamethasone and interleukin-15 effects on the phenotype, proliferation and interferon gamma release. Cytotherapy 2023; 25:330-340. [PMID: 36585293 PMCID: PMC9742221 DOI: 10.1016/j.jcyt.2022.12.001] [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: 06/13/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AIMS We have previously demonstrated the safety and feasibility of adoptive cell therapy with CD45RA- memory T cells containing severe acute respiratory syndrome coronavirus 2-specific T cells for patients with coronavirus disease 2019 from an unvaccinated donor who was chosen based on human leukocyte antigen compatibility and cellular response. In this study, we examined the durability of cellular and humoral immunity within CD45RA- memory T cells and the effect of dexamethasone, the current standard of care treatment, and interleukin-15, a cytokine critically involved in T-cell maintenance and survival. METHODS We performed a longitudinal analysis from previously severe acute respiratory syndrome coronavirus 2-infected and infection-naïve individuals covering 21 months from infection and 10 months after full vaccination with the BNT162b2 Pfizer/BioNTech vaccine. RESULTS We observed that cellular responses are maintained over time. Humoral responses increased after vaccination but were gradually lost. In addition, dexamethasone did not alter cell functionality or proliferation of CD45RA- T cells, and interleukin-15 increased the memory T-cell activation state, regulatory T cell expression, and interferon gamma release. CONCLUSIONS Our results suggest that the best donors for adoptive cell therapy would be recovered individuals and 2 months after vaccination, although further studies with larger cohorts would be needed to confirm this finding. Dexamethasone did not affect the characteristics of the memory T cells at a concentration used in the clinical practice and IL-15 showed a positive effect on SARS-CoV-2-specific CD45RA- T cells.
Collapse
Affiliation(s)
- Karima Al-Akioui-Sanz
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Bárbara Pascual-Miguel
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | | | - Carmen Mestre-Durán
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Alfonso Navarro-Zapata
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Laura Clares-Villa
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Carla Martín-Cortázar
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - José Luis Vicario
- Histocompatibility Unit, Transfusion Center of Madrid, Madrid, Spain
| | | | - Antonio Balas
- Histocompatibility Unit, Transfusion Center of Madrid, Madrid, Spain
| | - Raquel De Paz
- Cell Therapy Unit, Hematology Department, La Paz University Hospital, Madrid, Spain
| | - Jordi Minguillón
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain
| | - Antonio Pérez-Martínez
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain; Pediatric Hemato-oncology Department, La Paz University Hospital, Madrid, Spain; Faculty of Medicine Autonomous, University of Madrid, Madrid, Spain
| | - Cristina Ferreras
- IdiPAZ, Hospital La Paz Institute for Health Research, La Paz University Hospital, Madrid, Spain.
| |
Collapse
|
22
|
Lu H, Liu Z, Deng X, Chen S, Zhou R, Zhao R, Parandaman R, Thind A, Henley J, Tian L, Yu J, Comai L, Feng P, Yuan W. Potent NKT cell ligands overcome SARS-CoV-2 immune evasion to mitigate viral pathogenesis in mouse models. PLoS Pathog 2023; 19:e1011240. [PMID: 36961850 PMCID: PMC10128965 DOI: 10.1371/journal.ppat.1011240] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 04/25/2023] [Accepted: 02/24/2023] [Indexed: 03/25/2023] Open
Abstract
One of the major pathogenesis mechanisms of SARS-CoV-2 is its potent suppression of innate immunity, including blocking the production of type I interferons. However, it is unknown whether and how the virus interacts with different innate-like T cells, including NKT, MAIT and γδ T cells. Here we reported that upon SARS-CoV-2 infection, invariant NKT (iNKT) cells rapidly trafficked to infected lung tissues from the periphery. We discovered that the envelope (E) protein of SARS-CoV-2 efficiently down-regulated the cell surface expression of the antigen-presenting molecule, CD1d, to suppress the function of iNKT cells. E protein is a small membrane protein and a viroporin that plays important roles in virion packaging and envelopment during viral morphogenesis. We showed that the transmembrane domain of E protein was responsible for suppressing CD1d expression by specifically reducing the level of mature, post-ER forms of CD1d, suggesting that it suppressed the trafficking of CD1d proteins and led to their degradation. Point mutations demonstrated that the putative ion channel function was required for suppression of CD1d expression and inhibition of the ion channel function using small chemicals rescued the CD1d expression. Importantly, we discovered that among seven human coronaviruses, only E proteins from highly pathogenic coronaviruses including SARS-CoV-2, SARS-CoV and MERS suppressed CD1d expression, whereas the E proteins of human common cold coronaviruses, HCoV-OC43, HCoV-229E, HCoV-NL63 and HCoV-HKU1, did not. These results suggested that E protein-mediated evasion of NKT cell function was likely an important pathogenesis factor, enhancing the virulence of these highly pathogenic coronaviruses. Remarkably, activation of iNKT cells with their glycolipid ligands, both prophylactically and therapeutically, overcame the putative viral immune evasion, significantly mitigated viral pathogenesis and improved host survival in mice. Our results suggested a novel NKT cell-based anti-SARS-CoV-2 therapeutic approach.
Collapse
Affiliation(s)
- Hongjia Lu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Graduate Programs in Biomedical and Biological Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Zhewei Liu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Xiangxue Deng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Siyang Chen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Ruiting Zhou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Rongqi Zhao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Ramya Parandaman
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Amarjot Thind
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Jill Henley
- The Hastings and Wright Laboratories, Keck School of Medicine, University Southern California, California, United States of America
| | - Lei Tian
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California, United States of America
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California, United States of America
| | - Lucio Comai
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- The Hastings and Wright Laboratories, Keck School of Medicine, University Southern California, California, United States of America
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
23
|
Walter LO, Cardoso CC, Santos-Pirath ÍM, Costa HZ, Gartner R, Werle I, Mohr ETB, da Rosa JS, Lubschinski TL, Felisberto M, Kretzer IF, Masukawa II, Vanny PDA, Luiz MC, de Moraes ACR, Dalmarco EM, Santos-Silva MC. T cell maturation is significantly affected by SARS-CoV-2 infection. Immunology 2023. [PMID: 36855300 DOI: 10.1111/imm.13635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a respiratory tract infection caused by the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). An adequate T cell response is essential not only for fighting disease but also for the creation of immune memory. Thus, the present study aims to evaluate the T cells of patients with moderate, severe and critical COVID-19 not only at the time of illness but also 2 months after diagnosis to observe whether changes in this compartment persist. In this study, 166 COVID-19 patients were stratified into moderate/severe and critical disease categories. The maturation and activation of T cells were evaluated through flow cytometry. In addition, Treg cells were analysed. Until 15 days after diagnosis, patients presented a reduction in absolute and relative T lymphocyte counts. After 2 months, in moderate/severe patients, the counts returned to a similar level as that of the control group. In convalescent patients who had a critical illness, absolute T lymphocyte values increased considerably. Patients with active disease did not show differentiation of T cells. Nonetheless, after 2 months, patients with critical COVID-19 showed a significant increase in CD4+ EMRA (CD45RA+ effector memory) T lymphocytes. Furthermore, COVID-19 patients showed delayed T cell activation and reduced CD8+ suppressor T cells even 2 months after diagnosis. A reduction in CD4+ Treg cells was also observed, and their numbers returned to a similar level as that of healthy controls in convalescent patients. The results demonstrate that COVID-19 patients have a delayed activation and differentiation of T cells. In addition, these patients have a great reduction of T cells with a suppressor phenotype.
Collapse
Affiliation(s)
- Laura Otto Walter
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Chandra Chiappin Cardoso
- Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Íris Mattos Santos-Pirath
- Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Heloisa Zorzi Costa
- Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Rafaela Gartner
- Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Isabel Werle
- Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Julia Salvan da Rosa
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Mariano Felisberto
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Iara Fabricia Kretzer
- Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Ivete Ioshiko Masukawa
- Infectious Disease Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil.,Infectious Disease Service, Nereu Ramos Hospital, State Health Department, Florianópolis, Brazil
| | - Patrícia de Almeida Vanny
- Infectious Disease Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Magali Chaves Luiz
- Infectious Disease Service, Nereu Ramos Hospital, State Health Department, Florianópolis, Brazil
| | - Ana Carolina Rabello de Moraes
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil.,Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Eduardo Monguilhott Dalmarco
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil.,Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil
| | - Maria Cláudia Santos-Silva
- Postgraduate Program in Pharmacy, Federal University of Santa Catarina, Florianópolis, Brazil.,Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina, Florianópolis, Brazil.,Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
24
|
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: 22] [Impact Index Per Article: 11.0] [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
|
25
|
von Borstel A, Nguyen TH, Rowntree LC, Ashhurst TM, Allen LF, Howson LJ, Holmes NE, Smibert OC, Trubiano JA, Gordon CL, Cheng AC, Kent SJ, Rossjohn J, Kedzierska K, Davey MS. Circulating effector γδ T cell populations are associated with acute coronavirus disease 19 in unvaccinated individuals. Immunol Cell Biol 2023; 101:321-332. [PMID: 36698330 DOI: 10.1111/imcb.12623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/16/2022] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes severe coronavirus disease 2019 (COVID-19) in a small proportion of infected individuals. The immune system plays an important role in the defense against SARS-CoV-2, but our understanding of the cellular immune parameters that contribute to severe COVID-19 disease is incomplete. Here, we show that populations of effector γδ T cells are associated with COVID-19 in unvaccinated patients with acute disease. We found that circulating CD27neg CD45RA+ CX3CR1+ Vδ1effector cells expressing Granzymes (Gzms) were enriched in COVID-19 patients with acute disease. Moreover, higher frequencies of GzmB+ Vδ2+ T cells were observed in acute COVID-19 patients. SARS-CoV-2 infection did not alter the γδ T cell receptor repertoire of either Vδ1+ or Vδ2+ subsets. Our work demonstrates an association between effector populations of γδ T cells and acute COVID-19 in unvaccinated individuals.
Collapse
Affiliation(s)
- Anouk von Borstel
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Thi Ho Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Thomas M Ashhurst
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, Centenary Institute and University of Sydney, Sydney, NSW, Australia.,Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia
| | - Lilith F Allen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Lauren J Howson
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Natasha E Holmes
- Department of Infectious Diseases, Austin Health, Heidelberg, VIC, Australia.,Department of Critical Care, University of Melbourne, Parkville, VIC, Australia.,Data Analytics Research and Evaluation (DARE) Centre, Austin Health and University of Melbourne, Heidelberg, VIC, Australia.,Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC, Australia
| | - Olivia C Smibert
- Department of Infectious Diseases, Austin Health, Heidelberg, VIC, Australia.,Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jason A Trubiano
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC, Australia.,Department of Medicine (Austin Health), University of Melbourne, Heidelberg, VIC, Australia
| | - Claire L Gordon
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,Department of Infectious Diseases, Austin Health, Heidelberg, VIC, Australia
| | - Allen C Cheng
- Infection Prevention and Healthcare Epidemiology Unit, Alfred Health, Melbourne, VIC, Australia.,School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC, Australia.,Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Martin S Davey
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
26
|
Nateghi S, Hesari E, Mansouri F, Akrami M, Pourian M, Khosravani A, Taghizadeh F, Akbarpour S, Faraji N. Evaluation of Blood Biochemistry and Cardiopulmonary status of Hospitalized Covid-19 Patients in 3 Months Post Discharged Follow up Survey. Clin Med Insights Circ Respir Pulm Med 2022; 16:11795484221119332. [PMID: 36579139 PMCID: PMC9791284 DOI: 10.1177/11795484221119332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/29/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The new coronavirus causes systemic inflammation and damage to various organs. So, this study aimed to follow up patients with COVID -19 after recovery for three months by performing cardiac, pulmonary and laboratory tests. MATERIALS AND METHODS This was a prospective cohort study of COVID-19 patients who were discharged from Baharloo Hospital. The diagnosis of COVID-19 was confirmed by PCR or long CT scan. Inclusion criteria were age over 18 years and patients with more than 50% of pulmonary involvement in lung CT scan. The patients were called to Baharloo hospital three months after recovery and were examined for cardiac, pulmonary and blood tests. RESULT Our study included 178 participants with mean age of 55.70, and 50.6% of them were male. Among pulmonary factors in the hospital, 71.9%, 15.7%,1.7%,19.1% and 53.4% had positive GGO, consolidation, reverse halo sign, traction bronchiectasis and vascular enlargement, respectively. After three months follow up, percentage of patients who had reticulation, honeycombing, fibrotic brand and bullae were 12%, 1.1%,8.4% and 0%, respectively. The Mean values of FVC and FEV1 were reported 4.21 and 3.01, respectively. Among Cardiac factors, positive PVC, PAC and mean Pap were decreased after three months. Only Growth in myalgia and Decreased sense of taste were statistically significant. Also, D dimer, UA protein, PMN, Ferritin CRP, PMN, LDH and HB amounts had decreased significantly. CONCLUSION Our study indicated that in addition to pulmonary changes, rapid damage to other organs and the occurrence of cardiac symptoms and changes in laboratory result were also reported in patients recovered from COVID-19.
Collapse
Affiliation(s)
- Saeed Nateghi
- Department of Cardiology, Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Elahe Hesari
- School of public health, Department of Epidemiology and Biostatistics, Tehran University of Medical Science, Tehran, Iran
| | - Fariba Mansouri
- Department of Respiratory, Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Majid Akrami
- Department of Anesthesiology, Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Mandana Pourian
- Radiology, Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Arezoo Khosravani
- Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran
| | | | - Samaneh Akbarpour
- Occupational Sleep Research Center, Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Neda Faraji
- Department of Internist, Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran,Neda Faraji, Assistant professor of Internal Diseases, Department of Internist, Baharloo Hospital, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
27
|
Atmeh PA, Gay L, Levasseur A, La Scola B, Olive D, Mezouar S, Gorvel JP, Mege JL. Macrophages and γδ T cells interplay during SARS-CoV-2 variants infection. Front Immunol 2022; 13:1078741. [PMID: 36601113 PMCID: PMC9806226 DOI: 10.3389/fimmu.2022.1078741] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction The emergence of several SARS-CoV-2 variants during the COVID pandemic has revealed the impact of variant diversity on viral infectivity and host immune responses. While antibodies and CD8 T cells are essential to clear viral infection, the protective role of innate immunity including macrophages has been recognized. The aims of our study were to compare the infectivity of different SARS-CoV-2 variants in monocyte-derived macrophages (MDM) and to assess their activation profiles and the role of ACE2 (Angiotensin-converting enzyme 2), the main SARS-CoV-2 receptor. We also studied the ability of macrophages infected to affect other immune cells such as γδ2 T cells, another partner of innate immune response to viral infections. Results We showed that the SARS-CoV-2 variants α-B.1.1.7 (United Kingdom), β-B.1.351 (South Africa), γ-P.1 (Brazil), δ-B.1.617 (India) and B.1.1.529 (Omicron), infected MDM without replication, the γ-Brazil variant exhibiting increased infectivity for MDM. No clear polarization profile of SARS-CoV-2 variants-infected MDM was observed. The β-B.1.351 (South Africa) variant induced macrophage activation while B.1.1.529 (Omicron) was rather inhibitory. We observed that SARS-CoV-2 variants modulated ACE2 expression in MDM. In particular, the β-B.1.351 (South Africa) variant induced a higher expression of ACE2, related to MDM activation. Finally, all variants were able to activate γδ2 cells among which γ-P.1 (Brazil) and β-B.1.351 (South Africa) variants were the most efficient. Conclusion Our data show that SARS-CoV-2 variants can infect MDM and modulate their activation, which was correlated with the ACE2 expression. They also affect γδ2 T cell activation. The macrophage response to SARS-CoV-2 variants was stereotypical.
Collapse
Affiliation(s)
- Perla Abou Atmeh
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Laetitia Gay
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Anthony Levasseur
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Bernard La Scola
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Daniel Olive
- Institut Paoli-Calmettes; Aix-Marseille Univ, UM105, Centre National de la Recherche Scientifique (CNRS) UMR 7258, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
| | - Jean-Pierre Gorvel
- Aix-Marseille Univ, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d’Immunologie de Marseille Luminy (CIML), Marseille, France
| | - Jean-Louis Mege
- Aix-Marseille Univ, Institut de Recherche pour le Développement (IRD), Assistance Publique Hopitaux de Marseille (APHM), Microbe Evolution, Phylogeny and Infection (MEPHI), Marseille, France
- Institut Hospitalo-Universitaire (IHU)-Méditerranée Infection, Marseille, France
- Aix-Marseille Univ, Assitance Publique Hopitaux de Marseille (APHM), Hôpital de la Conception, Laboratoire d’Immunologie, Marseille, France
| |
Collapse
|
28
|
Fears AC, Walker EM, Chirichella N, Slisarenko N, Merino KM, Golden N, Picou B, Spencer S, Russell-Lodrigue KE, Doyle-Meyers LA, Blair RV, Beddingfield BJ, Maness NJ, Roy CJ, Rout N. The dynamics of γδ T cell responses in nonhuman primates during SARS-CoV-2 infection. Commun Biol 2022; 5:1380. [PMID: 36526890 PMCID: PMC9756695 DOI: 10.1038/s42003-022-04310-y] [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: 06/24/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Although most SARS-CoV-2 infections are mild, some patients develop systemic inflammation and progress to acute respiratory distress syndrome (ARDS). However, the cellular mechanisms underlying this spectrum of disease remain unclear. γδT cells are T lymphocyte subsets that have key roles in systemic and mucosal immune responses during infection and inflammation. Here we show that peripheral γδT cells are rapidly activated following aerosol or intra-tracheal/intra-nasal (IT/IN) SARS-CoV-2 infection in nonhuman primates. Our results demonstrate a rapid expansion of Vδ1 γδT cells at day1 that correlate significantly with lung viral loads during the first week of infection. Furthermore, increase in levels of CCR6 and Granzyme B expression in Vδ1 T cells during viral clearance imply a role in innate-like epithelial barrier-protective and cytotoxic functions. Importantly, the early activation and mobilization of circulating HLA-DR+CXCR3+ γδT cells along with significant correlations of Vδ1 T cells with IL-1Ra and SCF levels in bronchoalveolar lavage suggest a novel role for Vδ1 T cells in regulating lung inflammation during aerosol SARS-CoV-2 infection. A deeper understanding of the immunoregulatory functions of MHC-unrestricted Vδ1 T cells in lungs during early SARS-CoV-2 infection is particularly important in the wake of emerging new variants with increased transmissibility and immune evasion potential.
Collapse
Affiliation(s)
- Alyssa C Fears
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Edith M Walker
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Nicole Chirichella
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Nadia Slisarenko
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Kristen M Merino
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Nadia Golden
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Breanna Picou
- High Containment Research Performance Core, Tulane National Primate Research Center, Covington, LA, USA
| | - Skye Spencer
- High Containment Research Performance Core, Tulane National Primate Research Center, Covington, LA, USA
| | - Kasi E Russell-Lodrigue
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, USA
| | - Lara A Doyle-Meyers
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, USA
| | - Robert V Blair
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | | | - Nicholas J Maness
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane School of Medicine, New Orleans, LA, USA
| | - Chad J Roy
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane School of Medicine, New Orleans, LA, USA
| | - Namita Rout
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA.
- Department of Microbiology and Immunology, Tulane School of Medicine, New Orleans, LA, USA.
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
29
|
Di Simone M, Corsale AM, Lo Presti E, Scichilone N, Picone C, Giannitrapani L, Dieli F, Meraviglia S. Phenotypical and Functional Alteration of γδ T Lymphocytes in COVID-19 Patients: Reversal by Statins. Cells 2022; 11:3449. [PMID: 36359845 PMCID: PMC9656060 DOI: 10.3390/cells11213449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
(1) Background: statins have been considered an attractive class of drugs in the pharmacological setting of COVID-19 due to their pleiotropic properties and their use correlates with decreased mortality in hospitalized COVID-19 patients. Furthermore, it is well known that statins, which block the mevalonate pathway, affect γδ T lymphocyte activation. As γδ T cells participate in the inflammatory process of COVID-19, we have investigated the therapeutical potential of statins as a tool to inhibit γδ T cell pro-inflammatory activities; (2) Methods: we harvested peripheral blood mononuclear cells (PBMCs) from COVID-19 patients with mild clinical manifestations, COVID-19 recovered patients, and healthy controls. We performed ex vivo flow cytometry analysis to study γδ T cell frequency, phenotype, and exhaustion status. PBMCs were treated with Atorvastatin followed by non-specific and specific stimulation, to evaluate the expression of pro-inflammatory cytokines; (3) Results: COVID-19 patients had a lower frequency of circulating Vδ2+ T lymphocytes but showed a pronounced pro-inflammatory profile, which was inhibited by in vitro treatment with statins; (4) Conclusions: the in vitro capacity of statins to inhibit Vδ2+ T lymphocytes in COVID-19 patients highlights a new potential biological function of these drugs and supports their therapeutical use in these patients.
Collapse
Affiliation(s)
- Marta Di Simone
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AUOP Paolo Giaccone, 90127 Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy
| | - Anna Maria Corsale
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AUOP Paolo Giaccone, 90127 Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy
| | - Elena Lo Presti
- National Research Council (CNR), Institute for Biomedical Research and Innovation (IRIB), 90146 Palermo, Italy
| | - Nicola Scichilone
- Division of Respiratory Medicine, AUOP Paolo Giaccone, 90127 Palermo, Italy
- Internal Medicine Department Unit, Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialities Department (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Carmela Picone
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AUOP Paolo Giaccone, 90127 Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy
| | - Lydia Giannitrapani
- National Research Council (CNR), Institute for Biomedical Research and Innovation (IRIB), 90146 Palermo, Italy
- Internal Medicine Department Unit, Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialities Department (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AUOP Paolo Giaccone, 90127 Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AUOP Paolo Giaccone, 90127 Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
30
|
Bekbossynova M, Akhmaltdinova L, Dossybayeva K, Tauekelova A, Smagulova Z, Tsechoeva T, Turebayeva G, Sailybayeva A, Kalila Z, Mirashirova T, Muratov T, Poddighe D. Central and effector memory T cells in peripheral blood of patients with interstitial pneumonia: preliminary clues from a COVID-19 study. Respir Res 2022; 23:278. [PMID: 36217141 PMCID: PMC9549841 DOI: 10.1186/s12931-022-02190-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND SARS-CoV-2 pre-existing T-cell immune reactivity can be present in some people. A general perturbation of the main peripheral lymphocyte subsets has been described in severe COVID-19 patients, but very few studies assessed the general memory T-cell homeostasis in the acute phase of COVID-19. Here, we performed a general analysis of the main memory T cell populations in the peripheral blood of patients admitted to the hospital for a confirmed or probable COVID-19 diagnosis. METHODS In this cross-sectional study, adult patients (aged ≥ 18 years) needing hospital admission for respiratory disease due to confirmed or probable COVID-19, were recruited before starting the therapeutic protocol for this disease. In addition to the assessment of the general lymphocyte subpopulations in the early phase of COVID-19, central memory T cells (Tmcentr cells: CD45RO+CCR7+) and effector memory T cells (Tmeff cells: CD45RO+CCR7-) were assessed by multi-color flow cytometry, in comparison to a control group. RESULTS During the study period, 148 study participants were recruited. Among them, 58 patients turned out positive for SARS-CoV-2 PCR (including both patients with interstitial pneumonia [PCR+Pn+] and without this complication [PCR+Pn-]), whereas the remaining 90 patients resulted to be SARS-CoV-2 PCR negative, even though all were affected with interstitial pneumonia [PCR-Pn+]. Additionally, 28 control patients without any ongoing respiratory disease were recruited. A clear unbalance in the T memory compartment emerged from this analysis on the whole pool of T cells (CD3+ cells), showing a significant increase in Tmcentr cells and, conversely, a significant decrease in Tmeff cells in both pneumonia groups (PCR+Pn+ and PCR-Pn+) compared to the controls; PCR+Pn- group showed trends comprised between patients with pneumonia (from one side) and the control group (from the other side). This perturbation inside the memory T cell compartment was also observed in the individual analysis of the four main T cell subpopulations, based upon the differential expression of CD4 and/or CD8 markers. CONCLUSION Overall, we observed both absolute and relative increases of Tmcentr cells and decrease of Tmeff cells in patients affected with interstitial pneumonia (regardless of the positive or negative results of SARS-CoV-2 PCR), compared to controls. These results need confirmation from additional research, in order to consider this finding as a potential biological marker of interstitial lung involvement in patients affected with viral respiratory infections.
Collapse
Affiliation(s)
| | | | - Kuanysh Dossybayeva
- National Research Cardiac Surgery Center, 010000, Nur-Sultan, Kazakhstan
- Nazarbayev University School of Medicine (NUSOM), Kerei-Zhanibek Str. 5/1, 010000, Nur-Sultan, Kazakhstan
| | - Ainur Tauekelova
- National Research Cardiac Surgery Center, 010000, Nur-Sultan, Kazakhstan
| | - Zauresh Smagulova
- City Infectious Disease Center at Multidisciplinary Medical Center, 010000, Nur-Sultan, Kazakhstan
| | - Tatyana Tsechoeva
- City Infectious Disease Center at Multidisciplinary Medical Center, 010000, Nur-Sultan, Kazakhstan
| | - Gulsimzhan Turebayeva
- City Infectious Disease Center at Multidisciplinary Medical Center, 010000, Nur-Sultan, Kazakhstan
| | - Aliya Sailybayeva
- National Research Cardiac Surgery Center, 010000, Nur-Sultan, Kazakhstan
| | - Zhanar Kalila
- National Research Cardiac Surgery Center, 010000, Nur-Sultan, Kazakhstan
| | | | - Timur Muratov
- Department of Public Health of Nur‑Sultan City, 010000, Nur-Sultan, Kazakhstan
| | - Dimitri Poddighe
- Nazarbayev University School of Medicine (NUSOM), Kerei-Zhanibek Str. 5/1, 010000, Nur-Sultan, Kazakhstan.
- University Medical Center (UMC), 010000, Nur-Sultan, Kazakhstan.
| |
Collapse
|
31
|
Kudryavtsev IV, Arsentieva NA, Korobova ZR, Isakov DV, Rubinstein AA, Batsunov OK, Khamitova IV, Kuznetsova RN, Savin TV, Akisheva TV, Stanevich OV, Lebedeva AA, Vorobyov EA, Vorobyova SV, Kulikov AN, Sharapova MA, Pevtsov DE, Totolian AA. Heterogenous CD8+ T Cell Maturation and 'Polarization' in Acute and Convalescent COVID-19 Patients. Viruses 2022; 14:1906. [PMID: 36146713 PMCID: PMC9504186 DOI: 10.3390/v14091906] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The adaptive antiviral immune response requires interaction between CD8+ T cells, dendritic cells, and Th1 cells for controlling SARS-CoV-2 infection, but the data regarding the role of CD8+ T cells in the acute phase of COVID-19 and post-COVID-19 syndrome are still limited. METHODS . Peripheral blood samples collected from patients with acute COVID-19 (n = 71), convalescent subjects bearing serum SARS-CoV-2 N-protein-specific IgG antibodies (n = 51), and healthy volunteers with no detectable antibodies to any SARS-CoV-2 proteins (HC, n = 46) were analyzed using 10-color flow cytometry. RESULTS Patients with acute COVID-19 vs. HC and COVID-19 convalescents showed decreased absolute numbers of CD8+ T cells, whereas the frequency of CM and TEMRA CD8+ T cells in acute COVID-19 vs. HC was elevated. COVID-19 convalescents vs. HC had increased naïve and CM cells, whereas TEMRA cells were decreased compared to HC. Cell-surface CD57 was highly expressed by the majority of CD8+ T cells subsets during acute COVID-19, but convalescents had increased CD57 on 'naïve', CM, EM4, and pE1 2-3 months post-symptom onset. CXCR5 expression was altered in acute and convalescent COVID-19 subjects, whereas the frequencies of CXCR3+ and CCR4+ cells were decreased in both patient groups vs. HC. COVID-19 convalescents had increased CCR6-expressing CD8+ T cells. Moreover, CXCR3+CCR6- Tc1 cells were decreased in patients with acute COVID-19 and COVID-19 convalescents, whereas Tc2 and Tc17 levels were increased compared to HC. Finally, IL-27 negatively correlated with the CCR6+ cells in acute COVID-19 patients. CONCLUSIONS We described an abnormal CD8+ T cell profile in COVID-19 convalescents, which resulted in lower frequencies of effector subsets (TEMRA and Tc1), higher senescent state (upregulated CD57 on 'naïve' and memory cells), and higher frequencies of CD8+ T cell subsets expressing lung tissue and mucosal tissue homing molecules (Tc2, Tc17, and Tc17.1). Thus, our data indicate that COVID-19 can impact the long-term CD8+ T cell immune response.
Collapse
Affiliation(s)
- Igor V. Kudryavtsev
- Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Natalia A. Arsentieva
- Laboratory of Immunology, Saint Petersburg Pasteur Institute, Mira 14, 197101 Saint Petersburg, Russia
| | - Zoia R. Korobova
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Laboratory of Immunology, Saint Petersburg Pasteur Institute, Mira 14, 197101 Saint Petersburg, Russia
| | - Dmitry V. Isakov
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Artem A. Rubinstein
- Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
| | - Oleg K. Batsunov
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Laboratory of Immunology, Saint Petersburg Pasteur Institute, Mira 14, 197101 Saint Petersburg, Russia
| | - Irina V. Khamitova
- Laboratory of Immunology, Saint Petersburg Pasteur Institute, Mira 14, 197101 Saint Petersburg, Russia
| | - Raisa N. Kuznetsova
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Laboratory of Immunology, Saint Petersburg Pasteur Institute, Mira 14, 197101 Saint Petersburg, Russia
| | - Tikhon V. Savin
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Laboratory of Immunology, Saint Petersburg Pasteur Institute, Mira 14, 197101 Saint Petersburg, Russia
| | - Tatiana V. Akisheva
- Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
| | - Oksana V. Stanevich
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Smorodintsev Research Institute of Influenza, Prof. Popov St. 15/17, 197376 Saint Petersburg, Russia
| | - Aleksandra A. Lebedeva
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Evgeny A. Vorobyov
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Snejana V. Vorobyova
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Alexander N. Kulikov
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Maria A. Sharapova
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Dmitrii E. Pevtsov
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Areg A. Totolian
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Laboratory of Immunology, Saint Petersburg Pasteur Institute, Mira 14, 197101 Saint Petersburg, Russia
| |
Collapse
|
32
|
Sanz M, Mann BT, Chitrakar A, Soriano-Sarabia N. Defying convention in the time of COVID-19: Insights into the role of γδ T cells. Front Immunol 2022; 13:819574. [PMID: 36032159 PMCID: PMC9403327 DOI: 10.3389/fimmu.2022.819574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 is a complex disease which immune response can be more or less potent. In severe cases, patients might experience a cytokine storm that compromises their vital functions and impedes clearance of the infection. Gamma delta (γδ) T lymphocytes have a critical role initiating innate immunity and shaping adaptive immune responses, and they are recognized for their contribution to tumor surveillance, fighting infectious diseases, and autoimmunity. γδ T cells exist as both circulating T lymphocytes and as resident cells in different mucosal tissues, including the lungs and their critical role in other respiratory viral infections has been demonstrated. In the context of SARS-CoV-2 infection, γδ T cell responses are understudied. This review summarizes the findings on the antiviral role of γδ T cells in COVID-19, providing insight into how they may contribute to the control of infection in the mild/moderate clinical outcome.
Collapse
|
33
|
Tarique M, Suhail M, Naz H, Muhammad N, Tabrez S, Zughaibi TA, Abuzenadah AM, Hashem AM, Shankar H, Saini C, Sharma A. Where do T cell subsets stand in SARS-CoV-2 infection: an update. Front Cell Infect Microbiol 2022; 12:964265. [PMID: 36034704 PMCID: PMC9399648 DOI: 10.3389/fcimb.2022.964265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/12/2022] [Indexed: 01/08/2023] Open
Abstract
An outbreak of coronavirus disease 2019 (COVID-19) emerged in China in December 2019 and spread so rapidly all around the globe. It's continued and spreading more dangerously in India and Brazil with higher mortality rate. Understanding of the pathophysiology of COVID-19 depends on unraveling of interactional mechanism of SARS-CoV-2 and human immune response. The immune response is a complex process, which can be better understood by understanding the immunological response and pathological mechanisms of COVID-19, which will provide new treatments, increase treatment efficacy, and decrease mortality associated with the disease. In this review we present a amalgamate viewpoint based on the current available knowledge on COVID-19 which includes entry of the virus and multiplication of virus, its pathological effects on the cellular level, immunological reaction, systemic and organ presentation. T cells play a crucial role in controlling and clearing viral infections. Several studies have now shown that the severity of the COVID-19 disease is inversely correlated with the magnitude of the T cell response. Understanding SARS-CoV-2 T cell responses is of high interest because T cells are attractive vaccine targets and could help reduce COVID-19 severity. Even though there is a significant amount of literature regarding SARS-CoV-2, there are still very few studies focused on understanding the T cell response to this novel virus. Nevertheless, a majority of these studies focused on peripheral blood CD4+ and CD8+ T cells that were specific for viruses. The focus of this review is on different subtypes of T cell responses in COVID-19 patients, Th17, follicular helper T (TFH), regulatory T (Treg) cells, and less classical, invariant T cell populations, such as δγ T cells and mucosal-associated invariant T (MAIT) cells etc that could influence disease outcome.
Collapse
Affiliation(s)
- Mohammad Tarique
- Department of Child Health, University of Missouri, Columbia, MO, United States
| | - Mohd Suhail
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huma Naz
- Department of Child Health, University of Missouri, Columbia, MO, United States
| | - Naoshad Muhammad
- Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, United States
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Torki A. Zughaibi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adel M. Abuzenadah
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar M. Hashem
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hari Shankar
- India Council of Medical Research, New Delhi, India
| | - Chaman Saini
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
34
|
Du B, Guo Y, Li G, Zhu Y, Wang Y, Xi X. Non-structure protein ORF1ab (NSP8) in SARS-CoV-2 contains potential γδT cell epitopes. Front Microbiol 2022; 13:936272. [PMID: 35935236 PMCID: PMC9354780 DOI: 10.3389/fmicb.2022.936272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/30/2022] [Indexed: 12/05/2022] Open
Abstract
Upon activation by the pathogen through T-cell receptors (TCRs), γδT cells suppress the pathogenic replication and thus play important roles against viral infections. Targeting SARS-CoV-2 via γδT cells provides alternative therapeutic strategies. However, little is known about the recognition of SARS-CoV-2 antigens by γδT cells. We discovered a specific Vγ9/δ2 CDR3 by analyzing γδT cells derived from the patients infected by SARS-CoV-2. Using a cell model exogenously expressing γδ-TCR established, we further screened the structural motifs within the CDR3 responsible for binding to γδ-TCR. Importantly, these sequences were mapped to NSP8, a non-structural protein in SARS-CoV-2. Our results suggest that NSP8 mediates the recognition by γδT cells and thus could serve as a potential target for vaccines.
Collapse
Affiliation(s)
- Boyu Du
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
- Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yang Guo
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Gang Li
- Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yunhe Zhu
- Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yunfu Wang
- Institute of Neuroscience, Hubei University of Medicine, Shiyan, China
- *Correspondence: Yunfu Wang,
| | - Xueyan Xi
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
- Renmin Hospital, Hubei University of Medicine, Shiyan, China
- Xueyan Xi,
| |
Collapse
|
35
|
Gay L, Mezouar S, Cano C, Frohna P, Madakamutil L, Mège JL, Olive D. Role of Vγ9vδ2 T lymphocytes in infectious diseases. Front Immunol 2022; 13:928441. [PMID: 35924233 PMCID: PMC9340263 DOI: 10.3389/fimmu.2022.928441] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/27/2022] [Indexed: 12/22/2022] Open
Abstract
The T cell receptor Vγ9Vδ2 T cells bridge innate and adaptive antimicrobial immunity in primates. These Vγ9Vδ2 T cells respond to phosphoantigens (pAgs) present in microbial or eukaryotic cells in a butyrophilin 3A1 (BTN3) and butyrophilin 2A1 (BTN2A1) dependent manner. In humans, the rapid expansion of circulating Vγ9Vδ2 T lymphocytes during several infections as well as their localization at the site of active disease demonstrates their important role in the immune response to infection. However, Vγ9Vδ2 T cell deficiencies have been observed in some infectious diseases such as active tuberculosis and chronic viral infections. In this review, we are providing an overview of the mechanisms of Vγ9Vδ2 T cell-mediated antimicrobial immunity. These cells kill infected cells mainly by releasing lytic mediators and pro-inflammatory cytokines and inducing target cell apoptosis. In addition, the release of chemokines and cytokines allows the recruitment and activation of immune cells, promoting the initiation of the adaptive immune response. Finaly, we also describe potential new therapeutic tools of Vγ9Vδ2 T cell-based immunotherapy that could be applied to emerging infections.
Collapse
Affiliation(s)
- Laetitia Gay
- Aix-Marseille Univ, Intitut Recherche pour le Développement (IRT), Assistance Publique Hôpitaux de Marseille (APHM), Microbe, Evolution, Phylogeny, Infection (MEPHI), Marseille, France
- Immunology Department, IHU-Méditerranée Infection, Marseille, France
- ImCheck Therapeutics, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille Univ, Intitut Recherche pour le Développement (IRT), Assistance Publique Hôpitaux de Marseille (APHM), Microbe, Evolution, Phylogeny, Infection (MEPHI), Marseille, France
- Immunology Department, IHU-Méditerranée Infection, Marseille, France
| | | | | | | | - Jean-Louis Mège
- Aix-Marseille Univ, Intitut Recherche pour le Développement (IRT), Assistance Publique Hôpitaux de Marseille (APHM), Microbe, Evolution, Phylogeny, Infection (MEPHI), Marseille, France
- Immunology Department, IHU-Méditerranée Infection, Marseille, France
- Aix-Marseille Univ, APHM, Hôpital de la Conception, Laboratoire d’Immunologie, Marseille, France
| | - Daniel Olive
- Centre pour la Recherche sur le Cancer de Marseille (CRCM), Inserm UMR1068, Centre national de la recherche scientifique (CNRS) UMR7258, Institut Paoli Calmettes, Marseille, France
| |
Collapse
|
36
|
Singh K, Cogan S, Elekes S, Murphy DM, Cummins S, Curran R, Najda Z, Dunne MR, Jameson G, Gargan S, Martin S, Long A, Doherty DG. SARS-CoV-2 spike and nucleocapsid proteins fail to activate human dendritic cells or γδ T cells. PLoS One 2022; 17:e0271463. [PMID: 35834480 PMCID: PMC9282473 DOI: 10.1371/journal.pone.0271463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/30/2022] [Indexed: 11/20/2022] Open
Abstract
γδ T cells are thought to contribute to immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but the mechanisms by which they are activated by the virus are unknown. Using flow cytometry, we investigated if the two most abundant viral structural proteins, spike and nucleocapsid, can activate human γδ T cell subsets, directly or in the presence of dendritic cells (DC). Both proteins failed to induce interferon-γ production by Vδ1 or Vδ2 T cells within fresh mononuclear cells or lines of expanded γδ T cells generated from healthy donors, but the same proteins stimulated CD3+ cells from COVID-19 patients. The nucleocapsid protein stimulated interleukin-12 production by DC and downstream interferon-γ production by co-cultured Vδ1 and Vδ2 T cells, but protease digestion and use of an alternative nucleocapsid preparation indicated that this activity was due to contaminating non-protein material. Thus, SARS-CoV-2 spike and nucleocapsid proteins do not have stimulatory activity for DC or γδ T cells. We propose that γδ T cell activation in COVID-19 patients is mediated by immune recognition of viral RNA or other structural proteins by γδ T cells, or by other immune cells, such as DC, that produce γδ T cell-stimulatory ligands or cytokines.
Collapse
Affiliation(s)
- Kiran Singh
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Sita Cogan
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Stefan Elekes
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Dearbhla M. Murphy
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Sinead Cummins
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Rory Curran
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Zaneta Najda
- Molecular Cell Biology Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Margaret R. Dunne
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Gráinne Jameson
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Siobhan Gargan
- Discipline of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Seamus Martin
- Molecular Cell Biology Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Aideen Long
- Discipline of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Derek G. Doherty
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| |
Collapse
|
37
|
Vazquez-Alejo E, Tarancon-Diez L, Carrasco I, Vigil-Vázquez S, Muñoz-Chapuli M, Rincón-López E, Saavedra-Lozano J, Santos-Sebastián M, Aguilera-Alonso D, Hernanz-Lobo A, Santiago-García B, de León-Luis JA, Muñoz P, Sánchez-Luna M, Navarro ML, Muñoz-Fernández MÁ. SARS-CoV2 Infection During Pregnancy Causes Persistent Immune Abnormalities in Women Without Affecting the Newborns. Front Immunol 2022; 13:947549. [PMID: 35911743 PMCID: PMC9330630 DOI: 10.3389/fimmu.2022.947549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/20/2022] [Indexed: 12/21/2022] Open
Abstract
SARS-CoV2 infection in pregnancy and exposed newborns is poorly known. We performed a longitudinal analysis of immune system and determined soluble cytokine levels in pregnant women infected with SARS-CoV2 and in their newborns. Women with confirmed SARS-CoV2 infection and their exposed uninfected newborns were recruited from Hospital General Universitario Gregorio Marañón. Peripheral blood mononuclear cells (PBMCs), cord cells and plasma were collected at birth and 6 months later. Immunophenotyping of natural killer (NK), monocytes and CD4/CD8 T-cells were studied in cryopreserved PBMCs and cord cells by multiparametric flow cytometry. Up to 4 soluble pro/anti-inflammatory cytokines were assessed in plasma/cord plasma by ELISA assay. SARS-CoV2-infected mothers and their newborns were compared to matched healthy non-SARS-CoV2-infected mothers and their newborns. The TNFα and IL-10 levels of infected mothers were higher at baseline than those of healthy controls. Infected mothers showed increased NK cells activation and reduced expression of maturation markers that reverted after 6 months. They also had high levels of Central Memory and low Effector Memory CD4-T cell subsets. Additionally, the increased CD4- and CD8-T cell activation (CD154 and CD38) and exhaustion (TIM3/TIGIT) levels at baseline compared to controls remained elevated after 6 months. Regarding Treg cells, the levels were lower at infected mothers at baseline but reverted after 6 months. No newborn was infected at birth. The lower levels of monocytes, NK and CD4-T cells observed at SARS-CoV2-exposed newborns compared to unexposed controls significantly increased 6 months later. In conclusion, SARS-CoV2 infection during pregnancy shows differences in immunological components that could lead newborns to future clinical implications after birth. However, SARS-CoV2 exposed 6-months-old newborns showed no immune misbalance, whereas the infected mothers maintain increased activation and exhaustion levels in T-cells after 6 months.
Collapse
Affiliation(s)
- Elena Vazquez-Alejo
- Immunology Section, Laboratory of ImmunoBiology Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), HIV-HGM BioBank, Madrid, Spain
| | - Laura Tarancon-Diez
- Immunology Section, Laboratory of ImmunoBiology Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), HIV-HGM BioBank, Madrid, Spain
| | - Itzíar Carrasco
- Infectious Diseases in Paediatric Population, Gregorio Marañón Research Institute (IiSGM) and University Hospital, Madrid, Spain
- CIBER of Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - Sara Vigil-Vázquez
- Department of Neonatology, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
| | - Mar Muñoz-Chapuli
- Department of Obstetrics and Gynecology, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
| | - Elena Rincón-López
- Infectious Diseases in Paediatric Population, Gregorio Marañón Research Institute (IiSGM) and University Hospital, Madrid, Spain
- Infectious Diseases Section, Department of Paediatrics, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
- CIBER of Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - Jesús Saavedra-Lozano
- Infectious Diseases in Paediatric Population, Gregorio Marañón Research Institute (IiSGM) and University Hospital, Madrid, Spain
- Infectious Diseases Section, Department of Paediatrics, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
- Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- CIBER of Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - Mar Santos-Sebastián
- Infectious Diseases in Paediatric Population, Gregorio Marañón Research Institute (IiSGM) and University Hospital, Madrid, Spain
- Infectious Diseases Section, Department of Paediatrics, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
- CIBER of Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - David Aguilera-Alonso
- Infectious Diseases in Paediatric Population, Gregorio Marañón Research Institute (IiSGM) and University Hospital, Madrid, Spain
- Infectious Diseases Section, Department of Paediatrics, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
- CIBER of Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - Alicia Hernanz-Lobo
- Infectious Diseases in Paediatric Population, Gregorio Marañón Research Institute (IiSGM) and University Hospital, Madrid, Spain
- Infectious Diseases Section, Department of Paediatrics, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
- CIBER of Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - Begoña Santiago-García
- Infectious Diseases in Paediatric Population, Gregorio Marañón Research Institute (IiSGM) and University Hospital, Madrid, Spain
- Infectious Diseases Section, Department of Paediatrics, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
- CIBER of Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - Juan Antonio de León-Luis
- Department of Obstetrics and Gynecology, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
- Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón (HGUGM), CIBER Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Manuel Sánchez-Luna
- Department of Neonatology, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
- Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - María Luisa Navarro
- Infectious Diseases in Paediatric Population, Gregorio Marañón Research Institute (IiSGM) and University Hospital, Madrid, Spain
- Infectious Diseases Section, Department of Paediatrics, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
- Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- CIBER of Infectious Diseases (CIBERINFEC), Madrid, Spain
| | - Mª Ángeles Muñoz-Fernández
- Immunology Section, Laboratory of ImmunoBiology Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), HIV-HGM BioBank, Madrid, Spain
| |
Collapse
|
38
|
Wang D, Cao K, Shen X, Zhang B, Chen M, Yu W. Clinical Characteristics and Immune Status of Patients with Severe Fever with Thrombocytopenia Syndrome. Viral Immunol 2022; 35:465-473. [PMID: 35675657 DOI: 10.1089/vim.2021.0217] [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: 11/12/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is a novel infectious disease caused by bunya virus. The purpose of this study was to investigate the clinical characteristics of SFTS patients and their virus-related immune disorders in vivo. Patients with SFTS admitted to Nanjing Drum Tower Hospital from 2017 to 2020 were retrospectively analyzed, and divided into survival group and death group according to the 28-day survival. Clinical characteristics and laboratory examination results of SFTS patients were recorded, and dynamic changes of immune function and inflammatory factors were statistically analyzed. Prolonged activated prothrombin time (APTT) (p = 0.001), high viral load (p = 0.001), and elevated human leukocyte antigen DR (HLA-DR) level (p = 0.002) were independent prognostic risk factors for SFTS patients. Compared to the survival group, the nonsurvival group was more prone to hemorrhagic and neurological symptoms (p < 0.05). Natural kill (NK) cell count, interleukin-10, interferon-α, and tumor necrosis factor-α scores in the nonsurvival group continued to increase after admission, while CD3+ T, CD4+ T, and CD8+ T cell counts continued to decrease. CD3+ T lymphocyte count was negatively correlated with viral load (R = 0.3883, p < 0.001), CD4+ T lymphocyte count was negatively correlated with viral load (R = 0.28933, p < 0.001), CD8+ T lymphocyte count was negatively correlated with viral load (R = 0.781, p < 0.001), and HLA-DR was positively correlated with viral load (R = 0.489, p < 0.001). High viral load, prolonged APTT time, and elevated HLA-DR level are independent prognostic risk factors for SFTS patients. The T lymphocyte subsets of SFTS patients continue to decrease after infection, and the number of T lymphocyte subsets can reflect the severity of the disease.
Collapse
Affiliation(s)
- Dacheng Wang
- Department of Intensive Care, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Ke Cao
- Department of Intensive Care, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Xiaofei Shen
- Department of General Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Beiyuan Zhang
- Department of Intensive Care, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Ming Chen
- Department of Intensive Care, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Wenkui Yu
- Department of Intensive Care, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
39
|
Cimini E, Agrati C. γδ T Cells in Emerging Viral Infection: An Overview. Viruses 2022; 14:v14061166. [PMID: 35746638 PMCID: PMC9230790 DOI: 10.3390/v14061166] [Citation(s) in RCA: 3] [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: 04/20/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 02/05/2023] Open
Abstract
New emerging viruses belonging to the Coronaviridae, Flaviviridae, and Filoviridae families are serious threats to public health and represent a global concern. The surveillance to monitor the emergence of new viruses and their transmission is an important target for public health authorities. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is an excellent example of a pathogen able to cause a pandemic. In a few months, SARS-CoV-2 has spread globally from China, and it has become a world health problem. Gammadelta (γδ) T cell are sentinels of innate immunity and are able to protect the host from viral infections. They enrich many tissues, such as the skin, intestines, and lungs where they can sense and fight the microbes, thus contributing to the protective immune response. γδ T cells perform their direct antiviral activity by cytolytic and non-cytolytic mechanisms against a wide range of viruses, and they are able to orchestrate the cellular interplay between innate and acquired immunity. For their pleiotropic features, γδ T cells have been proposed as a target for immunotherapies in both cancer and viral infections. In this review, we analyzed the role of γδ T cells in emerging viral infections to define the profile of the response and to better depict their role in the host protection.
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
|
SARS-CoV-2 Envelope (E) Protein Binds and Activates TLR2 Pathway: A Novel Molecular Target for COVID-19 Interventions. Viruses 2022; 14:v14050999. [PMID: 35632741 PMCID: PMC9146335 DOI: 10.3390/v14050999] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
This paper presents a molecular characterization of the interaction between the SARS-CoV-2 envelope (E) protein and TLR2. We demonstrated that the E protein, both as a recombinant soluble protein and as a native membrane protein associated with SARS-CoV-2 viral particles, interacts physically with the TLR2 receptor in a specific and dose-dependent manner. Furthermore, we showed that the specific interaction with the TLR2 pathway activates the NF-κB transcription factor and stimulates the production of the CXCL8 inflammatory chemokine. In agreement with the importance of NF-κB in the TLR signaling pathway, we showed that the chemical inhibition of this transcription factor leads to significant inhibition of CXCL8 production, while the blockade of the P38 and ERK1/2 MAP kinases only results in partial CXCL8 inhibition. Overall, our findings propose the envelope (E) protein as a novel molecular target for COVID-19 interventions: either (i) by exploring the therapeutic effect of anti-E blocking/neutralizing antibodies in symptomatic COVID-19 patients, or (ii) as a promising non-spike SARS-CoV-2 antigen candidate for inclusion in the development of next-generation prophylactic vaccines against COVID-19 infection and disease.
Collapse
|
42
|
Tyurin AV, Salimgareeva MK, Miniakhmetov IR, Khusainova RI, Samorodov A, Pavlov VN, Kzhyshkowska J. Correlation of the Imbalance in the Circulating Lymphocyte Subsets With C-Reactive Protein and Cardio-Metabolic Conditions in Patients With COVID-19. Front Immunol 2022; 13:856883. [PMID: 35603207 PMCID: PMC9120577 DOI: 10.3389/fimmu.2022.856883] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/08/2022] [Indexed: 12/04/2022] Open
Abstract
The immune system is severely compromised in patients with COVID-19. The representative group of 43 patients were selected from the cohort of 342 patients with COVID-19 and pneumonia. This group of 43 patients was examined for the levels of C-reactive protein, biomarker of systemic inflammation, and for the subsets of adaptive immune cells. The immunological parameters were correlated with the metabolic parameters and cardiovascular pathology history. We identified that a decrease in the absolute number of T-lymphocytes, T-cytotoxic, T-activated and B-lymphocytes correlated with the higher levels of CRP. The absolute number of T-helpers and the absolute number of double positive T-lymphocytes positively correlated with the levels of iron in serum (Z= 0,310 and Z=0,394). The absolute numbers of T-activated lymphocytes positively correlated with serum levels of LDH (Z = 0,422), ferritin (Z = 0,407) and iron (Z = 0,418). When studying subpopulations of lymphocytes, depending on the combined pathology, we found that the absolute numbers of B-lymphocytes and double positive T-lymphocytes in the peripheral blood were significantly reduced in patients with arterial hypertension (p=0,0074 and p=0,0227, correspondingly). The increased levels of NK cell were found in patients with a history of coronary heart disease (p=0,0108). In addition, we found that deficiencies in the adaptive immune system correlated with the deficiencies in iron metabolism. The cardiovascular pathology upsets the balance in the adaptive and innate immune system in the circulation of patient with severe COVID-19.
Collapse
Affiliation(s)
- Anton V. Tyurin
- Internal Medicine Department, Bashkir State Medical University, Ufa, Russia
| | | | - Ildar R. Miniakhmetov
- Republic Medical Genetic Center, Ufa, Russia
- Medical Genetics Department, Bashkir State Medical University, Ufa, Russia
| | - Rita I. Khusainova
- Republic Medical Genetic Center, Ufa, Russia
- Medical Genetics Department, Bashkir State Medical University, Ufa, Russia
| | - Alexandr Samorodov
- Department of Pharmacology, Bashkir State Medical University, Ufa, Russia
| | | | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg – Hessen, Mannheim, Germany
- Laboratory for Translational Cell and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
- *Correspondence: Julia Kzhyshkowska,
| |
Collapse
|
43
|
Jedicke N, Stankov MV, Cossmann A, Dopfer‐Jablonka A, Knuth C, Ahrenstorf G, Ramos GM, Behrens GMN. Humoral immune response following prime and boost BNT162b2 vaccination in people living with HIV on antiretroviral therapy. HIV Med 2022; 23:558-563. [PMID: 34725907 PMCID: PMC8652991 DOI: 10.1111/hiv.13202] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/27/2021] [Accepted: 10/18/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVES People living with HIV (PLWH) with low CD4 T-cell counts may be at a higher risk for severe coronavirus disease 2019 (COVID-19) outcomes and in need of efficient vaccination. The World Health Organization (WHO) now recommends prioritizing PLHIV for COVID-19 vaccination. Data on immune responses after messenger RNA (mRNA) vaccination in PLHIV in relation to CD4 counts are scarce. We aimed at assessing the humoral immune response in PLHIV after mRNA vaccination against COVID-19. METHODS We examined a cohort of PLHIV after prime (n = 88) and boost (n = 52) vaccination with BNT162b2. We assessed levels of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein-specific immunoglobulin G (IgG)/IgA and circulating neutralizing antibodies in plasma and correlated results to the cellular immune status. BNT162b2-vaccinated health care workers served as controls. RESULTS All PLWH had a viral load of ≤ 200 HIV-1 RNA copies/mL and 96.5% had a viral load of < 50 copies/mL. Anti-S IgG and neutralizing antibody responses after BNT162b2 priming were significantly lower in PLHIV having a CD4:CD8 T-cell ratio of < 0.5. However, we observed robust humoral immunity in the majority of PLWH receiving antiretroviral therapy (ART) irrespective of CD4 T-cell nadir, current CD4 count or CD4:CD8 ratio after full BNT162b2 vaccination. Nevertheless, HIV-negative controls produced significantly higher mean anti-S IgG concentrations with less variability. CONCLUSIONS The majority of PLWH mounted robust responses after complete BNT162b2 vaccination but overall amounts of antibodies directed against the SARS-CoV-2 receptor-binding domain were variable. The impact on clinical efficacy remains unclear.
Collapse
Affiliation(s)
- Nils Jedicke
- Department of Gastroenterology, Hepatology and EndocrinologyHannover Medical SchoolHannoverGermany
| | - Metodi V. Stankov
- Department of Rheumatology and ImmunologyHannover Medical SchoolHannoverGermany
| | - Anne Cossmann
- Department of Rheumatology and ImmunologyHannover Medical SchoolHannoverGermany
| | - Alexandra Dopfer‐Jablonka
- Department of Gastroenterology, Hepatology and EndocrinologyHannover Medical SchoolHannoverGermany
- German Center for Infection Research (DZIF)Partner Site Hannover‐BraunschweigBraunschweigGermany
| | - Christine Knuth
- Department of Rheumatology and ImmunologyHannover Medical SchoolHannoverGermany
| | - Gerrit Ahrenstorf
- Department of Rheumatology and ImmunologyHannover Medical SchoolHannoverGermany
| | - Gema Morillas Ramos
- Department of Rheumatology and ImmunologyHannover Medical SchoolHannoverGermany
| | - Georg M. N. Behrens
- Department of Rheumatology and ImmunologyHannover Medical SchoolHannoverGermany
- German Center for Infection Research (DZIF)Partner Site Hannover‐BraunschweigBraunschweigGermany
- Centre for Individualized Infection Medicine (CiiM)HannoverGermany
| |
Collapse
|
44
|
Torres Rives B, Zúñiga Rosales Y, Mataran Valdés M, Roblejo Balbuena H, Martínez Téllez G, Rodríguez Pérez J, Caridad Marín Padrón L, Rodríguez Pelier C, Sotomayor Lugo F, Valdés Zayas A, Carmenate Portilla T, Sánchez Ramírez B, Carlos Silva Aycaguer L, Portal Miranda JA, Marcheco Teruel B. Assessment of changes in immune status linked to COVID-19 convalescent and its clinical severity in patients and uninfected exposed relatives. Immunobiology 2022; 227:152216. [PMID: 35436751 PMCID: PMC9004226 DOI: 10.1016/j.imbio.2022.152216] [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: 11/04/2021] [Revised: 02/23/2022] [Accepted: 04/09/2022] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The immune response during and after SARS-CoV-2 infection can be complex and heterogeneous, and it can be affected by the severity of the disease. It can also contribute to an unfavorable evolution and bring about short and long term effects. The aim of this study was to characterize the lymphocyte composition according to the severity of COVID-19, as well as its degree of relationship to the specific humoral response to SARS-CoV-2 in convalescents up to 106 days after the infection and in their exposed relatives. METHODS An applied research was carried out with a cross-section analytical design, from March 11 to June 11, 2020 in Cuba. The sample consisted of 251 convalescents from COVID-19 over 18 years of age and 88 exposed controls who did not become ill. The B and T cell subpopulations, including memory T cells, as well as the relationship with the humoral immune response against SARS-CoV-2, were identified by flow cytometry and enzyme immunoassay. RESULTS Convalescent patients, who evolved with severe forms, showed a decrease in frequency and a greater proportion of individuals with values lower than the minimum normal range of B cells, CD3 + CD4 + cells and the CD4 + / CD8 + ratio, as well as a higher frequency and a greater proportion of individuals with values above the normal maximum range of CD3 + CD8 + and NK cells. Convalescent patients with severe forms of COVID-19 that exhibited IgG / RBD titers ≥ 1/200 had a lower frequency of TEMRA CD8 + cells (p = 0.0128) and TEMRA CD4 + (p = 0.0068). IgG / RBD titers were positively correlated with the relative frequency of CD4 + CM T memory cells (r = 0.4352, p = 0.0018). CONCLUSIONS The identified alterations of B and T lymphocytes suggest that convalescent patients with the severe disease could be vulnerable to infectious, autoimmune or autotinflammatory processes; therefore, these individuals need medical follow-up after recovering from the acute disease. Furthermore, the role of T cells CD4 + CM in the production of antibodies against SARS-CoV-2 is confirmed, and it is noted that the defect of memory T cells CD8 + TEMRA could contribute to the development of severe forms of COVID-19.
Collapse
|
45
|
Liu Z, Kilic G, Li W, Bulut O, Gupta MK, Zhang B, Qi C, Peng H, Tsay HC, Soon CF, Mekonnen YA, Ferreira AV, van der Made CI, van Cranenbroek B, Koenen HJPM, Simonetti E, Diavatopoulos D, de Jonge MI, Müller L, Schaal H, Ostermann PN, Cornberg M, Eiz-Vesper B, van de Veerdonk F, van Crevel R, Joosten LAB, Domínguez-Andrés J, Xu CJ, Netea MG, Li Y. Multi-Omics Integration Reveals Only Minor Long-Term Molecular and Functional Sequelae in Immune Cells of Individuals Recovered From COVID-19. Front Immunol 2022; 13:838132. [PMID: 35464396 PMCID: PMC9022455 DOI: 10.3389/fimmu.2022.838132] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
The majority of COVID-19 patients experience mild to moderate disease course and recover within a few weeks. An increasing number of studies characterized the long-term changes in the specific anti-SARS-CoV-2 immune responses, but how COVID-19 shapes the innate and heterologous adaptive immune system after recovery is less well known. To comprehensively investigate the post-SARS-CoV-2 infection sequelae on the immune system, we performed a multi-omics study by integrating single-cell RNA-sequencing, single-cell ATAC-sequencing, genome-wide DNA methylation profiling, and functional validation experiments in 14 convalescent COVID-19 and 15 healthy individuals. We showed that immune responses generally recover without major sequelae after COVID-19. However, subtle differences persist at the transcriptomic level in monocytes, with downregulation of the interferon pathway, while DNA methylation also displays minor changes in convalescent COVID-19 individuals. However, these differences did not affect the cytokine production capacity of PBMCs upon different bacterial, viral, and fungal stimuli, although baseline release of IL-1Ra and IFN-γ was higher in convalescent individuals. In conclusion, we propose that despite minor differences in epigenetic and transcriptional programs, the immune system of convalescent COVID-19 patients largely recovers to the homeostatic level of healthy individuals.
Collapse
Affiliation(s)
- Zhaoli Liu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Gizem Kilic
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Wenchao Li
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Ozlem Bulut
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Manoj Kumar Gupta
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Bowen Zhang
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Cancan Qi
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - He Peng
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Hsin-Chieh Tsay
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Chai Fen Soon
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Yonatan Ayalew Mekonnen
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Anaísa Valido Ferreira
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Caspar I van der Made
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bram van Cranenbroek
- Department of Laboratory Medicine, Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hans J P M Koenen
- Department of Laboratory Medicine, Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Elles Simonetti
- Laboratory of Pediatric Infectious Diseases, Radboud Centre for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Dimitri Diavatopoulos
- Laboratory of Pediatric Infectious Diseases, Radboud Centre for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marien I de Jonge
- Department of Laboratory Medicine, Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Lisa Müller
- Institute of Virology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine University Duesseldorf, Dusseldorf, Germany
| | - Heiner Schaal
- Institute of Virology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine University Duesseldorf, Dusseldorf, Germany
| | - Philipp N Ostermann
- Institute of Virology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine University Duesseldorf, Dusseldorf, Germany
| | - Markus Cornberg
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Frank van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
46
|
Abstract
COVID-19 is a respiratory disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It was first documented in late 2019, but within months, a worldwide pandemic was declared due to the easily transmissible nature of the virus. Research to date on the immune response to SARS-CoV-2 has focused largely on conventional B and T lymphocytes. This review examines the emerging role of unconventional T cell subsets, including γδ T cells, invariant natural killer T (iNKT) cells and mucosal associated invariant T (MAIT) cells in human SARS-CoV-2 infection.Some of these T cell subsets have been shown to play protective roles in anti-viral immunity by suppressing viral replication and opsonising virions of SARS-CoV. Here, we explore whether unconventional T cells play a protective role in SARS-CoV-2 infection as well. Unconventional T cells are already under investigation as cell-based immunotherapies for cancer. We discuss the potential use of these cells as therapeutic agents in the COVID-19 setting. Due to the rapidly evolving situation presented by COVID-19, there is an urgent need to understand the pathogenesis of this disease and the mechanisms underlying its immune response. Through this, we may be able to better help those with severe cases and lower the mortality rate by devising more effective vaccines and novel treatment strategies.
Collapse
Affiliation(s)
- Kristen Orumaa
- Department of Clinical Microbiology and Department of Immunology, Trinity Translational Medicine Institute, St James's Hospital, Dublin 8, Ireland
| | - Margaret R Dunne
- Department of Clinical Microbiology and Department of Immunology, Trinity Translational Medicine Institute, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
47
|
Mortezaee K, Majidpoor J. CD8 + T Cells in SARS-CoV-2 Induced Disease and Cancer-Clinical Perspectives. Front Immunol 2022; 13:864298. [PMID: 35432340 PMCID: PMC9010719 DOI: 10.3389/fimmu.2022.864298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
Dysregulated innate and adaptive immunity is a sign of SARS-CoV-2-induced disease and cancer. CD8+ T cells are important cells of the immune system. The cells belong to the adaptive immunity and take a front-line defense against viral infections and cancer. Extreme CD8+ T-cell activities in the lung of patients with a SARS-CoV-2-induced disease and within the tumor microenvironment (TME) will change their functionality into exhausted state and undergo apoptosis. Such diminished immunity will put cancer cases at a high-risk group for SARS-CoV-2-induced disease, rendering viral sepsis and a more severe condition which will finally cause a higher rate of mortality. Recovering responses from CD8+ T cells is a purpose of vaccination against SARS-CoV-2. The aim of this review is to discuss the CD8+ T cellular state in SARS-CoV-2-induced disease and in cancer and to present some strategies for recovering the functionality of these critical cells.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
48
|
Al-Mterin MA, Alsalman A, Elkord E. Inhibitory Immune Checkpoint Receptors and Ligands as Prognostic Biomarkers in COVID-19 Patients. Front Immunol 2022; 13:870283. [PMID: 35432324 PMCID: PMC9008255 DOI: 10.3389/fimmu.2022.870283] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by SARS-CoV-2. During T-cell activation, the immune system uses different checkpoint pathways to maintain co-inhibitory and co-stimulatory signals. In COVID-19, expression of immune checkpoints (ICs) is one of the most important manifestations, in addition to lymphopenia and inflammatory cytokines, contributing to worse clinical outcomes. There is a controversy whether upregulation of ICs in COVID-19 patients might lead to T-cell exhaustion or activation. This review summarizes the available studies that investigated IC receptors and ligands in COVID-19 patients, as well as their effect on T-cell function. Several IC receptors and ligands, including CTLA-4, BTLA, TIM-3, VISTA, LAG-3, TIGIT, PD-1, CD160, 2B4, NKG2A, Galectin-9, Galectin-3, PD-L1, PD-L2, LSECtin, and CD112, were upregulated in COVID-19 patients. Based on the available studies, there is a possible relationship between disease severity and increased expression of IC receptors and ligands. Overall, the upregulation of some ICs could be used as a prognostic biomarker for disease severity.
Collapse
Affiliation(s)
| | - Alhasan Alsalman
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Eyad Elkord
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, United Kingdom
| |
Collapse
|
49
|
Odak I, Schultze-Florey CR, Hammerschmidt SI, Ritter C, Willenzon S, Friedrichsen M, Ravens I, Sikora R, Bayir LM, Gutierrez Jauregui R, Bernhardt G, Stankov MV, Cossmann A, Hansen G, Krey T, Cornberg M, Koenecke C, Behrens GMN, Bošnjak B, Förster R. Longitudinal Tracking of Immune Responses in COVID-19 Convalescents Reveals Absence of Neutralization Activity Against Omicron and Staggered Impairment to Other SARS-CoV-2 Variants of Concern. Front Immunol 2022; 13:863039. [PMID: 35359969 PMCID: PMC8964088 DOI: 10.3389/fimmu.2022.863039] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Evaluating long-term protection against SARS-CoV-2 variants of concern in convalescing individuals is of high clinical relevance. In this prospective study of a cohort of 46 SARS-CoV-2 patients infected with the Wuhan strain of SARS-CoV-2 we longitudinally analyzed changes in humoral and cellular immunity upon early and late convalescence. Antibody neutralization capacity was measured by surrogate virus neutralization test and cellular responses were investigated with 31-colour spectral flow cytometry. Spike-specific, isotype-switched B cells developed already during the disease phase, showed a memory phenotype and did not decrease in numbers even during late convalescence. Otherwise, no long-lasting perturbations of the immune compartment following COVID-19 clearance were observed. During convalescence anti-Spike (S1) IgG antibodies strongly decreased in all patients. We detected neutralizing antibodies against the Wuhan strain as well as the Alpha and Delta but not against the Beta, Gamma or Omicron variants for up to 7 months post COVID-19. Furthermore, correlation analysis revealed a strong association between sera anti-S1 IgG titers and their neutralization capacity against the Wuhan strain as well as Alpha and Delta. Overall, our data suggest that even 7 month after the clearance of COVID-19 many patients possess a protective layer of immunity, indicated by the persistence of Spike-specific memory B cells and by the presence of neutralizing antibodies against the Alpha and Delta variants. However, lack of neutralizing antibodies against the Beta, Gamma and Omicron variants even during the peak response is of major concern as this indicates viral evasion of the humoral immune response.
Collapse
Affiliation(s)
- Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christian R. Schultze-Florey
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | | | | | | | | - Inga Ravens
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Ruth Sikora
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Lâle M. Bayir
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Günter Bernhardt
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Metodi V. Stankov
- Clinic Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Anne Cossmann
- Clinic Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Guido Hansen
- Institute of Biochemistry, University of Lübeck, Lübeck, Germany
| | - Thomas Krey
- Institute of Biochemistry, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg-Lübeck-Borstel-Riems, Brunswick, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Markus Cornberg
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Sites Hannover-Braunschweig, Brunswick, Germany
- Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Christian Koenecke
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Georg M. N. Behrens
- Clinic Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Sites Hannover-Braunschweig, Brunswick, Germany
- Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
| | - Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Sites Hannover-Braunschweig, Brunswick, Germany
| |
Collapse
|
50
|
Fenizia C, Cetin I, Mileto D, Vanetti C, Saulle I, Di Giminiani M, Saresella M, Parisi F, Trabattoni D, Clerici M, Biasin M, Savasi V. Pregnant Women Develop a Specific Immunological Long-Lived Memory Against SARS-COV-2. Front Immunol 2022; 13:827889. [PMID: 35251011 PMCID: PMC8889908 DOI: 10.3389/fimmu.2022.827889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/24/2022] [Indexed: 12/04/2022] Open
Abstract
It is well established that pregnancy induces deep changes in the immune system. This is part of the physiological adaptation of the female organism to the pregnancy and the immunological tolerance toward the fetus. Indeed, over the three trimesters, the suppressive T regulatory lymphocytes are progressively more represented, while the expression of co-stimulatory molecules decreases overtime. Such adaptations relate to an increased risk of infections and progression to severe disease in pregnant women, potentially resulting in an altered generation of long-lived specific immunological memory of infection contracted during pregnancy. How potent is the immune response against SARS-CoV-2 in infected pregnant women and how long the specific SARS-CoV-2 immunity might last need to be urgently addressed, especially considering the current vaccinal campaign. To address these questions, we analyzed the long-term immunological response upon SARS-CoV-2 infection in pregnant women from delivery to a six-months follow-up. In particular, we investigated the specific antibody production, T cell memory subsets, and inflammation profile. Results show that 80% developed an anti-SARS-CoV-2-specific IgG response, comparable with the general population. While IgG were present only in 50% of the asymptomatic subjects, the antibody production was elicited by infection in all the mild-to-critical patients. The specific T-cell memory subsets rebalanced over-time, and the pro-inflammatory profile triggered by specific SARS-CoV-2 stimulation faded away. These results shed light on SARS-CoV-2-specific immunity in pregnant women; understanding the immunological dynamics of the immune system in response to SARS-CoV-2 is essential for defining proper obstetric management of pregnant women and fine tune gender-specific vaccinal plans.
Collapse
Affiliation(s)
- Claudio Fenizia
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Irene Cetin
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Woman, Mother and Neonate Buzzi Children’s Hospital, ASST Fatebenefratelli‐Sacco, Milan, Italy
| | - Davide Mileto
- Clinical Microbiology, Virology and Bio-emergence Diagnosis, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Claudia Vanetti
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Irma Saulle
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Maria Di Giminiani
- Unit of Obstetrics and Gynecology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | | | - Francesca Parisi
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Unit of Obstetrics and Gynecology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Daria Trabattoni
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Fondazione don Carlo Gnocchi, IRCCS, Milan, Italy
| | - Mara Biasin
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Valeria Savasi
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Unit of Obstetrics and Gynecology, ASST Fatebenefratelli-Sacco, Milan, Italy
| |
Collapse
|