1
|
Zhang Y, Hua J, Chen L. Identifying the plasma metabolome responsible for mediating immune cell action in severe COVID-19: a Mendelian randomization investigation. Front Cell Infect Microbiol 2024; 14:1393432. [PMID: 39224704 PMCID: PMC11366714 DOI: 10.3389/fcimb.2024.1393432] [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: 02/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction The immune response regulates the severity of COVID-19 (sCOVID-19). This study examined the cause-and-effect relationship between immune cell traits (ICTs) and the risk of severe COVID-19. Additionally, we discovered the potential role of plasma metabolome in modulating this risk. Methods Employing data from a genome-wide association study (GWAS), we conducted a two-sample Mendelian randomization (MR) assessment of 731 genetic ICTs and sCOVID-19 (5,101 cases, 1,383,241 controls) incidence. The MR analysis was utilized to further quantitate the degree of plasma metabolome-mediated regulation of immune traits in sCOVID-19. Results The inverse variance weighted method recognized 2 plasma metabolites (PMs) responsible for casual associations between immune cells and sCOVID-19 risk. These included Tridecenedioate (C13:1-DC) which regulated the association between CD27 on IgD- CD38br (OR 0.804, 95% CI 0.699-0.925, p = 0.002) and sCOVID-19 risk (mediated proportion: 18.7%); arginine to citrulline ratio which controlled the relationship of CD39 on monocyte (OR 1.053, 95% CI 1.013-1.094, p = 0.009) with sCOVID-19 risk (mediated proportion: -7.11%). No strong evidence that genetically predicted sCOVID-19 influenced the aforementioned immune traits. Conclusion In this study, we have successfully identified a cause-and-effect relationship between certain ICTs, PMs, and the likelihood of contracting severe COVID-19. Our findings can potentially improve the accuracy of COVID-19 prognostic evaluation and provide valuable insights into the underlying mechanisms of the disease.
Collapse
Affiliation(s)
- Yixia Zhang
- Department of Hematology, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Jie Hua
- Department of Gastroenterology, Jiangsu Province People’s Hospital, Nanjing, China
| | - Liang Chen
- Department of Infectious Diseases, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical College of Nanjing University, Nanjing, China
| |
Collapse
|
2
|
Endo HM, Bandeca SCS, Olchanheski LR, Schemczssen-Graeff Z, Pileggi M. Probiotics and the reduction of SARS-CoV-2 infection through regulation of host cell calcium dynamics. Life Sci 2024; 350:122784. [PMID: 38848939 DOI: 10.1016/j.lfs.2024.122784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/21/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Calcium is a secondary messenger that interacts with several cellular proteins, regulates various physiological processes, and plays a role in diseases such as viral infections. Next-generation probiotics and live biotherapeutic products are linked to the regulation of intracellular calcium levels. Some viruses can manipulate calcium channels, pumps, and membrane receptors to alter calcium influx and promote virion production and release. In this study, we examined the use of bacteria for the prevention and treatment of viral diseases, such as coronavirus of 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Vaccination programs have helped reduce disease severity; however, there is still a lack of well-recognized drug regimens for the clinical management of COVID-19. SARS-CoV-2 interacts with the host cell calcium (Ca2+), manipulates proteins, and disrupts Ca2+ homeostasis. This article explores how viruses exploit, create, or exacerbate calcium imbalances, and the potential role of probiotics in mitigating viral infections by modulating calcium signaling. Pharmacological strategies have been developed to prevent viral replication and block the calcium channels that serve as viral receptors. Alternatively, probiotics may interact with cellular calcium influx, such as Lactobacillus spp. The interaction between Akkermansia muciniphila and cellular calcium homeostasis is evident. A scientific basis for using probiotics to manipulate calcium channel activity needs to be established for the treatment and prevention of viral diseases while maintaining calcium homeostasis. In this review article, we discuss how intracellular calcium signaling can affect viral replication and explore the potential therapeutic benefits of probiotics.
Collapse
Affiliation(s)
- Hugo Massami Endo
- Environmental Microbiology Laboratory, Life Sciences and Health Institute, Structural and Molecular Biology, and Genetics Department, Ponta Grossa State University, Ponta Grossa, Brazil
| | | | - Luiz Ricardo Olchanheski
- Environmental Microbiology Laboratory, Life Sciences and Health Institute, Structural and Molecular Biology, and Genetics Department, Ponta Grossa State University, Ponta Grossa, Brazil
| | - Zelinda Schemczssen-Graeff
- Comparative Immunology Laboratory, Department of Microbiology, Parasitology and Pathology, Federal University of Paraná, Curitiba, Brazil
| | - Marcos Pileggi
- Environmental Microbiology Laboratory, Life Sciences and Health Institute, Structural and Molecular Biology, and Genetics Department, Ponta Grossa State University, Ponta Grossa, Brazil.
| |
Collapse
|
3
|
Zhou D, Zhao S, He K, Liu Q, Zhang F, Pu Z, Xiao L, Zhang L, Chen S, Qian X, Wu X, Shen Y, Yu L, Zhang H, Jin J, Xu M, Wang X, Zhu D, Xie Z, Xu X. Longitudinal dynamic single-cell mass cytometry analysis of peripheral blood mononuclear cells in COVID-19 patients within 6 months after viral RNA clearance. BMC Infect Dis 2024; 24:567. [PMID: 38844850 PMCID: PMC11157885 DOI: 10.1186/s12879-024-09464-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
This study investigates the longitudinal dynamic changes in immune cells in COVID-19 patients over an extended period after recovery, as well as the interplay between immune cells and antibodies. Leveraging single-cell mass spectrometry, we selected six COVID-19 patients and four healthy controls, dissecting the evolving landscape within six months post-viral RNA clearance, alongside the levels of anti-spike protein antibodies. The T cell immunophenotype ascertained via single-cell mass spectrometry underwent validation through flow cytometry in 37 samples. Our findings illuminate that CD8 + T cells, gamma-delta (gd) T cells, and NK cells witnessed an increase, in contrast to the reduction observed in monocytes, B cells, and double-negative T (DNT) cells over time. The proportion of monocytes remained significantly elevated in COVID-19 patients compared to controls even after six-month. Subpopulation-wise, an upsurge manifested within various T effector memory subsets, CD45RA + T effector memory, gdT, and NK cells, whereas declines marked the populations of DNT, naive and memory B cells, and classical as well as non-classical monocytes. Noteworthy associations surfaced between DNT, gdT, CD4 + T, NK cells, and the anti-S antibody titer. This study reveals the changes in peripheral blood mononuclear cells of COVID-19 patients within 6 months after viral RNA clearance and sheds light on the interactions between immune cells and antibodies. The findings from this research contribute to a better understanding of immune transformations during the recovery from COVID-19 and offer guidance for protective measures against reinfection in the context of viral variants.
Collapse
Affiliation(s)
- Diwenxin Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Shuai Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Keting He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Qiuhong Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Fen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Zhangya Pu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Lanlan Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Lingjian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Shangci Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Xiaohan Qian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Xiaoxin Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Yangfan Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Ling Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Huafen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Jiandi Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Min Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Xiaoyan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Zhongyang Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China.
| | - Xiaowei Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China.
| |
Collapse
|
4
|
Felisberto M, Walter LO, Cardoso CC, Santos-Pirath ÍM, Costa HZ, Gartner R, Werle I, Mohr ETB, Salvan da Rosa J, Lubschinski TL, Kretzer IF, Masukawa II, de Almeida Vanny P, Luiz MC, Rabello de Moraes AC, Santos-Silva MC, Dalmarco EM. Lymphocyte B Subtypes in Peripheral Blood: A Prognostic Biomarker for COVID-19 Patients. J Appl Lab Med 2024; 9:456-467. [PMID: 38321537 DOI: 10.1093/jalm/jfad123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND In view of the scientific gap in knowledge of the involvement of the B-cell compartment and clinical prognostic in SARS-CoV-2 infection, this work aims to evaluate the B-cell subsets and the presence of specific IgM and IgG, as well as neutralizing antibodies against SARS-CoV-2, in unvaccinated patients diagnosed with COVID-19. METHODS This study included 133 patients with COVID-19. Cellular components were assessed by flow cytometry, and immunoglobulin levels and reactivity were measured by indirect enzyme-linked immunosorbent assay. RESULTS Our results showed no changes in less differentiated B cells. However, non-switched memory B cells (NS-MBCs) and class-switched memory B cells (CS-MBCs) were reduced in the patients with moderate disease. Also, plasmablasts and double-negative (DN) or "atypical" memory B cells were increased in groups of patients with moderate to critical conditions. In addition, the production of IgM, IgG, and neutralizing antibodies against SARS-CoV-2 demonstrated a positive correlation between the positivity of antibodies against SARS-CoV-2 and disease severity. Besides being related to the development of a more severe course of the disease, the increase in DN B-cell count also contributed to a poorer disease outcome in patients with a higher percentage of these cells. On the other hand, we observed an increase in the absolute number of CS-MBCs in patients with greater chances of survival. CONCLUSIONS This study demonstrates that the B-cell compartment may contribute to the development of clinical symptoms of COVID-19, with changes in B-cell subset counts linked to disease course and patient prognosis.
Collapse
Affiliation(s)
- Mariano Felisberto
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Laura Otto Walter
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Chandra Chiappin Cardoso
- Clinical Analysis Department, Flow Cytometry Service, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Íris Mattos Santos-Pirath
- Clinical Analysis Department, Flow Cytometry Service, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Heloisa Zorzi Costa
- Clinical Analysis Department, Flow Cytometry Service, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Rafaela Gartner
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Isabel Werle
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Eduarda Talita Bramorski Mohr
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Julia Salvan da Rosa
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Tainá Larissa Lubschinski
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Iara Fabricia Kretzer
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Ivete Ioshiko Masukawa
- Infectious Disease Service, University Hospital-Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Infectious Disease Service, State Health Department, Hospital Nereu Ramos, Florianópolis, SC, Brazil
| | - Patrícia de Almeida Vanny
- Infectious Disease Service, University Hospital-Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Magali Chaves Luiz
- Infectious Disease Service, State Health Department, Hospital Nereu Ramos, Florianópolis, SC, Brazil
| | - Ana Carolina Rabello de Moraes
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Maria Claudia Santos-Silva
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Flow Cytometry Service, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Eduardo Monguilhott Dalmarco
- Postgraduate Program in Pharmacy, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Clinical Analysis Department, Health Sciences Center, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| |
Collapse
|
5
|
Suthar MS. Durability of immune responses to SARS-CoV-2 infection and vaccination. Semin Immunol 2024; 73:101884. [PMID: 38861769 PMCID: PMC11490408 DOI: 10.1016/j.smim.2024.101884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024]
Abstract
Infection with SARS-CoV-2 in humans has caused a pandemic of unprecedented dimensions. SARS-CoV-2 is primarily transmitted through respiratory droplets and targets ciliated epithelial cells in the nasal cavity, trachea, and lungs by utilizing the cellular receptor angiotensin-converting enzyme 2 (ACE2). The innate immune response, including type I and III interferons, inflammatory cytokines (IL-6, TNF-α, IL-1β), innate immune cells (monocytes, DCs, neutrophils, natural killer cells), antibodies (IgG, sIgA, neutralizing antibodies), and adaptive immune cells (B cells, CD8+ and CD4+ T cells) play pivotal roles in mitigating COVID-19 disease. Broad and durable B-cell- and T-cell immunity elicited by infection and vaccination is essential for protection against severe disease, hospitalization and death. However, the emergence of SARS-CoV-2 variants that evade neutralizing antibodies continue to jeopardize vaccine efficacy. In this review, we highlight our understanding the infection- and vaccine-mediated humoral, B and T cell responses, the durability of the immune responses, and how variants continue to threaten the efficacy of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Emory Vaccine Center, Emory University, Atlanta, GA, USA; Emory Center of Excellence of Influenza Research and Response (CEIRR), Atlanta, GA, USA; Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA; Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
6
|
De Biasi S, Lo Tartaro D, Neroni A, Rau M, Paschalidis N, Borella R, Santacroce E, Paolini A, Gibellini L, Ciobanu AL, Cuccorese M, Trenti T, Rubio I, Vitetta F, Cardi M, Argüello RJ, Ferraro D, Cossarizza A. Immunosenescence and vaccine efficacy revealed by immunometabolic analysis of SARS-CoV-2-specific cells in multiple sclerosis patients. Nat Commun 2024; 15:2752. [PMID: 38553477 PMCID: PMC10980723 DOI: 10.1038/s41467-024-47013-0] [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: 11/13/2023] [Accepted: 03/11/2024] [Indexed: 04/02/2024] Open
Abstract
Disease-modifying therapies (DMT) administered to patients with multiple sclerosis (MS) can influence immune responses to SARS-CoV-2 and vaccine efficacy. However, data on the detailed phenotypic, functional and metabolic characteristics of antigen (Ag)-specific cells following the third dose of mRNA vaccine remain scarce. Here, using flow cytometry and 45-parameter mass cytometry, we broadly investigate the phenotype, function and the single-cell metabolic profile of SARS-CoV-2-specific T and B cells up to 8 months after the third dose of mRNA vaccine in a cohort of 94 patients with MS treated with different DMT, including cladribine, dimethyl fumarate, fingolimod, interferon, natalizumab, teriflunomide, rituximab or ocrelizumab. Almost all patients display functional immune response to SARS-CoV-2. Different metabolic profiles characterize antigen-specific-T and -B cell response in fingolimod- and natalizumab-treated patients, whose immune response differs from all the other MS treatments.
Collapse
Affiliation(s)
- Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy.
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Moritz Rau
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | | | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Alin Liviu Ciobanu
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Michela Cuccorese
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, Azienda Unità Sanitaria Locale AUSL/AOU Policlinico, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, Azienda Unità Sanitaria Locale AUSL/AOU Policlinico, Modena, Italy
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Francesca Vitetta
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, Nuovo Ospedale Civile Sant'Agostino Estense, University of Modena and Reggio Emilia, Modena, Italy
| | - Martina Cardi
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, Nuovo Ospedale Civile Sant'Agostino Estense, University of Modena and Reggio Emilia, Modena, Italy
| | - Rafael José Argüello
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Diana Ferraro
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, Nuovo Ospedale Civile Sant'Agostino Estense, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy.
- National Institute for Cardiovascular Research, Bologna, Italy.
| |
Collapse
|
7
|
Wang E, Yang QJ, Xu XX, Zou QC, Long Y, Ma G, Deng ZH, Zhao JB, Li MH, Zeng J. Differential pathogenic and molecular features in neurological infection of SARS-CoV-2 Omicron BA.5.2 and BA.2.75 and Delta. J Med Virol 2024; 96:e29357. [PMID: 38235532 DOI: 10.1002/jmv.29357] [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: 07/31/2023] [Revised: 11/07/2023] [Accepted: 12/17/2023] [Indexed: 01/19/2024]
Abstract
The Coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a global threat, exacerbated by the emergence of viral variants. Two variants of SARS-CoV-2, Omicron BA.2.75 and BA.5, led to global infection peaks between May 2022 and May 2023, yet their precise characteristics in pathogenesis are not well understood. In this study, we compared these two Omicron sublineages with the previously dominant Delta variant using a human angiotensin-converting enzyme 2 knock-in mouse model. As expected, Delta exhibited higher viral replication in the lung and brain than both Omicron sublineages which induced less severe lung damage and immune activation. In contrast, the Omicron variants especially BA.5.2 showed a propensity for cellular proliferation and developmental pathways. Both Delta and BA.5.2 variants, but not BA.2.75, led to decreased pulmonary lymphocytes, indicating differential adaptive immune response. Neuroinvasiveness was shared with all strains, accompanied by vascular abnormalities, synaptic injury, and loss of astrocytes. However, Immunostaining assays and transcriptomic analysis showed that BA.5.2 displayed stronger immune suppression and neurodegeneration, while BA.2.75 exhibited more similar characteristics to Delta in the cortex. Such differentially infectious features could be partially attributed to the weakened interaction between Omicron Spike protein and host proteomes decoded via co-immunoprecipitation followed by mass spectrometry in neuronal cells. Our present study supports attenuated replication and pathogenicity of Omicron variants but also highlights their newly infectious characteristics in the lung and brain, especially with BA.5.2 demonstrating enhanced immune evasion and neural damage that could exacerbate neurological sequelae.
Collapse
Affiliation(s)
- Erlin Wang
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Qiao-Jiang Yang
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiang-Xiong Xu
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Qing-Cui Zou
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yanghaopeng Long
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Guanqin Ma
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhong-Hua Deng
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jie-Bin Zhao
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ming-Hua Li
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jianxiong Zeng
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming National High-level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Biodiversity Information, Kunming, Yunnan, China
| |
Collapse
|
8
|
Delgado AH, Fluxa R, Perez-Andres M, Diks AM, van Gaans-van den Brink JAM, Barkoff AM, Blanco E, Torres-Valle A, Berkowska MA, Grigore G, van Dongen J.J.M, Orfao A. Automated EuroFlow approach for standardized in-depth dissection of human circulating B-cells and plasma cells. Front Immunol 2023; 14:1268686. [PMID: 37915569 PMCID: PMC10616957 DOI: 10.3389/fimmu.2023.1268686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023] Open
Abstract
Background Multiparameter flow cytometry (FC) immunophenotyping is a key tool for detailed identification and characterization of human blood leucocytes, including B-lymphocytes and plasma cells (PC). However, currently used conventional data analysis strategies require extensive expertise, are time consuming, and show limited reproducibility. Objective Here, we designed, constructed and validated an automated database-guided gating and identification (AGI) approach for fast and standardized in-depth dissection of B-lymphocyte and PC populations in human blood. Methods For this purpose, 213 FC standard (FCS) datafiles corresponding to umbilical cord and peripheral blood samples from healthy and patient volunteers, stained with the 14-color 18-antibody EuroFlow BIgH-IMM panel, were used. Results The BIgH-IMM antibody panel allowed identification of 117 different B-lymphocyte and PC subsets. Samples from 36 healthy donors were stained and 14 of the datafiles that fulfilled strict inclusion criteria were analysed by an expert flow cytometrist to build the EuroFlow BIgH-IMM database. Data contained in the datafiles was then merged into a reference database that was uploaded in the Infinicyt software (Cytognos, Salamanca, Spain). Subsequently, we compared the results of manual gating (MG) with the performance of two classification algorithms -hierarchical algorithm vs two-step algorithm- for AGI of the cell populations present in 5 randomly selected FCS datafiles. The hierarchical AGI algorithm showed higher correlation values vs conventional MG (r2 of 0.94 vs. 0.88 for the two-step AGI algorithm) and was further validated in a set of 177 FCS datafiles against conventional expert-based MG. For virtually all identifiable cell populations a highly significant correlation was observed between the two approaches (r2>0.81 for 79% of all B-cell populations identified), with a significantly lower median time of analysis per sample (6 vs. 40 min, p=0.001) for the AGI tool vs. MG, respectively and both intra-sample (median CV of 1.7% vs. 10.4% by MG, p<0.001) and inter-expert (median CV of 3.9% vs. 17.3% by MG by 2 experts, p<0.001) variability. Conclusion Our results show that compared to conventional FC data analysis strategies, the here proposed AGI tool is a faster, more robust, reproducible, and standardized approach for in-depth analysis of B-lymphocyte and PC subsets circulating in human blood.
Collapse
Affiliation(s)
- Alejandro H. Delgado
- Cytognos SL, Salamanca, Spain
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
| | | | - Martin Perez-Andres
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Annieck M. Diks
- Department of Immunology (IMMU), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | | | | | - Elena Blanco
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Alba Torres-Valle
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Magdalena A. Berkowska
- Department of Immunology (IMMU), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | | | - J .J .M. van Dongen
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Department of Immunology (IMMU), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
9
|
Nooruzzaman M, Diel DG. Infection Dynamics, Pathogenesis, and Immunity to SARS-CoV-2 in Naturally Susceptible Animal Species. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1195-1201. [PMID: 37782853 PMCID: PMC10558081 DOI: 10.4049/jimmunol.2300378] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/13/2023] [Indexed: 10/04/2023]
Abstract
SARS-CoV-2, the causative agent of the COVID-19 pandemic, presents a broad host range. Domestic cats and white-tailed deer (WTD) are particularly susceptible to SARS-CoV-2 with multiple variant strains being associated with infections in these species. The virus replicates in the upper respiratory tract and in associated lymphoid tissues, and it is shed through oral and nasal secretions, which leads to efficient transmission of the virus to contact animals. Robust cell-mediated and humoral immune responses are induced upon infection in domestic cats, which curb the progression of clinical disease and are associated with control of infection. In WTD, high levels of neutralizing Abs are detected early upon infection. In this review, the current understanding of the infection dynamics, pathogenesis, and immune responses to SARS-CoV-2 infection in animals, with special focus on naturally susceptible felids and WTD, are discussed.
Collapse
Affiliation(s)
- Mohammed Nooruzzaman
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, United States of America
| | - Diego G. Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, United States of America
| |
Collapse
|
10
|
Aiello A, Najafi-Fard S, Goletti D. Initial immune response after exposure to Mycobacterium tuberculosis or to SARS-COV-2: similarities and differences. Front Immunol 2023; 14:1244556. [PMID: 37662901 PMCID: PMC10470049 DOI: 10.3389/fimmu.2023.1244556] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) and Coronavirus disease-2019 (COVID-19), whose etiologic agent is severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), are currently the two deadliest infectious diseases in humans, which together have caused about more than 11 million deaths worldwide in the past 3 years. TB and COVID-19 share several aspects including the droplet- and aerosol-borne transmissibility, the lungs as primary target, some symptoms, and diagnostic tools. However, these two infectious diseases differ in other aspects as their incubation period, immune cells involved, persistence and the immunopathological response. In this review, we highlight the similarities and differences between TB and COVID-19 focusing on the innate and adaptive immune response induced after the exposure to Mtb and SARS-CoV-2 and the pathological pathways linking the two infections. Moreover, we provide a brief overview of the immune response in case of TB-COVID-19 co-infection highlighting the similarities and differences of each individual infection. A comprehensive understanding of the immune response involved in TB and COVID-19 is of utmost importance for the design of effective therapeutic strategies and vaccines for both diseases.
Collapse
Affiliation(s)
| | | | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
11
|
Castleman MJ, Santos AL, Lesteberg KE, Maloney JP, Janssen WJ, Mould KJ, Beckham JD, Pelanda R, Torres RM. Activation and pro-inflammatory cytokine production by unswitched memory B cells during SARS-CoV-2 infection. Front Immunol 2023; 14:1213344. [PMID: 37638016 PMCID: PMC10449608 DOI: 10.3389/fimmu.2023.1213344] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/03/2023] [Indexed: 08/29/2023] Open
Abstract
Memory B cells are comprised of unswitched (CD27+IgD+) and switched (CD27+IgD-) subsets. The origin and function of unswitched human memory B cells are debated in the literature, whereas switched memory B cells are primed to respond to recurrent infection. Unswitched memory B cells have been described to be reduced in frequency with severe SARS-CoV2 infection and here we characterize their activation status, BCR functionality, and contribution to virally-induced cytokine production. Analyses of whole blood from healthy individuals, people immunized against SARS-CoV2, and those who have had mild and severe SARS-CoV2 infection, confirm a reduction in the frequency of unswitched memory B cells during severe SARS-CoV2 infection and demonstrate this reduction is associated with increased levels of systemic TNFα. We further document how severe viral infection is associated with an increased frequency of 'IgD+' only memory B cells that correlate with increased IgG autoantibody levels. Unswitched and switched memory B cells from severe SARS-CoV2 infection displayed evidence of heightened activation with a concomitant reduction in the expression of the inhibitory receptor CD72. Functionally, both populations of memory B cells from severe SARS-COV2 infection harbored a signaling-competent BCR that displayed enhanced BCR signaling activity in the unswitched population. Finally, we demonstrate that B cells from mild SARS-CoV2 infection are poised to secrete pro-inflammatory cytokines IL-6 and TNFα. Importantly, unswitched memory B cells were a major producer of IL-6 and switched memory B cells were a major producer of TNFα in response to viral TLR ligands. Together these data indicate that B cells contribute to the inflammatory milieu during viral infection.
Collapse
Affiliation(s)
- Moriah J. Castleman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Adriana Luna Santos
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Kelsey E. Lesteberg
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Division of Infectious Disease, University of Colorado School of Medicine, Aurora, CO, United States
| | - James P. Maloney
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - William J. Janssen
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Kara J. Mould
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - J. David Beckham
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Medicine, Division of Infectious Disease, University of Colorado School of Medicine, Aurora, CO, United States
- Rocky Mountain Regional VA, Medical Center, Aurora, CO, United States
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Raul M. Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
12
|
Starshinova A, Kudryavtsev I, Rubinstein A, Malkova A, Dovgaluk I, Kudlay D. Tuberculosis and COVID-19 Dually Affect Human Th17 Cell Immune Response. Biomedicines 2023; 11:2123. [PMID: 37626620 PMCID: PMC10452633 DOI: 10.3390/biomedicines11082123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 08/27/2023] Open
Abstract
COVID-19 infection not only profoundly impacts the detection of tuberculosis infection (Tbc) but also affects modality in tuberculosis patient immune response. It is important to determine immune response alterations in latent tuberculosis infection as well as in SARS-CoV-2-infected tuberculosis patients. Such changes may have underlying effects on the development and course of further tuberculosis. Here, we aimed to review the characteristics of immune response in TB patients or convalescent COVID-19 patients with latent TB infection (LTBI). MATERIALS AND METHODS We analyzed the features of immune response in tuberculosis and COVID-19 patients. For this, we analyzed publications released from December 2019 to March 2023; those which were published in accessible international databases ("Medline", "PubMed", "Scopus") and with keywords such as "COVID-19", "SARS-CoV-2", "tuberculosis", "pulmonary tuberculosis", "latent tuberculosis infection", "Treg", "follicular Treg", and "Treg subsets", we considered. RESULTS Through our analysis, we found that tuberculosis patients who had been infected with COVID-19 previously and elevated Th1 and Th2 cell levels. High levels of Th1 and Th2 cells may serve as a positive marker, characterizing activated immune response during TB infection. COVID-19 or post-COVID-19 subjects showed decreased Th17 levels, indicating a lack of tuberculosis development. Moreover, the typical course of tuberculosis is associated with an increase in Treg level, but COVID-19 contributes to a hyperinflammatory response. CONCLUSION According to the data obtained, the course of tuberculosis proceeds in a dissimilar way due to the distinct immune response, elicited by SARS-CoV-2. Importantly, the development of active tuberculosis with a severe course is associated with a decline in Treg levels. Both pathogens lead to disturbed immune responses, increasing the risk of developing severe TB. The insights and findings of this paper may be used to improve the future management of individuals with latent and active tuberculosis.
Collapse
Affiliation(s)
- Anna Starshinova
- Almazov National Medical Research Centre, 197341 St-Petersburg, Russia
| | - Igor Kudryavtsev
- Department of Immunology, Institution of Experimental Medicine, 197022 St-Petersburg, Russia; (I.K.); (A.R.)
| | - Artem Rubinstein
- Department of Immunology, Institution of Experimental Medicine, 197022 St-Petersburg, Russia; (I.K.); (A.R.)
| | - Anna Malkova
- Faculty of Natural Sciences, Ariel University, Ariel 40700, Israel;
| | - Irina Dovgaluk
- Phthisiopulmonology Department, Research Institute of Phthisiopulmonology, 191036 St-Petersburg, Russia;
| | - Dmitry Kudlay
- Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia;
- Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
| |
Collapse
|
13
|
Prebensen C, Lefol Y, Myhre PL, Lüders T, Jonassen C, Blomfeldt A, Omland T, Nilsen H, Berdal JE. Longitudinal whole blood transcriptomic analysis characterizes neutrophil activation and interferon signaling in moderate and severe COVID-19. Sci Rep 2023; 13:10368. [PMID: 37365222 PMCID: PMC10293211 DOI: 10.1038/s41598-023-37606-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 06/24/2023] [Indexed: 06/28/2023] Open
Abstract
A maladaptive inflammatory response has been implicated in the pathogenesis of severe COVID-19. This study aimed to characterize the temporal dynamics of this response and investigate whether severe disease is associated with distinct gene expression patterns. We performed microarray analysis of serial whole blood RNA samples from 17 patients with severe COVID-19, 15 patients with moderate disease and 11 healthy controls. All study subjects were unvaccinated. We assessed whole blood gene expression patterns by differential gene expression analysis, gene set enrichment, two clustering methods and estimated relative leukocyte abundance using CIBERSORT. Neutrophils, platelets, cytokine signaling, and the coagulation system were activated in COVID-19, and this broad immune activation was more pronounced in severe vs. moderate disease. We observed two different trajectories of neutrophil-associated genes, indicating the emergence of a more immature neutrophil phenotype over time. Interferon-associated genes were strongly enriched in early COVID-19 before falling markedly, with modest severity-associated differences in trajectory. In conclusion, COVID-19 necessitating hospitalization is associated with a broad inflammatory response, which is more pronounced in severe disease. Our data suggest a progressively more immature circulating neutrophil phenotype over time. Interferon signaling is enriched in COVID-19 but does not seem to drive severe disease.
Collapse
Affiliation(s)
- Christian Prebensen
- Department of Infectious Diseases, Oslo University Hospital, Kirkeveien 166, 0450, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Yohan Lefol
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Peder L Myhre
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Torben Lüders
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | | | - Anita Blomfeldt
- Department of Microbiology and Infection Control, Akershus University Hospital, Lørenskog, Norway
| | - Torbjørn Omland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Hilde Nilsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Jan-Erik Berdal
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
14
|
De Biasi S, Mattioli M, Meschiari M, Lo Tartaro D, Paolini A, Borella R, Neroni A, Fidanza L, Busani S, Girardis M, Coppi F, Mattioli AV, Guaraldi G, Mussini C, Cossarizza A, Gibellini L. Prognostic immune markers identifying patients with severe COVID-19 who respond to tocilizumab. Front Immunol 2023; 14:1123807. [PMID: 37215114 PMCID: PMC10196248 DOI: 10.3389/fimmu.2023.1123807] [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: 12/14/2022] [Accepted: 04/26/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction A growing number of evidences suggest that the combination of hyperinflammation, dysregulated T and B cell response and cytokine storm play a major role in the immunopathogenesis of severe COVID-19. IL-6 is one of the main pro-inflammatory cytokines and its levels are increased during SARS-CoV-2 infection. Several observational and randomized studies demonstrated that tocilizumab, an IL-6R blocker, improves survival in critically ill patients both in infectious disease and intensive care units. However, despite transforming the treatment options for COVID-19, IL-6R inhibition is still ineffective in a fraction of patients. Methods In the present study, we investigated the impact of two doses of tocilizumab in patients with severe COVID-19 who responded or not to the treatment by analyzing a panel of cytokines, chemokines and other soluble factors, along with the composition of peripheral immune cells, paying a particular attention to T and B lymphocytes. Results We observed that, in comparison with non-responders, those who responded to tocilizumab had different levels of several cytokines and different T and B cells proportions before starting therapy. Moreover, in these patients, tocilizumab was further able to modify the landscape of the aforementioned soluble molecules and cellular markers. Conclusions We found that tocilizumab has pleiotropic effects and that clinical response to this drug remain heterogenous. Our data suggest that it is possible to identify patients who will respond to treatment and that the administration of tocilizumab is able to restore the immune balance through the re-establishment of different cell populations affected by SARS-COV-2 infection, highlighting the importance of temporal examination of the pathological features from the diagnosis.
Collapse
Affiliation(s)
- Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Marianna Meschiari
- Infectious Diseases Clinics, Azienda Ospedaliera Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Lucia Fidanza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Stefano Busani
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Girardis
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Coppi
- Department of Metabolic Sciences and Neurosciences, Azienda Ospedaliera Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Vittoria Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, Azienda Ospedaliera Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, Azienda Ospedaliera Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
- National Institute for Cardiovascular Research, Bologna, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| |
Collapse
|
15
|
Pan T, Cao G, Tang E, Zhao Y, Penaloza-MacMaster P, Fang Y, Huang J. A single-cell atlas reveals shared and distinct immune responses and metabolic profiles in SARS-CoV-2 and HIV-1 infections. Front Genet 2023; 14:1105673. [PMID: 36992700 PMCID: PMC10040851 DOI: 10.3389/fgene.2023.1105673] [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: 11/22/2022] [Accepted: 03/01/2023] [Indexed: 03/16/2023] Open
Abstract
Introduction: Within the inflammatory immune response to viral infection, the distribution and cell type-specific profiles of immune cell populations and the immune-mediated viral clearance pathways vary according to the specific virus. Uncovering the immunological similarities and differences between viral infections is critical to understanding disease progression and developing effective vaccines and therapies. Insight into COVID-19 disease progression has been bolstered by the integration of single-cell (sc)RNA-seq data from COVID-19 patients with data from related viruses to compare immune responses. Expanding this concept, we propose that a high-resolution, systematic comparison between immune cells from SARS-CoV-2 infection and an inflammatory infectious disease with a different pathophysiology will provide a more comprehensive picture of the viral clearance pathways that underscore immunological and clinical differences between infections. Methods: Using a novel consensus single-cell annotation method, we integrate previously published scRNA-seq data from 111,566 single PBMCs from 7 COVID-19, 10 HIV-1+, and 3 healthy patients into a unified cellular atlas. We compare in detail the phenotypic features and regulatory pathways in the major immune cell clusters. Results: While immune cells in both COVID-19 and HIV-1+ cohorts show shared inflammation and disrupted mitochondrial function, COVID-19 patients exhibit stronger humoral immunity, broader IFN-I signaling, elevated Rho GTPase and mTOR pathway activity, and downregulated mitophagy. Discussion: Our results indicate that differential IFN-I signaling regulates the distinct immune responses in the two diseases, revealing insight into fundamental disease biology and potential therapeutic candidates.
Collapse
Affiliation(s)
- Tony Pan
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Guoshuai Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Erting Tang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Yu Zhao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | | | - Yun Fang
- Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Jun Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| |
Collapse
|
16
|
Desai N, Pradhan V, Chougule D, Tiwari S, Mandke C, Yadav RM, Athvale A, Kawle J, Pai V, Pawaskar S, Kharkar H, Bhosale S, Parab A, Ansari S, Kumar KH, Mhashal S, Redkar N, Madkaikar M. Perturbations of immune landscape in COVID-19 associated mucormycosis. Mycoses 2023; 66:226-236. [PMID: 36380699 DOI: 10.1111/myc.13546] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND A rise in secondary fungal infections during the COVID-19 pandemic necessitates a deeper understanding of the associated immunological perturbations. OBJECTIVES To evaluate the clinical and immunological characteristics observed in patients with COVID-19 associated mucormycosis (CAM) infection. PATIENTS/ METHODS Cases of mucormycosis with or post-COVID-19 infection were compared with cases of acute COVID-19 and convalescent COVID-19. Lymphocyte subsets, cytokines and other laboratory markers were compared between the groups. RESULTS The frequency of proposed risk factors for CAM was diabetes mellitus (77%), recent history of steroid use (69%) and hypoxia during COVID-19 infection (52%). Iron metabolism was dysregulated in CAM patients with low TIBC and total iron. Further, CAM was accompanied with lymphopenia with drastic reduction in B cell counts; however, plasmablasts were not altered. Further, CAM patients had low immunoglobulin levels and antibodies specific to mucor peptide did not increase in CAM suggesting dysfunction in B-cell response. There was increase in activated effector cytotoxic CD8 T cells and NK cells in CAM compared with COVID-19 infection and healthy controls. Among T helper cells, Tregs were reduced and Th-1 frequency was increased in CAM compared with COVID-19 infection. A distinct cytokine signature was evident in CAM with increase in IL-1β, IFN-γ, IL-6, IL-22, IL-17A, IL-10, IL-2, IL-8, IL-7, IL-21 and GM-CSF. CONCLUSION This is the first study on immunophenotyping in CAM suggesting the need for long-term monitoring of B-cell function after SARS-CoV-2 in patients with dysregulated glycaemic control and the possible benefit of therapeutic supplementation with intravenous immunoglobulins in CAM.
Collapse
Affiliation(s)
- Nidhi Desai
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Vandana Pradhan
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Durga Chougule
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Smrati Tiwari
- Department of Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Charuta Mandke
- Department of Ophthalmology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Reetika Malik Yadav
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Amita Athvale
- Department of Pulmonary Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Juhi Kawle
- Department of Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Vinayak Pai
- Department of Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Swapnal Pawaskar
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Harshada Kharkar
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Snehal Bhosale
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Ankita Parab
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Shazia Ansari
- Department of Ophthalmology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Kinnera Harish Kumar
- Department of Otorhinolaryngology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Shashikant Mhashal
- Department of Otolaryngology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Neelam Redkar
- Department of Medicine, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Manisha Madkaikar
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| |
Collapse
|
17
|
Lo Tartaro D, Paolini A, Mattioli M, Swatler J, Neroni A, Borella R, Santacroce E, Di Nella A, Gozzi L, Busani S, Cuccorese M, Trenti T, Meschiari M, Guaraldi G, Girardis M, Mussini C, Piwocka K, Gibellini L, Cossarizza A, De Biasi S. Detailed characterization of SARS-CoV-2-specific T and B cells after infection or heterologous vaccination. Front Immunol 2023; 14:1123724. [PMID: 36845156 PMCID: PMC9947839 DOI: 10.3389/fimmu.2023.1123724] [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: 12/14/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
The formation of a robust long-term antigen (Ag)-specific memory, both humoral and cell-mediated, is created following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination. Here, by using polychromatic flow cytometry and complex data analyses, we deeply investigated the magnitude, phenotype, and functionality of SARS-CoV-2-specific immune memory in two groups of healthy subjects after heterologous vaccination compared to a group of subjects who recovered from SARS-CoV-2 infection. We find that coronavirus disease 2019 (COVID-19) recovered patients show different long-term immunological profiles compared to those of donors who had been vaccinated with three doses. Vaccinated individuals display a skewed T helper (Th)1 Ag-specific T cell polarization and a higher percentage of Ag-specific and activated memory B cells expressing immunoglobulin (Ig)G compared to those of patients who recovered from severe COVID-19. Different polyfunctional properties characterize the two groups: recovered individuals show higher percentages of CD4+ T cells producing one or two cytokines simultaneously, while the vaccinated are distinguished by highly polyfunctional populations able to release four molecules, namely, CD107a, interferon (IFN)-γ, tumor necrosis factor (TNF), and interleukin (IL)-2. These data suggest that functional and phenotypic properties of SARS-CoV-2 adaptive immunity differ in recovered COVID-19 individuals and vaccinated ones.
Collapse
Affiliation(s)
- Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Julian Swatler
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Alessia Di Nella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Licia Gozzi
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Anesthesia and Intensive Care, Azienda Ospedaliero-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Michela Cuccorese
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, Azienda Unità Sanitaria Locale AUSL/AOU Policlinico, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, Azienda Unità Sanitaria Locale AUSL/AOU Policlinico, Modena, Italy
| | - Marianna Meschiari
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Anesthesia and Intensive Care, Azienda Ospedaliero-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
- National Institute for Cardiovascular Research, Bologna, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| |
Collapse
|
18
|
A Comprehensive Analysis of Cytokine Network in Centenarians. Int J Mol Sci 2023; 24:ijms24032719. [PMID: 36769039 PMCID: PMC9916918 DOI: 10.3390/ijms24032719] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Cytokines have been investigated extensively in elderly people, with conflicting results. We performed a comprehensive analysis of the plasma levels of 62 cytokines and growth factors involved in the regulation of the immune system, in healthy centenarians, and middle-aged controls. We confirmed the previously observed increase in the levels of several pro-inflammatory cytokines, such as TNF-α and IL-6, and found that several other cytokines, directly or indirectly involved in inflammation (such as IFN-α, IL-23, CCL-5), were present at higher levels in centenarians. We did not observe any increase in the levels of anti-inflammatory cytokines, with the notable exception of the Th2-shifting cytokine IL-19. No relevant difference was observed in cytokines regulating T cell immunity. Several growth factors having a role in regulating immunity, such as G-CSF, GM-CSF, EGF, and VEGF, were upregulated in centenarians, too. Principal component analysis of the cytokine dataset showed that pro and anti-inflammatory cytokines were the variables that contributed the most to the variability of the data we observed.
Collapse
|
19
|
Tirelli C, De Amici M, Albrici C, Mira S, Nalesso G, Re B, Corsico AG, Mondoni M, Centanni S. Exploring the Role of Immune System and Inflammatory Cytokines in SARS-CoV-2 Induced Lung Disease: A Narrative Review. BIOLOGY 2023; 12:biology12020177. [PMID: 36829456 PMCID: PMC9953200 DOI: 10.3390/biology12020177] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative pathogen of coronavirus disease 19 (COVID-19). COVID-19 can manifest with a heterogenous spectrum of disease severity, from mild upper airways infection to severe interstitial pneumonia and devastating acute respiratory distress syndrome (ARDS). SARS-CoV-2 infection may induce an over activation of the immune system and the release of high concentrations of pro-inflammatory cytokines, leading to a "cytokine storm", a recognized pathogenetic mechanism in the genesis of SARS-CoV-2-induced lung disease. This overproduction of inflammatory cytokines has been recognized as a poor prognostic factor, since it can lead to disease progression, organ failure, ARDS and death. Moreover, the immune system shows dysregulated activity, particularly through activated macrophages and T-helper cells and in the co-occurrent exhaustion of lymphocytes. We carried out a non-systematic literature review aimed at providing an overview of the current knowledge on the pathologic mechanisms played by the immune system and the inflammation in the genesis of SARS-CoV-2-induced lung disease. An overview on potential treatments for this harmful condition and for contrasting the "cytokine storm" has also been presented. Finally, a look at the experimented experimental vaccines against SARS-CoV-2 has been included.
Collapse
Affiliation(s)
- Claudio Tirelli
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy
- Correspondence:
| | - Mara De Amici
- Immuno-Allergology Laboratory of Clinical Chemistry and Department of Pediatrics, IRCCS Policlinico San Matteo University Hospital, 27100 Pavia, Italy
| | - Cristina Albrici
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy
| | - Sabrina Mira
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy
| | - Giulia Nalesso
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy
| | - Beatrice Re
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy
| | - Angelo Guido Corsico
- Pulmonology Unit, Department of Medical Sciences and Infectious Diseases, IRCCS Policlinico San Matteo University Hospital, 27100 Pavia, Italy
| | - Michele Mondoni
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy
| | - Stefano Centanni
- Respiratory Unit, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20142 Milan, Italy
| |
Collapse
|
20
|
COVID-19 Heart Lesions in Children: Clinical, Diagnostic and Immunological Changes. Int J Mol Sci 2023; 24:ijms24021147. [PMID: 36674665 PMCID: PMC9866514 DOI: 10.3390/ijms24021147] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
In the beginning of COVID-19, the proportion of confirmed cases in the pediatric population was relatively small and there was an opinion that children often had a mild or asymptomatic course of infection. Our understanding of the immune response, diagnosis and treatment of COVID-19 is highly oriented towards the adult population. At the same time, despite the fact that COVID-19 in children usually occurs in a mild form, there is an incomplete understanding of the course as an acute infection and its subsequent manifestations such as Long-COVID-19 or Post-COVID-19, PASC in the pediatric population, correlations with comorbidities and immunological changes. In mild COVID-19 in childhood, some authors explain the absence of population decreasing T and B lymphocytes. Regardless of the patient's condition, they can have the second phase, related to the exacerbation of inflammation in the heart tissue even if the viral infection was completely eliminated-post infectious myocarditis. Mechanism of myocardial dysfunction development in MIS-C are not fully understood. It is known that various immunocompetent cells, including both resident inflammatory cells of peripheral tissues (for example macrophages, dendritic cells, resident memory T-lymphocytes and so on) and also circulating in the peripheral blood immune cells play an important role in the immunopathogenesis of myocarditis. It is expected that hyperproduction of interferons and the enhanced cytokine response of T cells 1 and 2 types contribute to dysfunction of the myocardium. However, the role of Th1 in the pathogenesis of myocarditis remains highly controversial. At the same time, the clinical manifestations and mechanisms of damage, including the heart, both against the background and after COVID-19, in children differ from adults. Further studies are needed to evaluate whether transient or persistent cardiac complications are associated with long-term adverse cardiac events.
Collapse
|
21
|
Shafqat A, Omer MH, Ahmad O, Niaz M, Abdulkader HS, Shafqat S, Mushtaq AH, Shaik A, Elshaer AN, Kashir J, Alkattan K, Yaqinuddin A. SARS-CoV-2 epitopes inform future vaccination strategies. Front Immunol 2022; 13:1041185. [PMID: 36505475 PMCID: PMC9732895 DOI: 10.3389/fimmu.2022.1041185] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/11/2022] [Indexed: 11/27/2022] Open
Abstract
All currently approved COVID-19 vaccines utilize the spike protein as their immunogen. SARS-CoV-2 variants of concern (VOCs) contain mutations in the spike protein, enabling them to escape infection- and vaccination-induced immune responses to cause reinfection. New vaccines are hence being researched intensively. Studying SARS-CoV-2 epitopes is essential for vaccine design, as identifying targets of broadly neutralizing antibody responses and immunodominant T-cell epitopes reveal candidates for inclusion in next-generation COVID-19 vaccines. We summarize the major studies which have reported on SARS-CoV-2 antibody and T-cell epitopes thus far. These results suggest that a future of pan-coronavirus vaccines, which not only protect against SARS-CoV-2 but numerous other coronaviruses, may be possible. The T-cell epitopes of SARS-CoV-2 have gotten less attention than neutralizing antibody epitopes but may provide new strategies to control SARS-CoV-2 infection. T-cells target many SARS-CoV-2 antigens other than spike, recognizing numerous epitopes within these antigens, thereby limiting the chance of immune escape by VOCs that mainly possess spike protein mutations. Therefore, augmenting vaccination-induced T-cell responses against SARS-CoV-2 may provide adequate protection despite broad antibody escape by VOCs.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia,*Correspondence: Areez Shafqat,
| | - Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Omar Ahmad
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Mahnoor Niaz
- Medical College, Aga Khan University, Karachi, Pakistan
| | | | | | | | - Abdullah Shaik
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia,Department of Comparative Medicine, King Faisal Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | |
Collapse
|
22
|
Mendez-Cortina Y, Rodriguez-Perea AL, Chvatal-Medina M, Lopera TJ, Alvarez-Mesa N, Rodas-Marín JK, Moncada DC, Rugeles MT, Velilla PA. Dynamics of humoral immune response in SARS-CoV-2 infected individuals with different clinical stages. Front Immunol 2022; 13:1007068. [DOI: 10.3389/fimmu.2022.1007068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
BackgroundThe COVID-19 pandemic remains a global health problem. As in other viral infections, the humoral immune response against SARS-CoV-2 is thought to be crucial for controlling the infection. However, the dynamic of B cells in the clinical spectrum of this disease is still controversial. This study aimed to characterize B cell subsets and neutralizing responses in COVID-19 patients according to disease severity through a one-month follow-up.MethodsA cohort of 71 individuals with SARS-CoV-2 infection confirmed by RT-PCR were recruited and classified into four groups: i) asymptomatic; ii) symptomatic outpatients; iii) hospitalized in ward, and iv) intensive care unit patients (ICU). Samples were taken at days 0 (inclusion to the study), 7 and 30. B cell subsets and neutralizing antibodies were assessed using multiparametric flow cytometry and plaque reduction neutralization, respectively.ResultsOlder age, male gender and body mass index over 25 were common factors among hospitalized and ICU patients, compared to those with milder clinical presentations. In addition, those requiring hospitalization had more comorbidities. A significant increase in the frequencies of CD19+ cells at day 0 was observed in hospitalized and ICU patients compared to asymptomatic and symptomatic groups. Likewise, the frequency of plasmablasts was significantly increased at the first sample in the ICU group compared to the asymptomatic group, but then waned over time. The frequency of naïve B cells decreased at days 7 and 30 compared to day 0 in hospitalized and ICU patients. The neutralizing antibody titers were higher as the severity of COVID-19 increased; in asymptomatic individuals, it was strongly correlated with the percentage of IgM+ switched memory B cells, and a moderate correlation was found with plasmablasts.ConclusionThe humoral immune response is variable among SARS-CoV-2 infected people depending on the severity and time of clinical evolution. In severe COVID-19 patients, a higher plasmablast frequency and neutralizing antibody response were observed, suggesting that, despite having a robust humoral immunity, this response could be late, having a low impact on disease outcome.
Collapse
|
23
|
Öztürk-Kaygusuz T, Sağmak-Tartar A, Akbulut A. An Unusual Cause of Acute Isolated Hepatitis in a Cancer Patient Post-COVID Pneumonia: HSV-2. INFECTIOUS DISEASES & CLINICAL MICROBIOLOGY 2022; 4:210-213. [PMID: 38633396 PMCID: PMC10986694 DOI: 10.36519/idcm.2022.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 06/29/2022] [Indexed: 04/19/2024]
Abstract
Herpes simplex virus is a rare cause of hepatitis in immunosuppressed and immunocompetent individuals. It can cause clinical pictures in patients ranging from an asymptomatic course to fatal acute fulminant hepatitis. Early diagnosis and treatment may be delayed if it is not suspected because of the patients' nonspecific clinic. This study presents a case of isolated HSV-2 hepatitis in a patient with a history of cancer chemotherapy and a recent diagnosis of COVID-19 who received steroid therapy.
Collapse
Affiliation(s)
- Türkkan Öztürk-Kaygusuz
- Department of Infectious Diseases and Clinical Microbiology, Fırat University School of Medicine, Elazığ, Turkey
| | - Ayşe Sağmak-Tartar
- Department of Infectious Diseases and Clinical Microbiology, Fırat University School of Medicine, Elazığ, Turkey
| | - Ayhan Akbulut
- Department of Infectious Diseases and Clinical Microbiology, Fırat University School of Medicine, Elazığ, Turkey
| |
Collapse
|
24
|
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: 8.0] [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
|
25
|
Khare K, Pandey R. Cellular heterogeneity in disease severity and clinical outcome: Granular understanding of immune response is key. Front Immunol 2022; 13:973070. [PMID: 36072602 PMCID: PMC9441806 DOI: 10.3389/fimmu.2022.973070] [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: 06/19/2022] [Accepted: 07/26/2022] [Indexed: 12/15/2022] Open
Abstract
During an infectious disease progression, it is crucial to understand the cellular heterogeneity underlying the differential immune response landscape that will augment the precise information of the disease severity modulators, leading to differential clinical outcome. Patients with COVID-19 display a complex yet regulated immune profile with a heterogeneous array of clinical manifestation that delineates disease severity sub-phenotypes and worst clinical outcomes. Therefore, it is necessary to elucidate/understand/enumerate the role of cellular heterogeneity during COVID-19 disease to understand the underlying immunological mechanisms regulating the disease severity. This article aims to comprehend the current findings regarding dysregulation and impairment of immune response in COVID-19 disease severity sub-phenotypes and relate them to a wide array of heterogeneous populations of immune cells. On the basis of the findings, it suggests a possible functional correlation between cellular heterogeneity and the COVID-19 disease severity. It highlights the plausible modulators of age, gender, comorbidities, and hosts' genetics that may be considered relevant in regulating the host response and subsequently the COVID-19 disease severity. Finally, it aims to highlight challenges in COVID-19 disease that can be achieved by the application of single-cell genomics, which may aid in delineating the heterogeneity with more granular understanding. This will augment our future pandemic preparedness with possibility to identify the subset of patients with increased diseased severity.
Collapse
Affiliation(s)
- Kriti Khare
- Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajesh Pandey
- Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
26
|
Morales-Núñez JJ, García-Chagollán M, Muñoz-Valle JF, Díaz-Pérez SA, Torres-Hernández PC, Rodríguez-Reyes SC, Santoscoy-Ascencio G, Sierra García de Quevedo JJ, Hernández-Bello J. Differences in B-Cell Immunophenotypes and Neutralizing Antibodies Against SARS-CoV-2 After Administration of BNT162b2 (Pfizer-BioNTech) Vaccine in Individuals with and without Prior COVID-19 - A Prospective Cohort Study. J Inflamm Res 2022; 15:4449-4466. [PMID: 35958186 PMCID: PMC9361858 DOI: 10.2147/jir.s374304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/30/2022] [Indexed: 12/15/2022] Open
Abstract
Purpose Understanding the humoral immune response dynamics carried out by B cells in COVID-19 vaccination is little explored; therefore, we analyze the changes induced in the different cellular subpopulations of B cells after vaccination with BNT162b2 (Pfizer-BioNTech). Methods This prospective cohort study evaluated thirty-nine immunized health workers (22 with prior COVID-19 and 17 without prior COVID-19) and ten subjects not vaccinated against SARS-CoV-2 (control group). B cell subpopulations (transitional, mature, naïve, memory, plasmablasts, early plasmablast, and double-negative B cells) and neutralizing antibody levels were analyzed and quantified by flow cytometry and ELISA, respectively. Results The dynamics of the B cells subpopulations after vaccination showed the following pattern: the percentage of transitional B cells was higher in the prior COVID-19 group (p < 0.05), whereas virgin B cells were more prevalent in the group without prior COVID-19 (p < 0.05), mature B cells predominated in both vaccinated groups (p < 0.01), and memory B cells, plasmablasts, early plasmablasts, and double-negative B cells were higher in the not vaccinated group (p < 0.05). Conclusion BNT162b2 vaccine induces changes in B cell subpopulations, especially generating plasma cells and producing neutralizing antibodies against SARS-CoV-2. However, the previous infection with SARS-CoV-2 does not significantly alter the dynamics of these subpopulations but induces more rapid and optimal antibody production.
Collapse
Affiliation(s)
- José Javier Morales-Núñez
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Mariel García-Chagollán
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - José Francisco Muñoz-Valle
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Saúl Alberto Díaz-Pérez
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | | | - Saraí Citlalic Rodríguez-Reyes
- Institute of Translational Nutrigenetics and Nutrigenomics, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | | | | | - Jorge Hernández-Bello
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
- Correspondence: Jorge Hernández-Bello,s Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, 44340, Mexico, Tel +52 3334509355, Email
| |
Collapse
|
27
|
Jonigk D, Werlein C, Acker T, Aepfelbacher M, Amann KU, Baretton G, Barth P, Bohle RM, Büttner A, Büttner R, Dettmeyer R, Eichhorn P, Elezkurtaj S, Esposito I, Evert K, Evert M, Fend F, Gaßler N, Gattenlöhner S, Glatzel M, Göbel H, Gradhand E, Hansen T, Hartmann A, Heinemann A, Heppner FL, Hilsenbeck J, Horst D, Kamp JC, Mall G, Märkl B, Ondruschka B, Pablik J, Pfefferle S, Quaas A, Radbruch H, Röcken C, Rosenwald A, Roth W, Rudelius M, Schirmacher P, Slotta-Huspenina J, Smith K, Sommer L, Stock K, Ströbel P, Strobl S, Titze U, Weirich G, Weis J, Werner M, Wickenhauser C, Wiech T, Wild P, Welte T, von Stillfried S, Boor P. Organ manifestations of COVID-19: what have we learned so far (not only) from autopsies? Virchows Arch 2022; 481:139-159. [PMID: 35364700 PMCID: PMC8975445 DOI: 10.1007/s00428-022-03319-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 01/08/2023]
Abstract
The use of autopsies in medicine has been declining. The COVID-19 pandemic has documented and rejuvenated the importance of autopsies as a tool of modern medicine. In this review, we discuss the various autopsy techniques, the applicability of modern analytical methods to understand the pathophysiology of COVID-19, the major pathological organ findings, limitations or current studies, and open questions. This article summarizes published literature and the consented experience of the nationwide network of clinical, neuro-, and forensic pathologists from 27 German autopsy centers with more than 1200 COVID-19 autopsies. The autopsy tissues revealed that SARS-CoV-2 can be found in virtually all human organs and tissues, and the majority of cells. Autopsies have revealed the organ and tissue tropism of SARS-CoV-2, and the morphological features of COVID-19. This is characterized by diffuse alveolar damage, combined with angiocentric disease, which in turn is characterized by endothelial dysfunction, vascular inflammation, (micro-) thrombosis, vasoconstriction, and intussusceptive angiogenesis. These findings explained the increased pulmonary resistance in COVID-19 and supported the recommendations for antithrombotic treatment in COVID-19. In contrast, in extra-respiratory organs, pathological changes are often nonspecific and unclear to which extent these changes are due to direct infection vs. indirect/secondary mechanisms of organ injury, or a combination thereof. Ongoing research using autopsies aims at answering questions on disease mechanisms, e.g., focusing on variants of concern, and future challenges, such as post-COVID conditions. Autopsies are an invaluable tool in medicine and national and international interdisciplinary collaborative autopsy-based research initiatives are essential.
Collapse
Affiliation(s)
- Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany.
| | | | - Till Acker
- Institute of Neuropathology, University Hospital Giessen and Marburg, Giessen, Germany
| | - Martin Aepfelbacher
- Institute of Medical Microbiology, Virology, and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin U Amann
- Department of Nephropathology, University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Gustavo Baretton
- Department of Pathology, University Hospital Dresden, Dresden, Germany
| | - Peter Barth
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Rainer M Bohle
- Department of Pathology, University Hospital Saarland Homburg, Homburg, Germany
| | - Andreas Büttner
- Institute of Forensic Medicine, University Medical Center Rostock, Rostock, Germany
| | - Reinhard Büttner
- Department of Pathology, University Hospital Cologne, Cologne, Germany
| | - Reinhard Dettmeyer
- Department of Legal Medicine, University Hospital Giessen and Marburg, Giessen, Germany
| | - Philip Eichhorn
- Department of Pathology, University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Sefer Elezkurtaj
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Irene Esposito
- Department of Pathology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Katja Evert
- Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Evert
- Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - Falko Fend
- Department of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Nikolaus Gaßler
- Department of Pathology, University Hospital Jena, Jena, Germany
| | - Stefan Gattenlöhner
- Department of Pathology, University Hospital Giessen and Marburg, Giessen, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Heike Göbel
- Department of Pathology, University Hospital Cologne, Cologne, Germany
| | - Elise Gradhand
- Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt, Germany
| | - Torsten Hansen
- Department of Pathology, University Hospital OWL of the Bielefeld University, Campus Lippe, Detmold, Germany
| | - Arndt Hartmann
- Department of Pathology, University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Axel Heinemann
- Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank L Heppner
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
- Cluster of Excellence, NeuroCure, Berlin, Germany
| | - Julia Hilsenbeck
- Department of Pathology, University Hospital Dresden, Dresden, Germany
| | - David Horst
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jan C Kamp
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Gita Mall
- Department of Legal Medicine, University Hospital Jena, Jena, Germany
| | - Bruno Märkl
- General Pathology and Molecular Diagnostics, University Hospital Augsburg, Augsburg, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jessica Pablik
- Department of Pathology, University Hospital Dresden, Dresden, Germany
| | - Susanne Pfefferle
- Institute of Medical Microbiology, Virology, and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Quaas
- Department of Pathology, University Hospital Cologne, Cologne, Germany
| | - Helena Radbruch
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christoph Röcken
- Department of Pathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Wilfried Roth
- Department of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Martina Rudelius
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Peter Schirmacher
- Department of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Julia Slotta-Huspenina
- Department of Pathology, TUM School of Medicine of Technical University of Munich, Munich, Germany
| | - Kevin Smith
- Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt, Germany
| | - Linna Sommer
- Department of Pathology, University Hospital Dresden, Dresden, Germany
| | - Konrad Stock
- Department of Nephrology, TUM School of Medicine of Technical University of Munich, Munich, Germany
| | - Philipp Ströbel
- Department of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Stephanie Strobl
- Department of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Ulf Titze
- Department of Pathology, University Hospital OWL of the Bielefeld University, Campus Lippe, Detmold, Germany
| | - Gregor Weirich
- Department of Pathology, TUM School of Medicine of Technical University of Munich, Munich, Germany
| | - Joachim Weis
- Department of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Martin Werner
- Institute for Surgical Pathology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Claudia Wickenhauser
- Department of Pathology, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Thorsten Wiech
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Wild
- Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | | | - Peter Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany.
- Department of Nephrology and Immunology, University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
28
|
Paolini A, Borella R, Neroni A, Lo Tartaro D, Mattioli M, Fidanza L, Di Nella A, Santacroce E, Gozzi L, Busani S, Trenti T, Meschiari M, Guaraldi G, Girardis M, Mussini C, Gibellini L, De Biasi S, Cossarizza A. Patients Recovering from Severe COVID-19 Develop a Polyfunctional Antigen-Specific CD4+ T Cell Response. Int J Mol Sci 2022; 23:8004. [PMID: 35887351 PMCID: PMC9323836 DOI: 10.3390/ijms23148004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 12/10/2022] Open
Abstract
Specific T cells are crucial to control SARS-CoV-2 infection, avoid reinfection and confer protection after vaccination. We have studied patients with severe or moderate COVID-19 pneumonia, compared to patients who recovered from a severe or moderate infection that had occurred about 4 months before the analyses. In all these subjects, we assessed the polyfunctionality of virus-specific CD4+ and CD8+ T cells by quantifying cytokine production after in vitro stimulation with different SARS-CoV-2 peptide pools covering different proteins (M, N and S). In particular, we quantified the percentage of CD4+ and CD8+ T cells simultaneously producing interferon-γ, tumor necrosis factor, interleukin (IL)-2, IL-17, granzyme B, and expressing CD107a. Recovered patients who experienced a severe disease display high proportions of antigen-specific CD4+ T cells producing Th1 and Th17 cytokines and are characterized by polyfunctional SARS-CoV-2-specific CD4+ T cells. A similar profile was found in patients experiencing a moderate form of COVID-19 pneumonia. No main differences in polyfunctionality were observed among the CD8+ T cell compartments, even if the proportion of responding cells was higher during the infection. The identification of those functional cell subsets that might influence protection can thus help in better understanding the complexity of immune response to SARS-CoV-2.
Collapse
Affiliation(s)
- Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Lucia Fidanza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Alessia Di Nella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Licia Gozzi
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
- Department of Anesthesia and Intensive Care, AOU Policlinico and University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy;
| | - Marianna Meschiari
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
- Department of Anesthesia and Intensive Care, AOU Policlinico and University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
- National Institute for Cardiovascular Research, Via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
29
|
Dhanya CR, Shailaja A, Mary AS, Kandiyil SP, Savithri A, Lathakumari VS, Veettil JT, Vandanamthadathil JJ, Madhavan M. RNA Viruses, Pregnancy and Vaccination: Emerging Lessons from COVID-19 and Ebola Virus Disease. Pathogens 2022; 11:800. [PMID: 35890044 PMCID: PMC9322689 DOI: 10.3390/pathogens11070800] [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: 05/26/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 02/01/2023] Open
Abstract
Pathogenic viruses with an RNA genome represent a challenge for global human health since they have the tremendous potential to develop into devastating pandemics/epidemics. The management of the recent COVID-19 pandemic was possible to a certain extent only because of the strong foundations laid by the research on previous viral outbreaks, especially Ebola Virus Disease (EVD). A clear understanding of the mechanisms of the host immune response generated upon viral infections is a prime requisite for the development of new therapeutic strategies. Hence, we present here a comparative study of alterations in immune response upon SARS-CoV-2 and Ebola virus infections that illustrate many common features. Vaccination and pregnancy are two important aspects that need to be studied from an immunological perspective. So, we summarize the outcomes and immune responses in vaccinated and pregnant individuals in the context of COVID-19 and EVD. Considering the significance of immunomodulatory approaches in combating both these diseases, we have also presented the state of the art of such therapeutics and prophylactics. Currently, several vaccines against these viruses have been approved or are under clinical trials in various parts of the world. Therefore, we also recapitulate the latest developments in these which would inspire researchers to look for possibilities of developing vaccines against many other RNA viruses. We hope that the similar aspects in COVID-19 and EVD open up new avenues for the development of pan-viral therapies.
Collapse
Affiliation(s)
| | - Aswathy Shailaja
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Aarcha Shanmugha Mary
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur 610105, India;
| | | | - Ambili Savithri
- Department of Biochemistry, Sree Narayana College, Kollam 691001, India;
| | | | | | | | - Maya Madhavan
- Department of Biochemistry, Government College for Women, Thiruvananthapuram 695014, India
| |
Collapse
|
30
|
Lamacchia G, Mazzoni A, Spinicci M, Vanni A, Salvati L, Peruzzi B, Bencini S, Capone M, Carnasciali A, Farahvachi P, Rocca A, Kiros ST, Graziani L, Zammarchi L, Mencarini J, Colao MG, Caporale R, Liotta F, Cosmi L, Rossolini GM, Bartoloni A, Maggi L, Annunziato F. Clinical and Immunological Features of SARS-CoV-2 Breakthrough Infections in Vaccinated Individuals Requiring Hospitalization. J Clin Immunol 2022; 42:1379-1391. [PMID: 35809212 PMCID: PMC9674730 DOI: 10.1007/s10875-022-01325-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/30/2022] [Indexed: 12/29/2022]
Abstract
Background and Purpose Waning immunity and the surge of SARS-CoV-2 variants are responsible for breakthrough infections, i.e., infections in fully vaccinated individuals. Although the majority of vaccinated infected subjects report mild or no symptoms, some others require hospitalization. The clinical and immunological features of vaccinated hospitalized COVID-19 patients are currently unknown. Methods Twenty-nine unvaccinated and 36 vaccinated hospitalized COVID-19 patients were prospectively enrolled and clinical and laboratory data were gathered. Immunophenotyping of leukocytes’ subsets, T and B cell SARS-CoV-2-specific responses were evaluated via flow cytometry. Anti-IFN-α autoantibodies were measured via ELISA. Results Despite vaccinated patients were older and with more comorbidities, unvaccinated subjects showed higher levels of pro-inflammatory markers, more severe disease, and increased mortality rate. Accordingly, they presented significant alterations in the circulating leukocyte composition, typical of severe COVID-19. Vaccinated patients displayed higher levels of anti-Spike IgGs and Spike-specific B cells. Of all participants, survivors showed higher levels of anti-Spike IgGs and Spike-specific CD4+ T cells than non-survivors. At hospital admission, 6 out of 65 patients (9.2%) displayed high serum concentrations of autoantibodies targeting IFN-α. Remarkably, 3 were unvaccinated and eventually died, while the other 3 were vaccinated and survived. Conclusion Despite more severe pre-existing clinical conditions, vaccinated patients have good outcome. A rapid activation of anti-SARS-CoV-2-specific immunity is fundamental for the resolution of the infection. Therefore, prior immunization through vaccination provides a significant contribution to prevention of disease worsening and can even overcome the presence of high-risk factors (i.e., older age, comorbidities, anti-IFN-α autoantibodies). Supplementary Information The online version contains supplementary material available at 10.1007/s10875-022-01325-2.
Collapse
Affiliation(s)
- Giulia Lamacchia
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Anna Vanni
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Benedetta Peruzzi
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Sara Bencini
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Alberto Carnasciali
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Parham Farahvachi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Arianna Rocca
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Seble Tekle Kiros
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Lucia Graziani
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Lorenzo Zammarchi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Jessica Mencarini
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Maria Grazia Colao
- Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Roberto Caporale
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy.,Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy. .,Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
31
|
Rossi CM, Lenti MV, Merli S, Di Sabatino A. Role of IgM Memory B Cells and Spleen Function in COVID-19. Front Immunol 2022; 13:889876. [PMID: 35844543 PMCID: PMC9280616 DOI: 10.3389/fimmu.2022.889876] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
IgM memory B cells, are a peculiar subset of memory B cells, which probably originates in the spleen and outside germinal centers and provide a rapid line of defence against mucosal infections. Their role in counteracting COVID-19 is still elusive but, recent evidence, mainly boosted by studies on spleen function/involvement in COVID-19, seems to support the notion that this subset of memory B cells could exert a protective role against this virus, along with other coronaviruses, particularly in the acute setting of the infection, as outlined by worst clinical outcomes observed in unvaccinated patients with impaired IgM B memory response and spleen function. Herein we critically summarise the current landscape of studies on IgM memory B cells, focusing on the clinical impact of their depletion, by comparing the COVID-19-related splenic dysfunction with other hypo- and asplenic conditions and by adding recent data on follow-up studies and postulate a mechanistic explanation for their reduced numbers. The early detection of an impaired IgM memory B cell response in patients with COVID-19 may contribute to their improved care through different strategies, such as through tailored vaccine strategies, prompt hospital admission and/or administration of anti-infective treatments, thus resulting in an better prognosis, although at present management algorithms are still unavailable. Moreover, further studies with longer follow-up are needed to assess the evolution of COVID-19-associated/exacerbated immune deficit.
Collapse
|
32
|
Wiech M, Chroscicki P, Swatler J, Stepnik D, De Biasi S, Hampel M, Brewinska-Olchowik M, Maliszewska A, Sklinda K, Durlik M, Wierzba W, Cossarizza A, Piwocka K. Remodeling of T Cell Dynamics During Long COVID Is Dependent on Severity of SARS-CoV-2 Infection. Front Immunol 2022; 13:886431. [PMID: 35757700 PMCID: PMC9226563 DOI: 10.3389/fimmu.2022.886431] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022] Open
Abstract
Several COVID-19 convalescents suffer from the post-acute COVID-syndrome (PACS)/long COVID, with symptoms that include fatigue, dyspnea, pulmonary fibrosis, cognitive dysfunctions or even stroke. Given the scale of the worldwide infections, the long-term recovery and the integrative health-care in the nearest future, it is critical to understand the cellular and molecular mechanisms as well as possible predictors of the longitudinal post-COVID-19 responses in convalescent individuals. The immune system and T cell alterations are proposed as drivers of post-acute COVID syndrome. However, despite the number of studies on COVID-19, many of them addressed only the severe convalescents or the short-term responses. Here, we performed longitudinal studies of mild, moderate and severe COVID-19-convalescent patients, at two time points (3 and 6 months from the infection), to assess the dynamics of T cells immune landscape, integrated with patients-reported symptoms. We show that alterations among T cell subsets exhibit different, severity- and time-dependent dynamics, that in severe convalescents result in a polarization towards an exhausted/senescent state of CD4+ and CD8+ T cells and perturbances in CD4+ Tregs. In particular, CD8+ T cells exhibit a high proportion of CD57+ terminal effector cells, together with significant decrease of naïve cell population, augmented granzyme B and IFN-γ production and unresolved inflammation 6 months after infection. Mild convalescents showed increased naïve, and decreased central memory and effector memory CD4+ Treg subsets. Patients from all severity groups can be predisposed to the long COVID symptoms, and fatigue and cognitive dysfunctions are not necessarily related to exhausted/senescent state and T cell dysfunctions, as well as unresolved inflammation that was found only in severe convalescents. In conclusion, the post-COVID-19 functional remodeling of T cells could be seen as a two-step process, leading to distinct convalescent immune states at 6 months after infection. Our data imply that attenuation of the functional polarization together with blocking granzyme B and IFN-γ in CD8+ cells might influence post-COVID alterations in severe convalescents. However, either the search for long COVID predictors or any treatment to prevent PACS and further complications is mandatory in all patients with SARS-CoV-2 infection, and not only in those suffering from severe COVID-19.
Collapse
Affiliation(s)
- Milena Wiech
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Chroscicki
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Julian Swatler
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Dawid Stepnik
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Michal Hampel
- Department of Gastroenterological Surgery and Transplantology, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland
| | - Marta Brewinska-Olchowik
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Maliszewska
- Department of Gastroenterological Surgery and Transplantology, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland
| | - Katarzyna Sklinda
- Department of Radiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marek Durlik
- Department of Gastroenterological Surgery and Transplantology, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland.,Departament of Gastroenterological Surgery and Transplantology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Waldemar Wierzba
- Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland.,University of Humanities and Economics, Lodz, Poland
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy.,National Institute for Cardiovascular Research, Bologna, Italy
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
33
|
Lo Tartaro D, Neroni A, Paolini A, Borella R, Mattioli M, Fidanza L, Quong A, Petes C, Awong G, Douglas S, Lin D, Nieto J, Gozzi L, Franceschini E, Busani S, Nasi M, Mattioli AV, Trenti T, Meschiari M, Guaraldi G, Girardis M, Mussini C, Gibellini L, Cossarizza A, De Biasi S. Molecular and cellular immune features of aged patients with severe COVID-19 pneumonia. Commun Biol 2022; 5:590. [PMID: 35710943 PMCID: PMC9203559 DOI: 10.1038/s42003-022-03537-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022] Open
Abstract
Aging is a major risk factor for developing severe COVID-19, but few detailed data are available concerning immunological changes after infection in aged individuals. Here we describe main immune characteristics in 31 patients with severe SARS-CoV-2 infection who were >70 years old, compared to 33 subjects <60 years of age. Differences in plasma levels of 62 cytokines, landscape of peripheral blood mononuclear cells, T cell repertoire, transcriptome of central memory CD4+ T cells, specific antibodies are reported along with features of lung macrophages. Elderly subjects have higher levels of pro-inflammatory cytokines, more circulating plasmablasts, reduced plasmatic level of anti-S and anti-RBD IgG3 antibodies, lower proportions of central memory CD4+ T cells, more immature monocytes and CD56+ pro-inflammatory monocytes, lower percentages of circulating follicular helper T cells (cTfh), antigen-specific cTfh cells with a less activated transcriptomic profile, lung resident activated macrophages that promote collagen deposition and fibrosis. Our study underlines the importance of inflammation in the response to SARS-CoV-2 and suggests that inflammaging, coupled with the inability to mount a proper anti-viral response, could exacerbate disease severity and the worst clinical outcome in old patients. Patients over the age of 70 show inflammaging and a weaker anti-viral response to SARS-CoV-2, pointing at the immunological changes associated with COVID-19 severity and outcome for aged patients.
Collapse
Affiliation(s)
- Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Lucia Fidanza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Andrew Quong
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Carlene Petes
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Geneve Awong
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Samuel Douglas
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Dongxia Lin
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Jordan Nieto
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Licia Gozzi
- Infectious Diseases Clinics, AOU Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy
| | - Erica Franceschini
- Infectious Diseases Clinics, AOU Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.,Department of Anesthesia and Intensive Care, AOU Policlinico and University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy
| | - Milena Nasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy
| | - Anna Vittoria Mattioli
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.,National Institute for Cardiovascular Research, via Irnerio 48, 40126, Bologna, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124, Modena, Italy
| | - Marianna Meschiari
- Infectious Diseases Clinics, AOU Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, AOU Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy.,Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.,Department of Anesthesia and Intensive Care, AOU Policlinico and University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, AOU Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy.,Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy. .,National Institute for Cardiovascular Research, via Irnerio 48, 40126, Bologna, Italy.
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy.
| |
Collapse
|
34
|
SARS CoV-2 (Delta Variant) Infection Kinetics and Immunopathogenesis in Domestic Cats. Viruses 2022; 14:v14061207. [PMID: 35746678 PMCID: PMC9230585 DOI: 10.3390/v14061207] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 02/04/2023] Open
Abstract
Continued emergence of SARS-CoV-2 variants highlights the critical need for adaptable and translational animal models for acute COVID-19. Limitations to current animal models for SARS CoV-2 (e.g., transgenic mice, non-human primates, ferrets) include subclinical to mild lower respiratory disease, divergence from clinical COVID-19 disease course, and/or the need for host genetic modifications to permit infection. We therefore established a feline model to study COVID-19 disease progression and utilized this model to evaluate infection kinetics and immunopathology of the rapidly circulating Delta variant (B.1.617.2) of SARS-CoV-2. In this study, specific-pathogen-free domestic cats (n = 24) were inoculated intranasally and/or intratracheally with SARS CoV-2 (B.1.617.2). Infected cats developed severe clinical respiratory disease and pulmonary lesions at 4- and 12-days post-infection (dpi), even at 1/10 the dose of previously studied wild-type SARS-CoV-2. Infectious virus was isolated from nasal secretions of delta-variant infected cats in high amounts at multiple timepoints, and viral antigen was co-localized in ACE2-expressing cells of the lungs (pneumocytes, vascular endothelium, peribronchial glandular epithelium) and strongly associated with severe pulmonary inflammation and vasculitis that were more pronounced than in wild-type SARS-CoV-2 infection. RNA sequencing of infected feline lung tissues identified upregulation of multiple gene pathways associated with cytokine receptor interactions, chemokine signaling, and viral protein–cytokine interactions during acute infection with SARS-CoV-2. Weighted correlation network analysis (WGCNA) of differentially expressed genes identified several distinct clusters of dysregulated hub genes that are significantly correlated with both clinical signs and lesions during acute infection. Collectively, the results of these studies help to delineate the role of domestic cats in disease transmission and response to variant emergence, establish a flexible translational model to develop strategies to prevent the spread of SARS-CoV-2, and identify potential targets for downstream therapeutic development.
Collapse
|
35
|
Kudryavtsev I, Rubinstein A, Golovkin A, Kalinina O, Vasilyev K, Rudenko L, Isakova-Sivak I. Dysregulated Immune Responses in SARS-CoV-2-Infected Patients: A Comprehensive Overview. Viruses 2022; 14:1082. [PMID: 35632823 PMCID: PMC9147674 DOI: 10.3390/v14051082] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first detected in humans more than two years ago and caused an unprecedented socio-economic burden on all countries around the world. Since then, numerous studies have attempted to identify various mechanisms involved in the alterations of innate and adaptive immunity in COVID-19 patients, with the ultimate goal of finding ways to correct pathological changes and improve disease outcomes. State-of-the-art research methods made it possible to establish precise molecular mechanisms which the new virus uses to trigger multisystem inflammatory syndrome and evade host antiviral immune responses. In this review, we present a comprehensive analysis of published data that provide insight into pathological changes in T and B cell subsets and their phenotypes, accompanying the acute phase of the SARS-CoV-2 infection. This knowledge might help reveal new biomarkers that can be utilized to recognize case severity early as well as to provide additional objective information on the effective formation of SARS-CoV-2-specific immunity and predict long-term complications of COVID-19, including a large variety of symptoms termed the 'post-COVID-19 syndrome'.
Collapse
Affiliation(s)
- Igor Kudryavtsev
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Artem Rubinstein
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Alexey Golovkin
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.G.); (O.K.)
| | - Olga Kalinina
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.G.); (O.K.)
| | - Kirill Vasilyev
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Larisa Rudenko
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Irina Isakova-Sivak
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| |
Collapse
|
36
|
Romano A, Parrinello NL, Barchitta M, Manuele R, Puglisi F, Maugeri A, Barbato A, Triolo AM, Giallongo C, Tibullo D, La Ferla L, Botta C, Siragusa S, Iacobello C, Montineri A, Volti GL, Agodi A, Palumbo GA, Di Raimondo F. In-vitro NET-osis induced by COVID-19 sera is associated to severe clinical course in not vaccinated patients and immune-dysregulation in breakthrough infection. Sci Rep 2022; 12:7237. [PMID: 35508575 PMCID: PMC9065667 DOI: 10.1038/s41598-022-11157-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 04/18/2022] [Indexed: 11/18/2022] Open
Abstract
Since neutrophil extracellular traps formation (NET-osis) can be assessed indirectly by treating healthy neutrophils with blood-derived fluids from patients and then measuring the NETs response, we designed a pilot study to convey high-dimensional cytometry of peripheral blood immune cells and cytokines, combined with clinical features, to understand if NET-osis assessment could be included in the immune risk profiling to early prediction of clinical patterns, disease severity, and viral clearance at 28 days in COVID-19 patients. Immune cells composition of peripheral blood, cytokines concentration and in-vitro NETosis were detected in peripheral blood of 41 consecutive COVID-19 inpatients, including 21 mild breakthrough infections compared to 20 healthy donors, matched for sex and age. Major immune dysregulation in peripheral blood in not-vaccinated COVID-19 patients compared to healthy subjects included: a significant reduction of percentage of unswitched memory B-cells and transitional B-cells; loss of naïve CD3+CD4+CD45RA+ and CD3+CD8+CD45RA+ cells, increase of IL-1β, IL-17A and IFN-γ. Myeloid compartment was affected as well, due to the increase of classical (CD14++CD16−) and intermediate (CD14++CD16+) monocytes, overexpressing the activation marker CD64, negatively associated to the absolute counts of CD8+ CD45R0+ cells, IFN-γ and IL-6, and expansion of monocytic-like myeloid derived suppressor cells. In not-vaccinated patients who achieved viral clearance by 28 days we found at hospital admission lower absolute counts of effector cells, namely CD8+T cells, CD4+ T-cells and CD4+CD45RO+ T cells. Percentage of in-vitro NET-osis induced by patients’ sera and NET-osis density were progressively higher in moderate and severe COVID-19 patients than in mild disease and controls. The percentage of in-vitro induced NET-osis was positively associated to circulating cytokines IL-1β, IFN-γ and IL-6. In breakthrough COVID-19 infections, characterized by mild clinical course, we observed increased percentage of in-vitro NET-osis, higher CD4+ CD45RO+ and CD8+ CD45RO+ T cells healthy or mild-COVID-19 not-vaccinated patients, reduced by 24 h of treatment with ACE inhibitor ramipril. Taken together our data highlight the role of NETs in orchestrating the complex immune response to SARS-COV-2, that should be considered in a multi-target approach for COVID-19 treatment.
Collapse
Affiliation(s)
- Alessandra Romano
- Division of Hematology, Azienda Policlinico-Rodolico San Marco, Catania, Italy. .,Dipartimento di Chirurgia Generale e Specialità Medico Chirurgiche, University of Catania, Catania, Italy.
| | | | - Martina Barchitta
- Department of Medical and Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, 95123, Catania, Italy
| | - Rosy Manuele
- U.O.C. di Malattie Infettive, Azienda Policlinico-Rodolico San Marco, Catania, Italy
| | - Fabrizio Puglisi
- Division of Hematology, Azienda Policlinico-Rodolico San Marco, Catania, Italy
| | - Andrea Maugeri
- Department of Medical and Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, 95123, Catania, Italy
| | - Alessandro Barbato
- Division of Hematology, Azienda Policlinico-Rodolico San Marco, Catania, Italy
| | - Anna Maria Triolo
- Division of Hematology, Azienda Policlinico-Rodolico San Marco, Catania, Italy
| | - Cesarina Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, 95123, Catania, Italy
| | - Daniele Tibullo
- Dipartimento di Scienze Biomediche e Biotecnologiche, University of Catania, Catania, Italy
| | - Lucia La Ferla
- U.O.C. di Malattie Infettive, Azienda Cannizzaro, Catania, Italy
| | - Ciro Botta
- Division of Hematology, Università degli Studi di Palermo, Palermo, Italy
| | - Sergio Siragusa
- Division of Hematology, Università degli Studi di Palermo, Palermo, Italy
| | | | - Arturo Montineri
- U.O.C. di Malattie Infettive, Azienda Policlinico-Rodolico San Marco, Catania, Italy
| | - Giovanni Li Volti
- Dipartimento di Scienze Biomediche e Biotecnologiche, University of Catania, Catania, Italy
| | - Antonella Agodi
- Department of Medical and Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, 95123, Catania, Italy
| | - Giuseppe Alberto Palumbo
- Division of Hematology, Azienda Policlinico-Rodolico San Marco, Catania, Italy.,Department of Medical and Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, 95123, Catania, Italy
| | - Francesco Di Raimondo
- Division of Hematology, Azienda Policlinico-Rodolico San Marco, Catania, Italy.,Dipartimento di Chirurgia Generale e Specialità Medico Chirurgiche, University of Catania, Catania, Italy
| |
Collapse
|
37
|
Kuchroo M, Huang J, Wong P, Grenier JC, Shung D, Tong A, Lucas C, Klein J, Burkhardt DB, Gigante S, Godavarthi A, Rieck B, Israelow B, Simonov M, Mao T, Oh JE, Silva J, Takahashi T, Odio CD, Casanovas-Massana A, Fournier J, Farhadian S, Dela Cruz CS, Ko AI, Hirn MJ, Wilson FP, Hussin JG, Wolf G, Iwasaki A, Krishnaswamy S. Multiscale PHATE identifies multimodal signatures of COVID-19. Nat Biotechnol 2022; 40:681-691. [PMID: 35228707 PMCID: PMC10015653 DOI: 10.1038/s41587-021-01186-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 12/10/2021] [Indexed: 01/21/2023]
Abstract
As the biomedical community produces datasets that are increasingly complex and high dimensional, there is a need for more sophisticated computational tools to extract biological insights. We present Multiscale PHATE, a method that sweeps through all levels of data granularity to learn abstracted biological features directly predictive of disease outcome. Built on a coarse-graining process called diffusion condensation, Multiscale PHATE learns a data topology that can be analyzed at coarse resolutions for high-level summarizations of data and at fine resolutions for detailed representations of subsets. We apply Multiscale PHATE to a coronavirus disease 2019 (COVID-19) dataset with 54 million cells from 168 hospitalized patients and find that patients who die show CD16hiCD66blo neutrophil and IFN-γ+ granzyme B+ Th17 cell responses. We also show that population groupings from Multiscale PHATE directly fed into a classifier predict disease outcome more accurately than naive featurizations of the data. Multiscale PHATE is broadly generalizable to different data types, including flow cytometry, single-cell RNA sequencing (scRNA-seq), single-cell sequencing assay for transposase-accessible chromatin (scATAC-seq), and clinical variables.
Collapse
Affiliation(s)
- Manik Kuchroo
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Jessie Huang
- Department of Computer Science, Yale University, New Haven, CT, USA
| | - Patrick Wong
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | | | - Dennis Shung
- Department of Medicine, Yale University, New Haven, CT, USA
| | - Alexander Tong
- Department of Computer Science, Yale University, New Haven, CT, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Jon Klein
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | | | - Scott Gigante
- Computational Biology, Bioinformatics Program, Yale University, New Haven, CT, USA
| | | | - Bastian Rieck
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Benjamin Israelow
- Department of Immunobiology, Yale University, New Haven, CT, USA
- Section of Infectious Diseases, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Tianyang Mao
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Ji Eun Oh
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Julio Silva
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | | | - Camila D Odio
- Department of Medicine, Yale University, New Haven, CT, USA
| | - Arnau Casanovas-Massana
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - John Fournier
- Section of Infectious Diseases, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Shelli Farhadian
- Section of Infectious Diseases, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Charles S Dela Cruz
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Medicine, West Haven Connecticut Veterans Affairs Medical Center, West Haven, CT, USA
| | - Albert I Ko
- Section of Infectious Diseases, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Matthew J Hirn
- Department of Mathematics, Michigan State University, East Lansing, MI, USA
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - F Perry Wilson
- Clinical and Translational Research Accelerator, Department of Medicine, Yale University, New Haven, CT, USA
| | - Julie G Hussin
- Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Guy Wolf
- Mila - Quebec AI institute, Montreal, Quebec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montreal, Quebec, Canada
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Smita Krishnaswamy
- Department of Computer Science, Yale University, New Haven, CT, USA.
- Department of Genetics, Yale University, New Haven, CT, USA.
| |
Collapse
|
38
|
Gibellini L, De Biasi S, Meschiari M, Gozzi L, Paolini A, Borella R, Mattioli M, Lo Tartaro D, Fidanza L, Neroni A, Busani S, Girardis M, Guaraldi G, Mussini C, Cozzi-Lepri A, Cossarizza A. Plasma Cytokine Atlas Reveals the Importance of TH2 Polarization and Interferons in Predicting COVID-19 Severity and Survival. Front Immunol 2022; 13:842150. [PMID: 35386702 PMCID: PMC8979161 DOI: 10.3389/fimmu.2022.842150] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/21/2022] [Indexed: 12/11/2022] Open
Abstract
Although it is now widely accepted that host inflammatory response contributes to COVID-19 immunopathogenesis, the pathways and mechanisms driving disease severity and clinical outcome remain poorly understood. In the effort to identify key soluble mediators that characterize life-threatening COVID-19, we quantified 62 cytokines, chemokines and other factors involved in inflammation and immunity in plasma samples, collected at hospital admission, from 80 hospitalized patients with severe COVID-19 disease who were stratified on the basis of clinical outcome (mechanical ventilation or death by day 28). Our data confirm that age, as well as neutrophilia, lymphocytopenia, procalcitonin, D-dimer and lactate dehydrogenase are strongly associated with the risk of fatal COVID-19. In addition, we found that cytokines related to TH2 regulations (IL-4, IL-13, IL-33), cell metabolism (lep, lep-R) and interferons (IFNα, IFNβ, IFNγ) were also predictive of life-threatening COVID-19.
Collapse
Affiliation(s)
- Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Marianna Meschiari
- Infectious Diseases Clinics, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Licia Gozzi
- Infectious Diseases Clinics, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Lucia Fidanza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Stefano Busani
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Girardis
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Cozzi-Lepri
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation, Institute for Global Health, London, United Kingdom
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy.,National Institute for Cardiovascular Research, Bologna, Italy
| |
Collapse
|
39
|
Bartlett ML, Suwanmanee S, Peart Akindele N, Ghimire S, Chan AK, Guo C, Gould SJ, Cox AL, Griffin DE. Continued Virus-Specific Antibody-Secreting Cell Production, Avidity Maturation and B Cell Evolution in Patients Hospitalized with COVID-19. Viral Immunol 2022; 35:259-272. [PMID: 35285743 PMCID: PMC9063170 DOI: 10.1089/vim.2021.0191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Understanding the development and sustainability of the virus-specific protective immune response to infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) remains incomplete with respect to the appearance and disappearance of virus-specific antibody-secreting cells (ASCs) in circulation. Therefore, we performed cross-sectional and longitudinal analyses of peripheral blood mononuclear cells and plasma collected from 55 hospitalized patients up to 4 months after onset of COVID-19 symptoms. Spike (S)- and nucleocapsid (N)-specific IgM and IgG ASCs appeared within 2 weeks accompanied by flow cytometry increases in double negative plasmablasts consistent with a rapid extrafollicular B cell response. Total and virus-specific IgM and IgG ASCs peaked at 3-4 weeks and were still being produced at 3-4 months accompanied by increasing antibody avidity consistent with a slower germinal center B cell response. N-specific ASCs were produced for longer than S-specific ASCs and avidity maturation was greater for antibody to N than S. Patients with more severe disease produced more S-specific IgM and IgG ASCs than those with mild disease and had higher levels of N- and S-specific antibody. Women had more B cells in circulation than men and produced more S-specific IgA and IgG and N-specific IgG ASCs. Flow cytometry analysis of B cell phenotypes showed an increase in circulating B cells at 4-6 weeks with decreased percentages of switched and unswitched memory B cells. These data indicate ongoing antigen-specific stimulation, maturation, and production of ASCs for several months after onset of symptoms in patients hospitalized with COVID-19.
Collapse
Affiliation(s)
- Maggie L. Bartlett
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - San Suwanmanee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Nadine Peart Akindele
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shristi Ghimire
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andy K.P. Chan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Chenxu Guo
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stephen J. Gould
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrea L. Cox
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Diane E. Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
40
|
Chai M, Guo Y, Yang L, Li J, Liu S, Chen L, Shen Y, Yang Y, Wang Y, Xu L, Yu C. A high-throughput single cell-based antibody discovery approach against the full-length SARS-CoV-2 spike protein suggests a lack of neutralizing antibodies targeting the highly conserved S2 domain. Brief Bioinform 2022; 23:6561436. [PMID: 35362510 DOI: 10.1093/bib/bbac070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/25/2022] [Accepted: 02/12/2022] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 pandemic continues globally with a growing number of infections, but there are currently no effective antibody drugs against the virus. In addition, 90% amino acid sequence identity between the S2 subunit of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and SARS-CoV S proteins attracts us to examine S2-targeted cross-neutralizing antibodies that are not yet well defined. We therefore immunized RenMab mice with the full-length S protein and constructed a high-throughput antibody discovery method based on single-cell sequencing technology to isolate SARS-CoV-2 S-targeted neutralizing antibodies and cross-neutralizing antibodies against the S2 region of SARS-CoV-2/SARS-CoV S. Diversity of antibody sequences in RenMab mice and consistency in B-cell immune responses between RenMab mice and humans enabled screening of fully human virus-neutralizing antibodies. From all the frequency >1 paired clonotypes obtained from single-cell V(D)J sequencing, 215 antibodies with binding affinities were identified and primarily bound S2. However, only two receptor-binding domain-targeted clonotypes had neutralizing activity against SARS-CoV-2. Moreover, 5' single-cell RNA sequencing indicated that these sorted splenic B cells are mainly plasmablasts, germinal center (GC)-dependent memory B-cells and GC B-cells. Among them, plasmablasts and GC-dependent memory B-cells were considered the most significant possibility of producing virus-specific antibodies. Altogether, using a high-throughput single cell-based antibody discovery approach, our study highlighted the challenges of developing S2-binding neutralizing antibodies against SARS-CoV-2 and provided a novel direction for the enrichment of antigen-specific B-cells.
Collapse
Affiliation(s)
- Mengya Chai
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yajuan Guo
- Beijing Biocytogen Co., Ltd, Beijing 101111, China
| | - Liu Yang
- Beijing Biocytogen Co., Ltd, Beijing 101111, China
| | - Jianhui Li
- Beijing Biocytogen Co., Ltd, Beijing 101111, China
| | - Shuo Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing 102629, China
| | - Lei Chen
- Beijing Biocytogen Co., Ltd, Beijing 101111, China
| | - Yuelei Shen
- Beijing Biocytogen Co., Ltd, Beijing 101111, China
| | - Yi Yang
- Beijing Biocytogen Co., Ltd, Beijing 101111, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing 102629, China
| | - Lida Xu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Changyuan Yu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
41
|
Garcia-Gasalla M, Berman-Riu M, Pons J, Rodríguez A, Iglesias A, Martínez-Pomar N, Llompart-Alabern I, Riera M, Ferré Beltrán A, Figueras-Castilla A, Murillas J, Ferrer JM. Hyperinflammatory State and Low T1 Adaptive Immune Response in Severe and Critical Acute COVID-19 Patients. Front Med (Lausanne) 2022; 9:828678. [PMID: 35425776 PMCID: PMC9002349 DOI: 10.3389/fmed.2022.828678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/07/2022] [Indexed: 01/08/2023] Open
Abstract
Background A better understanding of COVID-19 immunopathology is needed to identify the most vulnerable patients and improve treatment options. Objective We aimed to identify immune system cell populations, cytokines, and inflammatory markers related to severity in COVID-19. Methods 139 hospitalized patients with COVID-19-58 mild/moderate and 81 severe/critical-and 74 recovered patients were included in a prospective longitudinal study. Clinical data and blood samples were obtained on admission for laboratory markers, cytokines, and lymphocyte subsets study. In the recovered patients, lymphocyte subsets were analyzed 8-12 weeks after discharge. Results A National Early Warning Score 2 >2 (OR:41.4; CI:10.38-167.0), ferritin >583 pg/mL (OR:16.3; CI: 3.88-69.9), neutrophil/lymphocyte ratio >3 (OR: 3.5; CI: 1.08-12.0), sIL-2rα (sCD25) >512 pg/mL (OR: 3.3; CI: 1.48-7.9), IL-1Ra >94 pg/mL (OR: 3.2; IC: 1.4-7.3), and IL-18 >125 pg/mL (OR: 2.4; CI: 1.1-5.0) were associated with severe/critical COVID-19 in the multivariate models used. Lower absolute values of CD3, CD4, CD8, and CD19 lymphocytes together with higher frequencies of NK cells, a CD4 and CD8 activated (CD38+HLA-DR+) memory T cell and effector memory CD45RA+ (EMRA) phenotype, and lower T regulatory cell frequencies were found in severe/critical patients relative to mild/moderate and recovered COVID-19 patients. A significant reduction in Th1, Tfh1, and Tc1 with higher Th2, Tfh2, Tc2, and plasma cell frequencies was found in the most severe cases. Conclusion A characteristic hyperinflammatory state with significantly elevated neutrophil/lymphocyte ratio and ferritin, IL-1Ra, sIL-2rα, and IL-18 levels together with a "low T1 lymphocyte signature" was found in severe/critical COVID-19 patients.
Collapse
Affiliation(s)
- Mercedes Garcia-Gasalla
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - María Berman-Riu
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Jaime Pons
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Centro de Investigación Biomedica en Red (CIBER) de Enfermedades Respiratorias, Hospital Universitari Son Espases, Palma, Spain
| | - Adrián Rodríguez
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Department of Internal Medicine, Hospital Universitari Son Llàtzer, Palma, Spain
| | - Amanda Iglesias
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Centro de Investigación Biomedica en Red (CIBER) de Enfermedades Respiratorias, Hospital Universitari Son Espases, Palma, Spain
| | - Natalia Martínez-Pomar
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
| | - Isabel Llompart-Alabern
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Análisis Clínicos, Hospital Universitari Son Espases, Palma, Spain
| | - Melchor Riera
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Adrián Ferré Beltrán
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain
| | | | - Javier Murillas
- Department of Internal Medicine, Hospital Universitari Son Espases, Palma, Spain
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
| | - Joana M. Ferrer
- Balearic Islands Health Research Institute (IdISBa), Palma, Spain
- Department of Immunology, Hospital Universitari Son Espases, Palma, Spain
- Centro de Investigación Biomedica en Red (CIBER) de Enfermedades Respiratorias, Hospital Universitari Son Espases, Palma, Spain
| |
Collapse
|
42
|
Shafqat A, Shafqat S, Salameh SA, Kashir J, Alkattan K, Yaqinuddin A. Mechanistic Insights Into the Immune Pathophysiology of COVID-19; An In-Depth Review. Front Immunol 2022; 13:835104. [PMID: 35401519 PMCID: PMC8989408 DOI: 10.3389/fimmu.2022.835104] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), which causes coronavirus-19 (COVID-19), has caused significant morbidity and mortality globally. In addition to the respiratory manifestations seen in severe cases, multi-organ pathologies also occur, making management a much-debated issue. In addition, the emergence of new variants can potentially render vaccines with a relatively limited utility. Many investigators have attempted to elucidate the precise pathophysiological mechanisms causing COVID-19 respiratory and systemic disease. Spillover of lung-derived cytokines causing a cytokine storm is considered the cause of systemic disease. However, recent studies have provided contradictory evidence, whereby the extent of cytokine storm is insufficient to cause severe illness. These issues are highly relevant, as management approaches considering COVID-19 a classic form of acute respiratory distress syndrome with a cytokine storm could translate to unfounded clinical decisions, detrimental to patient trajectory. Additionally, the precise immune cell signatures that characterize disease of varying severity remain contentious. We provide an up-to-date review on the immune dysregulation caused by COVID-19 and highlight pertinent discussions in the scientific community. The response from the scientific community has been unprecedented regarding the development of highly effective vaccines and cutting-edge research on novel therapies. We hope that this review furthers the conversations held by scientists and informs the aims of future research projects, which will potentially further our understanding of COVID-19 and its immune pathogenesis.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | | | - Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Center of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | |
Collapse
|
43
|
Siu JHY, Pitcher MJ, Tull TJ, Velounias RL, Guesdon W, Montorsi L, Mahbubani KT, Ellis R, Dhami P, Todd K, Kadolsky UD, Kleeman M, D'Cruz DP, Saeb-Parsy K, Bemark M, Pettigrew GJ, Spencer J. Two subsets of human marginal zone B cells resolved by global analysis of lymphoid tissues and blood. Sci Immunol 2022; 7:eabm9060. [PMID: 35302862 DOI: 10.1126/sciimmunol.abm9060] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
B cells generate antibodies that are essential for immune protection, but their subgroups are poorly defined. Here, we perform undirected deep profiling of B cells in matched human lymphoid tissues from deceased transplant organ donors and blood. In addition to identifying unanticipated features of tissue-based B cell differentiation, we resolve two subsets of marginal zone B (MZB) cells differing in cell surface and transcriptomic profiles, clonal relationships to other subsets, enrichment of genes in the NOTCH pathway, distribution bias within splenic marginal zone microenvironment, and immunoglobulin repertoire diversity and hypermutation frequency. Each subset is present in spleen, gut-associated lymphoid tissue, mesenteric lymph nodes, and blood. MZB cells and the lineage from which they are derived are depleted in lupus nephritis. Here, we show that this depletion is of only one MZB subset. The other remains unchanged as a proportion of total B cells compared with health. Thus, it is important to factor MZB cell heterogeneity into studies of human B cell responses and pathology.
Collapse
Affiliation(s)
- Jacqueline H Y Siu
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Michael J Pitcher
- School of Immunology and Microbial Sciences, King's College London, Guy's Campus, London SE1 9RT, UK
| | - Thomas J Tull
- School of Immunology and Microbial Sciences, King's College London, Guy's Campus, London SE1 9RT, UK
| | - Rebekah L Velounias
- School of Immunology and Microbial Sciences, King's College London, Guy's Campus, London SE1 9RT, UK
| | - William Guesdon
- School of Immunology and Microbial Sciences, King's College London, Guy's Campus, London SE1 9RT, UK
| | - Lucia Montorsi
- School of Cancer Sciences, King's College London, Guy's Campus, London, UK.,Cancer Systems Biology Laboratory, Francis Crick Institute, London, UK
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Richard Ellis
- NIHR Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas NHS Foundation Trust, Guy's Hospital, London SE1 9RT, UK
| | - Pawan Dhami
- NIHR Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas NHS Foundation Trust, Guy's Hospital, London SE1 9RT, UK
| | - Katrina Todd
- NIHR Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas NHS Foundation Trust, Guy's Hospital, London SE1 9RT, UK
| | - Ulrich D Kadolsky
- NIHR Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas NHS Foundation Trust, Guy's Hospital, London SE1 9RT, UK
| | - Michelle Kleeman
- NIHR Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas NHS Foundation Trust, Guy's Hospital, London SE1 9RT, UK
| | - David P D'Cruz
- School of Immunology and Microbial Sciences, King's College London, Guy's Campus, London SE1 9RT, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Mats Bemark
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE 405 30 Gothenburg, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gavin J Pettigrew
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Jo Spencer
- School of Immunology and Microbial Sciences, King's College London, Guy's Campus, London SE1 9RT, UK
| |
Collapse
|
44
|
Park U, Cho NH. Protective and pathogenic role of humoral responses in COVID-19. J Microbiol 2022; 60:268-275. [PMID: 35235178 PMCID: PMC8890013 DOI: 10.1007/s12275-022-2037-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 12/24/2022]
Abstract
Since the advent of SARS-CoV-2 in Dec. 2019, the global endeavor to identify the pathogenic mechanism of COVID-19 has been ongoing. Although humoral immunity including neutralizing activity play an important role in protection from the viral pathogen, dysregulated antibody responses may be associated with the pathogenic progression of COVID-19, especially in high-risk individuals. In addition, SARS-CoV-2 spike-specific antibodies acquired by prior infection or vaccination act as immune pressure, driving continuous population turnover by selecting for antibody-escaping mutations. Here, we review accumulating knowledge on the potential role of humoral immune responses in COVID-19, primarily focusing on their beneficial and pathogenic properties. Understanding the multifaceted regulatory mechanisms of humoral responses during SARS-CoV-2 infection can help us to develop more effective therapeutics, as well as protective measures against the ongoing pandemic.
Collapse
Affiliation(s)
- Uni Park
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, 03080, Republic of Korea.
- Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea.
- Wide River Institute of Immunology, Seoul National University, Hongcheon, 25159, Republic of Korea.
| |
Collapse
|
45
|
Atanasov P, Ivanova S, Kobakova Y, Moneva-Sakelarieva M, Obreshkova D, Petkova V, Laleva-Jordanova P. Immunogenesis in patients with medium and severe coronavirus infection – dynamics in different age groups. PHARMACIA 2022. [DOI: 10.3897/pharmacia.69.e81063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The results of a one-year prospective study, during which the process of immunogenesis in patients over 18 years of age with moderate and severe coronavirus infection was monitored and analyzed in clinical and paraclinical (clinical laboratory) aspects, are summarized and presented.
The study included 2683 patients, all treated in the Clinic of Internal Diseases at the University Multiprofile Hospital for Active Treatment and Emergency Medicine “N. I. Pirogov” EAD, Sofia for the period from April 2020 to December 2020. Patients were followed for one year after recovering from moderate to severe coronavirus infection. Patients are grouped into four age categories as follows: 18–45 years; 46–65 years; 66–80 years and over 80 years.
The results of our study show that during the study period in 97% of patients the level of anti-SARS-CoV2, rose and in the remaining three percent it was flat or followed by subsequent waning (in less than 1% of patients), but does not reach critically low levels (i. e. below the positivity conditional threshold). The level of IgG reached a peak and then waned, but on the other hand, as mentioned above, the amount of Ig-Total tested shows a significant increase. This trend is observed in all age groups, with a difference in the level of IgG and Ig-Total depending on age.
The results of the additional screening in the target period in terms of virulence and virus segregation, categorically rule out the suspicion of the presence of “silent spreader”. During the follow-up period, no patients were re-hospitalized due to recurrence of Coronavirus infection (re-infection and illness).
Collapse
|
46
|
Yousefi Z, Taheri N, Dargahi M, Chaman R, Binesh E, Emamian MH, Jafari R. Long-Term Persistence of Anti-SARS-COV-2 IgG Antibodies. Curr Microbiol 2022; 79:96. [PMID: 35150319 PMCID: PMC8853199 DOI: 10.1007/s00284-022-02800-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 02/02/2022] [Indexed: 12/17/2022]
Abstract
Antibodies against severe acute respiratory syndrome coronavirus-2 (Anti-SARS-COV-2) can be detected in patients with COVID-19 in 7 to 10 days post onset of symptoms (POS). However, there is no firm evidence of the long-term persistence of these antibodies in recovered COVID-19 patients. Therefore, this study aimed to evaluate the stability of anti-SARS-COV-2 IgG in recovered COVID-19 patients in a 15-month follow-up testing. Thirty hospitalized patients with real-time PCR-confirmed SARS-COV-2 infections were included in the study and five serum samples (1st, 2nd, 3rd, 4th, and 5th) were collected from each participant. The serum levels of N and S specific anti-SARS-COV-2 IgG and IgM antibodies were evaluated by the immunoassay technique at the same time. To determine the correlation between levels of anti-SARS-CoV-2 IgG/IgM with severity of disease, neutrophil-to-lymphocyte ratio (NLR %), and the serum levels of C-reactive protein were evaluated using an automated analyzer and turbidimetry assays, respectively. The mean serum level of anti-SARS-CoV-2 IgG antibody was at the highest level up to 90 days and then decreased significantly 1 year POS (P < 0.0001). However, it was still detectable in a 15-month follow-up testing. There were no significant differences in the mean levels of IgG antibody in patients with mild, moderate, and severe diseases. The results from this study suggest that the titer of anti-SARS-COV-2 IgG antibody is detectable at high levels up to 3 months and then decreases over time. However, these antibodies can be reliably detected in up to 15 months, and they may persist for a long time.
Collapse
Affiliation(s)
- Zahra Yousefi
- School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Niloofar Taheri
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Motahareh Dargahi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Reza Chaman
- Department of Community Medicine, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ehsan Binesh
- Clinical Research Development Unit, Imam Hossein Hospital, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Hassan Emamian
- Ophthalmic Epidemiology Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Reza Jafari
- School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran.
| |
Collapse
|
47
|
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
|
48
|
Xia J, Chen S, Li Y, Li H, Gan M, Wu J, Prohaska CC, Bai Y, Gao L, Gu L, Zhang D. Immune Response Is Key to Genetic Mechanisms of SARS-CoV-2 Infection With Psychiatric Disorders Based on Differential Gene Expression Pattern Analysis. Front Immunol 2022; 13:798538. [PMID: 35185890 PMCID: PMC8854505 DOI: 10.3389/fimmu.2022.798538] [Citation(s) in RCA: 2] [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: 10/20/2021] [Accepted: 01/13/2022] [Indexed: 12/11/2022] Open
Abstract
Existing evidence demonstrates that coronavirus disease 2019 (COVID-19) leads to psychiatric illness, despite its main clinical manifestations affecting the respiratory system. People with mental disorders are more susceptible to COVID-19 than individuals without coexisting mental health disorders, with significantly higher rates of severe illness and mortality in this population. The incidence of new psychiatric diagnoses after infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is also remarkably high. SARS-CoV-2 has been reported to use angiotensin-converting enzyme-2 (ACE2) as a receptor for infecting susceptible cells and is expressed in various tissues, including brain tissue. Thus, there is an urgent need to investigate the mechanism linking psychiatric disorders to COVID-19. Using a data set of peripheral blood cells from patients with COVID-19, we compared this to data sets of whole blood collected from patients with psychiatric disorders and used bioinformatics and systems biology approaches to identify genetic links. We found a large number of overlapping immune-related genes between patients infected with SARS-CoV-2 and differentially expressed genes of bipolar disorder (BD), schizophrenia (SZ), and late-onset major depressive disorder (LOD). Many pathways closely related to inflammatory responses, such as MAPK, PPAR, and TGF-β signaling pathways, were observed by enrichment analysis of common differentially expressed genes (DEGs). We also performed a comprehensive analysis of protein-protein interaction network and gene regulation networks. Chemical-protein interaction networks and drug prediction were used to screen potential pharmacologic therapies. We hope that by elucidating the relationship between the pathogenetic processes and genetic mechanisms of infection with SARS-CoV-2 with psychiatric disorders, it will lead to innovative strategies for future research and treatment of psychiatric disorders linked to COVID-19.
Collapse
Affiliation(s)
- Jing Xia
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Shuhan Chen
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Yaping Li
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Hua Li
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Minghong Gan
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Jiashuo Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Clare Colette Prohaska
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Yang Bai
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lu Gao
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Li Gu
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| | - Dongfang Zhang
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
49
|
Boraldi F, Lofaro FD, Cossarizza A, Quaglino D. The "Elastic Perspective" of SARS-CoV-2 Infection and the Role of Intrinsic and Extrinsic Factors. Int J Mol Sci 2022; 23:ijms23031559. [PMID: 35163482 PMCID: PMC8835950 DOI: 10.3390/ijms23031559] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/20/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023] Open
Abstract
Elastin represents the structural component of the extracellular matrix providing elastic recoil to tissues such as skin, blood vessels and lungs. Elastogenic cells secrete soluble tropoelastin monomers into the extracellular space where these monomers associate with other matrix proteins (e.g., microfibrils and glycoproteins) and are crosslinked by lysyl oxidase to form insoluble fibres. Once elastic fibres are formed, they are very stable, highly resistant to degradation and have an almost negligible turnover. However, there are circumstances, mainly related to inflammatory conditions, where increased proteolytic degradation of elastic fibres may lead to consequences of major clinical relevance. In severely affected COVID-19 patients, for instance, the massive recruitment and activation of neutrophils is responsible for the profuse release of elastases and other proteolytic enzymes which cause the irreversible degradation of elastic fibres. Within the lungs, destruction of the elastic network may lead to the permanent impairment of pulmonary function, thus suggesting that elastases can be a promising target to preserve the elastic component in COVID-19 patients. Moreover, intrinsic and extrinsic factors additionally contributing to damaging the elastic component and to increasing the spread and severity of SARS-CoV-2 infection are reviewed.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.B.); (F.D.L.)
| | - Francesco Demetrio Lofaro
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.B.); (F.D.L.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.B.); (F.D.L.)
- Correspondence:
| |
Collapse
|
50
|
Pan T, Cao G, Tang E, Zhao Y, Penaloza-MacMaster P, Fang Y, Huang J. A single-cell atlas reveals shared and distinct immune responses and metabolism during SARS-CoV-2 and HIV-1 infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.01.10.475725. [PMID: 35043114 PMCID: PMC8764725 DOI: 10.1101/2022.01.10.475725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
UNLABELLED SARS-CoV-2 and HIV-1 are RNA viruses that have killed millions of people worldwide. Understanding the similarities and differences between these two infections is critical for understanding disease progression and for developing effective vaccines and therapies, particularly for 38 million HIV-1 + individuals who are vulnerable to SARS-CoV-2 co-infection. Here, we utilized single-cell transcriptomics to perform a systematic comparison of 94,442 PBMCs from 7 COVID-19 and 9 HIV-1 + patients in an integrated immune atlas, in which 27 different cell types were identified using an accurate consensus single-cell annotation method. While immune cells in both cohorts show shared inflammation and disrupted mitochondrial function, COVID-19 patients exhibit stronger humoral immunity, broader IFN-I signaling, elevated Rho GTPase and mTOR pathway activities, and downregulated mitophagy. Our results elucidate transcriptional signatures associated with COVID-19 and HIV-1 that may reveal insights into fundamental disease biology and potential therapeutic targets to treat these viral infections. HIGHLIGHTS COVID-19 and HIV-1 + patients show disease-specific inflammatory immune signatures COVID-19 patients show more productive humoral responses than HIV-1 + patients SARS-CoV-2 elicits more enriched IFN-I signaling relative to HIV-IDivergent, impaired metabolic programs distinguish SARS-CoV-2 and HIV-1 infections.
Collapse
|