1
|
Bain V, Correa-Silva S, Matsuo OM, Silva-Avelar I, Zheng Y, Rangel-Santos A, Souza Gonçalves G, de Toledo Fink T, Suguita P, Caires O A Ferreira J, Fernandes Ferreira AE, Litvinov N, Andrade Macaferri da Fonseca F, Astley C, Martins F, Carneiro Sampaio M, de Sousa Marques HH, da Silva CAA, Palmeira P, Bádue Pereira MF. Comprehensive Characterization of Innate and Adaptive Immune Profiles in the Pediatric COVID-19 Convalescent Phase. Pediatr Infect Dis J 2025; 44:557-563. [PMID: 40067781 DOI: 10.1097/inf.0000000000004713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
BACKGROUND Knowledge of the effect of SARS-CoV-2 on the innate and adaptive immune responses of children is currently lacking. We investigated the immune profile of recovered pediatric patients 3 to 11 weeks after acute COVID-19. METHODS Children who were previously healthy or had a preexisting chronic disease and had a positive reverse transcription polymerase chain reaction/serology were enrolled (n=23). The control group was composed of 25 patients without COVID-19 paired by age, sex and baseline chronic conditions. We performed immunophenotyping, hematologic and inflammatory markers analysis, cytokines and T-cell receptor excision circle (TREC) quantifications. RESULTS Most COVID-19 convalescent pediatric patients (COVID-19 CPP) had chronic conditions (73.9%), as well as 80% of the controls. Five children developed multisystem inflammatory syndrome in children. COVID-19 CPP had higher lymphocyte numbers than controls due to an increase in CD4+ T cells. Naive, effector memory (EM) reexpressing CD45RA T cells and follicular CD4+ T cells, as well as TRECs and HLA-DR+ and CD38+CD4+ activated T lymphocytes, were increased in those patients. EM2 and EM3 CD4+ T cells, EM2 CD8+ T cells and memory B cells were elevated in the COVID-19 CPP group. Numbers of neutrophils, monocytes and natural killer cells were equivalent but with increased activation in the recovered patients. CONCLUSIONS In the short-term following infection, COVID-19-recovered patients show persistent activation profiles in phagocytes, T-cell subtypes and natural killer cells. Meanwhile, increased production of lymphocytes, TRECs and naive T cells suggests immune response recovery, even in immunosuppressed patients and children with comorbidities. The clinical implications of these findings should be further studied.
Collapse
Affiliation(s)
- Vera Bain
- From the Departamento de Pediatria, Faculdade de Medicina FMUSP
| | | | - Olivia M Matsuo
- From the Departamento de Pediatria, Faculdade de Medicina FMUSP
| | | | - Yingying Zheng
- From the Departamento de Pediatria, Faculdade de Medicina FMUSP
| | - Andreia Rangel-Santos
- Laboratorio de Pediatria Clinica (LIM-36), Departamento de Pediatria, Hospital das Clinicas HCFMUSP
| | | | | | | | | | | | | | | | - Camilla Astley
- Applied Physiology and Nutrition Research Group-Center of Lifestyle Medicine, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil
| | | | | | | | | | - Patricia Palmeira
- Laboratorio de Pediatria Clinica (LIM-36), Departamento de Pediatria, Hospital das Clinicas HCFMUSP
| | | |
Collapse
|
2
|
Luo Q, Song Q, Li Y, Zong K, Liu T, He J, Mei G, Du H, Xia Z, Liu M, Song J, Gao C, Xia D, Xue G, Tian W, Qu Y, Kou Z, Dong Z, Han J. Reduced immune response to SARS-CoV-2 infection in the elderly after 6 months. Front Immunol 2025; 16:1596065. [PMID: 40416973 PMCID: PMC12098630 DOI: 10.3389/fimmu.2025.1596065] [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: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 05/27/2025] Open
Abstract
Objectives To evaluate the immune persistence and cross-immune response of elderly individuals after Omicron BA.5 infections. Method The neutralizing antibodies against WT, BA.5, XBB.1 and EG.5 strains were analyzed. The T/B-cell subsets' responses were tested through intracellular cytokine staining and flow cytometry. Results The neutralizing antibodies titers against WT and BA.5 strain, remaining high level for at least 6 months, were higher than that of both XBB.1 and EG.5 variants. The neutralizing antibodies of WT, BA.5, XBB.1, and EG.5 strains in the elderly were slightly lower than those in middle-age. The memory B cells decreased rapidly in the elderly, and Tfh, Th17 cells of the elderly continued to increase for only 3 months, while Tfh and Th17 cells increased in the middle-aged for over 6 months. For the elderly, after peptide stimulation, unswitched/switched memory B cells decreased, while double negative B cells displayed higher proliferation. The proportions of both naïve and Temra cells in CD4+ and CD8+ T cells declined, whereas those of Tcm and Tem cells elevated. In the meantime, both CD69+ and CD38+ T cells decreased, but the frequencies of PD-1+ and CTLA-4+ of CD4+ and CD8+ T cells showed an increasing trend. The proportions of PD-1+ and CTLA-4+ cells also increased in older people with long COVID symptoms at 3m post-infection. Conclusions Omicron BA.5 infection induced lower neutralizing antibodies against XBB.1 and EG.5 variant. The decrease of memory B cells, CD69+ and CD38+T cells, as well as the increase of PD-1+, CTLA-4+ of CD4+/CD8+T cells and double negative B cells, indicate that sustained immune responses against BA.5 infection may wane more rapidly in elderly populations.
Collapse
Affiliation(s)
- Qin Luo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qinqin Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Li
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Intelligent Monitoring, Early Warning, and Prevention and Control of Infectious Diseases, Shandong Institute of Preventive Medicine, Jinan, China
| | - Kexin Zong
- Department of Infectious Diseases, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ti Liu
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Intelligent Monitoring, Early Warning, and Prevention and Control of Infectious Diseases, Shandong Institute of Preventive Medicine, Jinan, China
| | - Junming He
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Guoyong Mei
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haijun Du
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhiqiang Xia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Mi Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Juan Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chen Gao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dong Xia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guangyu Xue
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wenyan Tian
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yinli Qu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zengqiang Kou
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Intelligent Monitoring, Early Warning, and Prevention and Control of Infectious Diseases, Shandong Institute of Preventive Medicine, Jinan, China
| | - Zhongjun Dong
- The First Affiliated Hospital of Anhui Medical University and Institute of Clinical Immunology, Anhui Medical University, Hefei, China
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, Beijing, China
| | - Jun Han
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
3
|
Yuan L, Stoddard M, Sarkar S, van Egeren D, Mangalaganesh S, Nolan RP, Rogers MS, Hather G, White LF, Chakravarty A. The Impact of Vaccination Frequency on COVID-19 Public Health Outcomes: A Model-Based Analysis. Vaccines (Basel) 2025; 13:368. [PMID: 40333247 PMCID: PMC12031506 DOI: 10.3390/vaccines13040368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/16/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Background: While the rapid deployment of SARS-CoV-2 vaccines had a significant impact on the ongoing COVID-19 pandemic, rapid viral immune evasion and waning neutralizing antibody titers have degraded vaccine efficacy. Nevertheless, vaccine manufacturers and public health authorities have a number of options at their disposal to maximize the benefits of vaccination. In particular, the effect of booster schedules on vaccine performance bears further study. Methods: To better understand the effect of booster schedules on vaccine performance, we used an agent-based modeling framework and a population pharmacokinetic model to simulate the impact of boosting frequency on the durability of vaccine protection against infection and severe acute disease. Results: Our work suggests that repeated dosing at frequent intervals (three or more times a year) may offset the degradation of vaccine efficacy, preserving the utility of vaccines in managing the ongoing pandemic. Conclusions: Given the practical significance of potential improvements in vaccine utility, clinical research to better understand the effects of repeated vaccination would be highly impactful. These findings are particularly relevant as public health authorities worldwide have reduced the frequency of boosters to once a year or less.
Collapse
Affiliation(s)
- Lin Yuan
- Fractal Therapeutics, Lexington, MA 02420, USA; (L.Y.); (M.S.)
| | | | - Sharanya Sarkar
- Department of Microbiology and Immunology, Dartmouth College, Hanover, NH 03755, USA;
| | - Debra van Egeren
- Department of Oncology, School of Medicine, Stanford University, Stanford, CA 94305, USA;
| | - Shruthi Mangalaganesh
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia;
| | | | - Michael S. Rogers
- Department of Surgery, Harvard Medical School, Boston, MA 02114, USA;
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Greg Hather
- Sage Therapeutics, Cambridge, MA 02142, USA;
| | - Laura F. White
- School of Public Health, Boston University, Boston, MA 02118, USA;
| | | |
Collapse
|
4
|
de Oliveira G, Costa-Rocha IA, Oliveira-Carvalho N, dos Santos TMAF, Campi-Azevedo AC, Peruhype-Magalhães V, Miranda VHS, Prado RO, Pereira AAS, Alves CC, Brito-de-Sousa JP, Reis LR, Costa-Pereira C, da Mata CPSM, Almeida VES, dos Santos LM, Almeida GG, Antonelli LRDV, Coelho-dos-Reis JG, Teixeira-Carvalho A, Martins-Filho OA. Phenotypic Timeline Kinetics, Integrative Networks, and Performance of T- and B-Cell Subsets Associated with Distinct Clinical Outcome of Severe COVID-19 Patients. Microorganisms 2024; 12:2272. [PMID: 39597661 PMCID: PMC11596994 DOI: 10.3390/microorganisms12112272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 11/29/2024] Open
Abstract
The present study aimed to evaluate the kinetics of the phenotypic profile and integrative networks of T/B-cells in severe COVID-19 patients, categorized according to disease outcome, during the circulation of the B.1.1.28 and B.1.1.33 SARS-CoV-2 strains in Brazil. Peripheral blood obtained at distinct time points (baseline/D0; D7; D14-28) was used for ex vivo flow cytometry immunophenotyping. The data demonstrated a decrease at D0 in the frequency of CD3+ T-cells and CD4+ T-cells and an increase in B-cells with mixed activation/exhaustion profiles. Higher changes in B-cell and CD4+ T-cells at D7 were associated with discharge/death outcomes, respectively. Regardless of the lower T/B-cell connectivity at D0, distinct profiles from D7/D14-28 revealed that, while discharge was associated with increasing connectivity for B-cells, CD4+ and CD8+ T-cells death was related to increased connectivity involving B-cells, but with lower connections mediated by CD4+ T-cells. The CD4+CD38+ and CD8+CD69+ subsets accurately classified COVID-19 vs. healthy controls throughout the kinetic analysis. Binary logistic regression identified CD4+CD107a+, CD4+T-bet+, CD8+CD69+, and CD8+T-bet+ at D0 and CD4+CD45RO+CD27+ at D7 as subsets associated with disease outcomes. Results showed that distinct phenotypic timeline kinetics and integrative networks of T/B-cells are associated with COVID-19 outcomes that may subsidize the establishment of applicable biomarkers for clinical/therapeutic monitoring.
Collapse
Affiliation(s)
- Gabriela de Oliveira
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Ismael Artur Costa-Rocha
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Nani Oliveira-Carvalho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Tâmilla Mayane Alves Fidelis dos Santos
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Ana Carolina Campi-Azevedo
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Vanessa Peruhype-Magalhães
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Vitor Hugo Simões Miranda
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Roberta Oliveira Prado
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Agnes Antônia Sampaio Pereira
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Clarice Carvalho Alves
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Joaquim Pedro Brito-de-Sousa
- Departamento de Imunologia e Parasitologia, Universidade Federal de Uberlândia, Uberlândia 38408-100, MG, Brazil;
| | - Laise Rodrigues Reis
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Christiane Costa-Pereira
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | | | | | - Liliane Martins dos Santos
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Gregório Guilherme Almeida
- Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.G.A.); (L.R.d.V.A.)
| | - Lis Ribeiro do Valle Antonelli
- Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.G.A.); (L.R.d.V.A.)
| | - Jordana Grazziela Coelho-dos-Reis
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Andréa Teixeira-Carvalho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ-Minas, Belo Horizonte 30190-002, MG, Brazil; (G.d.O.); (I.A.C.-R.); (N.O.-C.); (T.M.A.F.d.S.); (A.C.C.-A.); (V.P.-M.); (V.H.S.M.); (R.O.P.); (A.A.S.P.); (C.C.A.); (L.R.R.); (C.C.-P.); (L.M.d.S.); (J.G.C.-d.-R.); (A.T.-C.)
| |
Collapse
|
5
|
Chen Z, Liu X, Guan J, Shi Y, Liu W, Peng Z, Hu J. Impact of COVID-19 Interventions on Respiratory and Intestinal Infectious Disease Notifications - Jiangsu Province, China, 2020-2023. China CDC Wkly 2024; 6:1059-1064. [PMID: 39502400 PMCID: PMC11532534 DOI: 10.46234/ccdcw2024.219] [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: 06/21/2024] [Accepted: 09/19/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Many measures implemented to control the coronavirus disease 2019 (COVID-19) pandemic have reshaped the epidemic patterns of other infectious diseases. This study estimated the impact of the COVID-19 pandemic on respiratory and intestinal infectious diseases and potential changes following reopening. Methods The optimal intervention and counterfactual models were selected from the seasonal autoregressive integrated moving average (SARIMA), neural network autoregression (NNAR), and hybrid models based on the minimum mean absolute percentage error (MAPE) in the test set. The relative change rate between the actual notification rate and that predicted by the optimal model was calculated for the entire COVID-19 epidemic prevention period and the "reopening" period. Results Compared with the predicted notification rate based on the counterfactual model, the total relative change rates for the 9 infectious diseases were -44.24%, respiratory infections (-55.41%), and intestinal infections (-26.59%) during 2020-2022. Compared with the predicted notification rate based on the intervention model, the total relative change rates were +247.98%, respiratory infections (+389.59%), and intestinal infections (+50.46%) in 2023. Among them, the relative increases in influenza (+499.98%) and hand-foot-mouth disease (HFMD) (+70.97%) were significant. Conclusions Measures taken in Jiangsu Province in response to COVID-19 effectively constrained the spread of respiratory and intestinal infectious diseases. Influenza and HFMD rebounded significantly after the lifting of COVID-19 intervention restrictions.
Collapse
Affiliation(s)
- Ziying Chen
- School of Public Health, Nanjing Medical University, Nanjing City, Jiangsu Province, China
| | - Xin Liu
- School of Public Health, Nanjing Medical University, Nanjing City, Jiangsu Province, China
| | - Jinxing Guan
- School of Public Health, Nanjing Medical University, Nanjing City, Jiangsu Province, China
| | - Yingying Shi
- Department of Acute Infectious Diseases Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing City, Jiangsu Province, China
| | - Wendong Liu
- Department of Acute Infectious Diseases Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing City, Jiangsu Province, China
| | - Zhihang Peng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jianli Hu
- School of Public Health, Nanjing Medical University, Nanjing City, Jiangsu Province, China
- Department of Acute Infectious Diseases Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing City, Jiangsu Province, China
- Jiangsu Province Engineering Research Center of Health Emergency, Nanjing City, Jiangsu Province, China
| |
Collapse
|
6
|
Liu Y, Bai Z, Yang T, Yuan B, Han Y, Xiang Y, Zhou R, Sun J, Chen M, Hao C, Wang H. Changes in the epidemiology of pediatric brain abscesses pre- and post-COVID-19 pandemic: a single-center study. BMC Pediatr 2024; 24:600. [PMID: 39306664 PMCID: PMC11416000 DOI: 10.1186/s12887-024-05082-6] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND An increased incidence of brain abscesses was observed post-COVID-19 pandemic. However, it remains unclear how the COVID-19 pandemic influenced the epidemiology of brain abscesses. This study aimed to investigate changes in the epidemiology of brain abscesses pre- and post-COVID-19 pandemic. METHODS A retrospective study of demographic, clinical, radiological, and laboratory characteristics of patients with brain abscesses in Children's Hospital of Soochow University from 2015-2023 was performed. RESULTS A total of 34 patients were admitted to the hospital during the study. The post-COVID-19 cohort had an average of 5.5 cases/year, which is a 129.2% increase compared to the pre-COVID-19 cohort's average of 2.4 cases/year. Additionally, the rates of fever upon admission (86.36% vs 50%, p = 0.04) and experiencing high-grade fever within 6 weeks before admission (40.91% vs 8.33%, p = 0.044) were significantly increased. A potential rise in the rate of intensive care unit admission was observed (36.36% vs 8.33%, p = 0.113). The average value of globulin in the post-COVID cohort was significantly higher compared to the pre-COVID cohort (31.60 ± 5.97 vs 25.50 ± 5.08, p = 0.009). Streptococcal infections were the predominant cause of brain abscesses in both cohorts (40% vs 43.75%, p = 0.57). CONCLUSIONS There was a significant increase in the number of brain abscess patients after the COVID-19 pandemic. This underscores the importance of children receiving the streptococcal vaccine.
Collapse
Affiliation(s)
- Yuchen Liu
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Zhenjiang Bai
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Tianquan Yang
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Bin Yuan
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yong Han
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yongjun Xiang
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Ruxuan Zhou
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jingxuan Sun
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Min Chen
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Chuangli Hao
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Hangzhou Wang
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| |
Collapse
|
7
|
Lisowska KA, Ciesielska-Figlon K, Komorniczak M, Bułło-Piontecka B, Dębska-Ślizień A, Wardowska A. Peripheral Blood Mononuclear Cells and Serum Cytokines in Patients with Lupus Nephritis after COVID-19. Int J Mol Sci 2024; 25:8278. [PMID: 39125849 PMCID: PMC11311954 DOI: 10.3390/ijms25158278] [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: 07/01/2024] [Revised: 07/18/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Systemic lupus erythematosus (SLE) patients have an increased risk of infections and infection-related mortality. Therefore, during the global SARS-CoV-2 pandemic, SLE patients were particularly vulnerable to SARS-CoV-2 infections. Also, compared to other patients, SLE patients seem to develop more severe manifestations of coronavirus disease 2019 (COVID-19), with higher rates of hospitalization, invasive ventilation requirements, or death. This study evaluated the immune parameters after SARS-CoV-2 infection in SLE patients. We analyzed subpopulations of peripheral blood cells collected from patients with renal manifestation of SLE (lupus nephritis, LN). LN patients were divided into two subgroups: those unexposed to SARS-CoV-2 (LN CoV-2(-)) and those who had confirmed COVID-19 (LN-CoV-2(+)) six months earlier. We analyzed basic subpopulations of T cells, B cells, monocytes, dendritic cells (DCs), and serum cytokines using flow cytometry. All collected data were compared to a healthy control group without SARS-CoV-2 infection in medical history. LN patients were characterized by a decreased percentage of helper T (Th) cells and an increased percentage of cytotoxic T (Tc) cells regardless of SARS-CoV-2 infection. LN CoV-2(+) patients had a higher percentage of regulatory T cells (Tregs) and plasmablasts (PBs) and a lower percentage of non-switched memory (NSM) B cells compared to LN CoV-2(-) patients or healthy controls (HC CoV-2(-)). LN patients had a higher percentage of total monocytes compared with HC CoV-2(-). LN CoV-2(+) patients had a higher percentage of classical and intermediate monocytes than LN CoV-2(-) patients and HC CoV-2(-). LN CoV-2(+) patients had higher serum IL-6 levels than HC CoV-2(-), while LN CoV-2(-) patients had higher levels of serum IL-10. LN patients are characterized by disturbances in the blood's basic immunological parameters. However, SARS-CoV-2 infection influences B-cell and monocyte compartments.
Collapse
Affiliation(s)
- Katarzyna A. Lisowska
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (K.A.L.)
| | - Klaudia Ciesielska-Figlon
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (K.A.L.)
| | - Michał Komorniczak
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Barbara Bułło-Piontecka
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Alicja Dębska-Ślizień
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Anna Wardowska
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (K.A.L.)
| |
Collapse
|
8
|
Zhang L, Woltering I, Holzner M, Brandhofer M, Schaefer CC, Bushati G, Ebert S, Yang B, Muenchhoff M, Hellmuth JC, Scherer C, Wichmann C, Effinger D, Hübner M, El Bounkari O, Scheiermann P, Bernhagen J, Hoffmann A. CD74 is a functional MIF receptor on activated CD4 + T cells. Cell Mol Life Sci 2024; 81:296. [PMID: 38992165 PMCID: PMC11335222 DOI: 10.1007/s00018-024-05338-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024]
Abstract
Next to its classical role in MHC II-mediated antigen presentation, CD74 was identified as a high-affinity receptor for macrophage migration inhibitory factor (MIF), a pleiotropic cytokine and major determinant of various acute and chronic inflammatory conditions, cardiovascular diseases and cancer. Recent evidence suggests that CD74 is expressed in T cells, but the functional relevance of this observation is poorly understood. Here, we characterized the regulation of CD74 expression and that of the MIF chemokine receptors during activation of human CD4+ T cells and studied links to MIF-induced T-cell migration, function, and COVID-19 disease stage. MIF receptor profiling of resting primary human CD4+ T cells via flow cytometry revealed high surface expression of CXCR4, while CD74, CXCR2 and ACKR3/CXCR7 were not measurably expressed. However, CD4+ T cells constitutively expressed CD74 intracellularly, which upon T-cell activation was significantly upregulated, post-translationally modified by chondroitin sulfate and could be detected on the cell surface, as determined by flow cytometry, Western blot, immunohistochemistry, and re-analysis of available RNA-sequencing and proteomic data sets. Applying 3D-matrix-based live cell-imaging and receptor pathway-specific inhibitors, we determined a causal involvement of CD74 and CXCR4 in MIF-induced CD4+ T-cell migration. Mechanistically, proximity ligation assay visualized CD74/CXCR4 heterocomplexes on activated CD4+ T cells, which were significantly diminished after MIF treatment, pointing towards a MIF-mediated internalization process. Lastly, in a cohort of 30 COVID-19 patients, CD74 surface expression was found to be significantly upregulated on CD4+ and CD8+ T cells in patients with severe compared to patients with only mild disease course. Together, our study characterizes the MIF receptor network in the course of T-cell activation and reveals CD74 as a novel functional MIF receptor and MHC II-independent activation marker of primary human CD4+ T cells.
Collapse
Affiliation(s)
- Lin Zhang
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Iris Woltering
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Mathias Holzner
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Markus Brandhofer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Carl-Christian Schaefer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Genta Bushati
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Simon Ebert
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Bishan Yang
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Maximilian Muenchhoff
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Johannes C Hellmuth
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Department of Medicine III, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Clemens Scherer
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Department of Medicine I, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Christian Wichmann
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - David Effinger
- Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistraße 15, 81377, Munich, Germany
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Max Hübner
- Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistraße 15, 81377, Munich, Germany
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Omar El Bounkari
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Patrick Scheiermann
- Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
- German Centre of Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| | - Adrian Hoffmann
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
- Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistraße 15, 81377, Munich, Germany.
- German Centre of Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
9
|
Berger L, Wolf J, Kalbitz S, Kellner N, Lübbert C, Borte S. Comparative Analysis of Lymphocyte Populations in Post-COVID-19 Condition and COVID-19 Convalescent Individuals. Diagnostics (Basel) 2024; 14:1286. [PMID: 38928701 PMCID: PMC11202600 DOI: 10.3390/diagnostics14121286] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Reduced lymphocyte counts in peripheral blood are one of the most common observations in acute phases of viral infections. Although many studies have already examined the impact of immune (dys)regulation during SARS-CoV-2 infection, there are still uncertainties about the long-term consequences for lymphocyte homeostasis. Furthermore, as persistent cellular aberrations have been described following other viral infections, patients with "Post-COVID-19 Condition" (PCC) may present similarly. In order to investigate cellular changes in the adaptive immune system, we performed a retrospective analysis of flow cytometric data from lymphocyte subpopulations in 106 patients with confirmed SARS-CoV-2 infection who received medical care at our institution. The patients were divided into three groups according to the follow-up date; laboratory analyses of COVID-19 patients were compared with 28 unexposed healthy controls. Regarding B lymphocyte subsets, levels of IgA + CD27+, IgG + CD27+, IgM + CD27- and switched B cells were significantly reduced at the last follow-up compared to unexposed healthy controls (UHC). Of the 106 COVID-19 patients, 56 were clinically classified as featuring PCC. Significant differences between PCC and COVID-19 convalescents compared to UHC were observed in T helper cells and class-switched B cells. However, we did not detect specific or long-lasting immune cellular changes in PCC compared to the non-post-COVID-19 condition.
Collapse
Affiliation(s)
- Luisa Berger
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
| | - Johannes Wolf
- Department of Laboratory Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| | - Sven Kalbitz
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
| | - Nils Kellner
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Stephan Borte
- Department of Laboratory Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| |
Collapse
|
10
|
Kado A, Tsutsumi T, Yotsuyanagi H, Ikeuchi K, Okushin K, Moriya K, Koike K, Fujishiro M. Differential peripheral memory T cell subsets sensitively indicate the severity of nonalcoholic fatty liver disease. Hepatol Res 2024; 54:525-539. [PMID: 38157267 DOI: 10.1111/hepr.14009] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
AIM Differential patterns of peripheral memory T cell subsets in nonalcoholic fatty liver disease (NAFLD) were assessed using flow cytometry (FCM) to elucidate their association with NAFLD severity and provide a new noninvasive method to sensitively detect the disease severity in addition to existing biomarkers. METHODS We assessed the differential frequencies of peripheral memory T cell subsets in 103 patients with NAFLD according to the degree of liver fibrosis (FIB) using FCM analysis. We focused on the following populations: CCR7+ CD45RA+ naïve T, CCR7+ CD45RA- central memory T cells (TCM), CCR7- CD45RA- effector memory T, and CCR7- CD45RA+ terminally differentiated effector memory T (TEMRA) cells in CD4+ and CD8+ T, Th1, Th2, and Th17 cells, respectively. To evaluate the pathological progression of the disease, these frequencies were also examined according to the degree of the NAFLD activity score (NAS). RESULTS Several significant correlations were observed between laboratory parameters and peripheral memory T lymphocyte frequencies according to the degree of liver FIB and NAS in NAFLD. In univariate and multivariate analyses, the frequency of CD8+ TEMRA cells predicted severe FIB, and the predictive power was validated in an independent cohort. Furthermore, the frequencies of several memory T cell subsets sensitively indicated the pathological progression of NAFLD (Th17 TCM: steatosis, CD4+ TCM: lobular inflammation, and CD8+ TEMRA and effector memory T cells: hepatocellular ballooning). CONCLUSIONS Our results suggest that the analysis of peripheral memory T lymphocyte frequencies can noninvasively predict severe FIB and sensitively indicate the pathological progression of NAFLD.
Collapse
Affiliation(s)
- Akira Kado
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Division for Health Service Promotion, The University of Tokyo, Tokyo, Japan
| | - Takeya Tsutsumi
- Department of Infection Control and Prevention, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Infectious Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Yotsuyanagi
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Ikeuchi
- Department of Infectious Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuya Okushin
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Infection Control and Prevention, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kyoji Moriya
- Division of Infection Control and Prevention, Education Research Center, The Tokyo Health Care University, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Gastroenterology, Kanto Central Hospital, Tokyo, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Suryawanshi P, Patil‐Takbhate B, Athavale P, Mirza S, Tripathy A, Kanitkar S, Shivnitwar S, Barthwal MS, Dole S, Chavan H, Jali P, Pawale S, Kakad D, Kakrani AL, Bhawalkar J, Gandhi M, Chaturvedi S, Karandikar M, Tripathy S. T-cell responses in COVID-19 survivors 6-8 months after infection: A longitudinal cohort study in Pune. Immun Inflamm Dis 2024; 12:e1238. [PMID: 38860770 PMCID: PMC11165687 DOI: 10.1002/iid3.1238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/26/2024] [Accepted: 03/20/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune response is crucial for disease management, although diminishing immunity raises the possibility of reinfection. METHODS We examined the immunological response to SARS-CoV-2 in a cohort of convalescent COVID-19 patients in matched samples collected at 1 and 6-8 months after infection. The peripheral blood mononuclear cells were isolated from enrolled study participants and flow cytometry analysis was done to assess the lymphocyte subsets of naive, effector, central memory, and effector memory CD4+ or CD8+ T cells in COVID-19 patients at 1 and 6-8 months after infection. Immunophenotypic characterization of immune cell subsets was performed on individuals who were followed longitudinally for 1 month (n = 44) and 6-8 months (n = 25) after recovery from COVID infection. RESULTS We observed that CD4 +T cells in hospitalized SARS-CoV-2 patients tended to decrease, whereas CD8+ T cells steadily recovered after 1 month, while there was a sustained increase in the population of effector T cells and effector memory T cells. Furthermore, COVID-19 patients showed persistently low B cells and a small increase in the NK cell population. CONCLUSION Our findings show that T cell responses were maintained at 6-8 months after infection. This opens new pathways for further research into the long-term effects in COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
- Poonam Suryawanshi
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Bhagyashri Patil‐Takbhate
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Prachi Athavale
- Department of Microbiology, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Shahzad Mirza
- Department of Microbiology, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | | | - Shubhangi Kanitkar
- Department of General Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Sachin Shivnitwar
- Department of General Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Madhusudan S. Barthwal
- Department of Respiratory Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, Pimpri, (deemed to be University)PuneIndia
| | - Sachin Dole
- Department of Respiratory Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, Pimpri, (deemed to be University)PuneIndia
| | - Hanumant Chavan
- Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Priyanka Jali
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Sujata Pawale
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Dhanashree Kakad
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Arjun Lal Kakrani
- Department of General Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Jitendra Bhawalkar
- Department of Community Medicine, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Madhura Gandhi
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | | | - Mahesh Karandikar
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| | - Srikanth Tripathy
- Central Research Facility, Dr D. Y. Patil Medical College, Hospital and Research CentreDr D. Y. Patil Vidyapeeth, (deemed to be University)PimpriPuneIndia
| |
Collapse
|
12
|
Andersson NW, Thiesson EM, Lassaunière R, Hansen JV, Hviid A. SARS-CoV-2 Infection and Postacute Risk of Non-Coronavirus Disease 2019 Infectious Disease Hospitalizations: A Nationwide Cohort Study of Danish Adults Aged ≥50 Years. Clin Infect Dis 2024; 78:603-612. [PMID: 37740392 DOI: 10.1093/cid/ciad531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Reports suggest that the potential long-lasting health consequences of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may involve persistent dysregulation of some immune populations, but the potential clinical implications are unknown. We investigated the associated risk of hospitalization due to non-coronavirus disease 2019 (COVID-19) infectious diseases following the postacute phase of SARS-CoV-2 infection. METHODS By cross-linking data from the comprehensive Danish test and surveillance system for COVID-19 together with nationwide healthcare and demographic registers, we established a study cohort of 2 430 694 individuals aged ≥50 years, from 1 January 2021 to 10 December 2022, with no evidence of SARS-CoV-2 infection prior to study entry. Using Poisson regression, we compared the outcome rates of non-COVID-19 infectious disease hospitalizations following the acute phase of (a first) SARS-CoV-2 infection (defined as ≥29 days since the day of infection) in recovered individuals with rates among SARS-CoV-2-uninfected individuals. RESULTS Among 2 430 694 included individuals (mean age, 66.8 [standard deviation, 11.3] years), 930 071 acquired SARS-CoV-2 infection during follow-up totaling 4 519 913 person-years. The postacute phase of SARS-CoV-2 infection was associated with an incidence rate ratio (IRR) of 0.90 (95% confidence interval [CI]: .88-.92) for any infectious disease hospitalization. Findings (IRR [95% CI]) were similar for upper respiratory tract (1.08 [.97-1.20]), lower respiratory tract (0.90 [.87-.93]), influenza (1.04 [.94-1.15]), gastrointestinal (1.28 [.78-2.09]), skin (0.98 [.93-1.03]), urinary tract (1.01 [.96-1.08]), certain invasive bacterial (0.96 [.91-1.01]), and other (0.96 [.92-1.00]) infectious disease hospitalizations and in subgroups. CONCLUSIONS Our study does not support an increased susceptibility to non-COVID-19 infectious disease hospitalization following SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | | | - Ria Lassaunière
- Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Denmark
| | | | - Anders Hviid
- Department of Epidemiology Research, Statens Serum Institut, Denmark
- Pharmacovigilance Research Center, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
13
|
Safont G, Villar-Hernández R, Smalchuk D, Stojanovic Z, Marín A, Lacoma A, Pérez-Cano C, López-Martínez A, Molina-Moya B, Solis AJ, Arméstar F, Matllo J, Díaz-Fernández S, Romero I, Casas I, Strecker K, Preyer R, Rosell A, Latorre I, Domínguez J. Measurement of IFN-γ and IL-2 for the assessment of the cellular immunity against SARS-CoV-2. Sci Rep 2024; 14:1137. [PMID: 38212416 PMCID: PMC10784529 DOI: 10.1038/s41598-024-51505-w] [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: 07/17/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024] Open
Abstract
The study of specific T-cell responses against SARS-CoV-2 is important for understanding long-term immunity and infection management. The aim of this study was to assess the dual IFN-γ and IL-2 detection, using a SARS-CoV-2 specific fluorescence ELISPOT, in patients undergoing acute disease, during convalescence, and after vaccination. We also evaluated humoral response and compared with T-cells with the aim of correlating both types of responses, and increase the number of specific response detection. Blood samples were drawn from acute COVID-19 patients and convalescent individuals classified according to disease severity; and from unvaccinated and vaccinated uninfected individuals. IgGs against Spike and nucleocapsid, IgMs against nucleocapsid, and neutralizing antibodies were also analyzed. Our results show that IFN-γ in combination with IL-2 increases response detection in acute and convalescent individuals (p = 0.023). In addition, IFN-γ detection can be a useful biomarker for monitoring severe acute patients, as our results indicate that those individuals with a poor outcome have lower levels of this cytokine. In some cases, the lack of cellular immunity is compensated by antibodies, confirming the role of both types of immune responses in infection, and confirming that their dual detection can increase the number of specific response detections. In summary, IFN-γ/IL-2 dual detection is promising for characterizing and assessing the immunization status, and helping in the patient management.
Collapse
Affiliation(s)
- Guillem Safont
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Raquel Villar-Hernández
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- Genome Identification Diagnostics GmbH (GenID), Straßberg, Germany
| | - Daria Smalchuk
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- Odesa I. I. Mechnykov National University, Odesa, Ukraine
| | - Zoran Stojanovic
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Alicia Marín
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Alicia Lacoma
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Pérez-Cano
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Anabel López-Martínez
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Bárbara Molina-Moya
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alan Jhunior Solis
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Fernando Arméstar
- Intensive Care Medicine Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
| | - Joan Matllo
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Sergio Díaz-Fernández
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Iris Romero
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irma Casas
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Preventive Medicine Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Kevin Strecker
- Genome Identification Diagnostics GmbH (GenID), Straßberg, Germany
| | - Rosemarie Preyer
- Genome Identification Diagnostics GmbH (GenID), Straßberg, Germany
| | - Antoni Rosell
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Irene Latorre
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jose Domínguez
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain.
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
14
|
Hultberg J, Blixt E, Göransson R, Adolfsson J, Govender M, Larsson M, Nilsdotter-Augustinsson Å, Ernerudh J, Nyström S. In-depth immune profiling reveals advanced B- and T-cell differentiation to be associated with Th1-driven immune dysregulation in common variable immunodeficiency. Clin Immunol 2023; 257:109816. [PMID: 37918468 DOI: 10.1016/j.clim.2023.109816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Common variable immunodeficiency (CVID) is an inborn error of immunity characterized by low levels of antibodies. In addition to infections, many patients also suffer from T-helper 1-driven immune dysregulation, which is associated with increased mortality. The aim of this study was to perform in-depth characterization of the T and the B cell compartments in a well-defined cohort of patients affected by CVID and correlate the findings to the level of clinical immune dysregulation. We used mass cytometry, targeted proteomics, flow cytometry and functional assays to delineate the immunological phenotype of 15 CVID-affected patients with different levels of immune dysregulation. Unbiased clustering of T cell mass cytometry data correlated with CVID-related immune dysregulation and plasma protein profiles. Expanded CXCR3+ T-bet-expressing B cells correlated with effector memory CD4+ T cell clusters, and increased plasma levels of CXCR3-ligands. Our findings indicate an interplay between B cells and T cells in CVID-related immune dysregulation and provide a better understanding of the underlying pathological mechanisms.
Collapse
Affiliation(s)
- Jonas Hultberg
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Emelie Blixt
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Robin Göransson
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Jörgen Adolfsson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Melissa Govender
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Marie Larsson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Åsa Nilsdotter-Augustinsson
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Sweden
| | - Sofia Nyström
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Sweden.
| |
Collapse
|
15
|
Yadav S, Mehta P, Soni J, Chattopadhyay P, Devi P, Habyarimana T, Tardalkar K, Joshi M, Pandey R. Single-cell RNA-Seq reveals intracellular microbial diversity within immune cells during SARS-CoV-2 infection and recovery. iScience 2023; 26:108357. [PMID: 38026191 PMCID: PMC10663746 DOI: 10.1016/j.isci.2023.108357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/05/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Intracellular microorganisms, like viruses, bacteria, and fungi, pose challenges in detection due to their non-culturable forms. Transcriptomic analysis at cellular level enables exploration of distributions and the impact of these microorganisms on host cells, a domain that remains underexplored because of methodological limitations. Single-cell technology shows promise in addressing this by capturing polyadenine-tailed transcripts, because recent studies confirmed polyadenylation in microbial transcriptomes. We utilized single-cell RNA-seq from PBMCs to probe intracellular microbes in healthy, SARS-CoV-2-positive, and recovered individuals. Among 76 bacterial species detected, 16 showed significant abundance differences. Buchnera aphidicola, Streptomyces clavuligerus, and Ehrlichia canis emerged significantly in memory-B, Naïve-T, and Treg cells. Staphylococcus aureus, Mycoplasma mycoides, Leptospira interrogans, and others displayed elevated levels in SARS-CoV-2-positive patients, suggesting possible disease association. This highlights the strength of single-cell technology in revealing potential microorganism's cell-specific functions. Further research is essential for functional understanding of their cell-specific abundance across physiological states.
Collapse
Affiliation(s)
- Sunita Yadav
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi 110007, India
| | - Priyanka Mehta
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jyoti Soni
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Partha Chattopadhyay
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Priti Devi
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Thierry Habyarimana
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi 110007, India
- Department of Biomedical Laboratory Sciences, INES-Ruhengeri, Ruhengeri, Rwanda
| | - Kishore Tardalkar
- Dr. D. Y. Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra 416003, India
| | - Meghnad Joshi
- Dr. D. Y. Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra 416003, India
| | - Rajesh Pandey
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
16
|
Berentschot JC, Drexhage HA, Aynekulu Mersha DG, Wijkhuijs AJM, GeurtsvanKessel CH, Koopmans MPG, Voermans JJC, Hendriks RW, Nagtzaam NMA, de Bie M, Heijenbrok-Kal MH, Bek LM, Ribbers GM, van den Berg-Emons RJG, Aerts JGJV, Dik WA, Hellemons ME. Immunological profiling in long COVID: overall low grade inflammation and T-lymphocyte senescence and increased monocyte activation correlating with increasing fatigue severity. Front Immunol 2023; 14:1254899. [PMID: 37881427 PMCID: PMC10597688 DOI: 10.3389/fimmu.2023.1254899] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/14/2023] [Indexed: 10/27/2023] Open
Abstract
Background Many patients with SARS-CoV-2 infection develop long COVID with fatigue as one of the most disabling symptoms. We performed clinical and immune profiling of fatigued and non-fatigued long COVID patients and age- and sex-matched healthy controls (HCs). Methods Long COVID symptoms were assessed using patient-reported outcome measures, including the fatigue assessment scale (FAS, scores ≥22 denote fatigue), and followed up to one year after hospital discharge. We assessed inflammation-related genes in circulating monocytes, serum levels of inflammation-regulating cytokines, and leukocyte and lymphocyte subsets, including major monocyte subsets and senescent T-lymphocytes, at 3-6 months post-discharge. Results We included 37 fatigued and 36 non-fatigued long COVID patients and 42 HCs. Fatigued long COVID patients represented a more severe clinical profile than non-fatigued patients, with many concurrent symptoms (median 9 [IQR 5.0-10.0] vs 3 [1.0-5.0] symptoms, p<0.001), and signs of cognitive failure (41%) and depression (>24%). Immune abnormalities that were found in the entire group of long COVID patients were low grade inflammation (increased inflammatory gene expression in monocytes, increased serum pro-inflammatory cytokines) and signs of T-lymphocyte senescence (increased exhausted CD8+ TEMRA-lymphocytes). Immune profiles did not significantly differ between fatigued and non-fatigued long COVID groups. However, the severity of fatigue (total FAS score) significantly correlated with increases of intermediate and non-classical monocytes, upregulated gene levels of CCL2, CCL7, and SERPINB2 in monocytes, increases in serum Galectin-9, and higher CD8+ T-lymphocyte counts. Conclusion Long COVID with fatigue is associated with many concurrent and persistent symptoms lasting up to one year after hospitalization. Increased fatigue severity associated with stronger signs of monocyte activation in long COVID patients and potentially point in the direction of monocyte-endothelial interaction. These abnormalities were present against a background of immune abnormalities common to the entire group of long COVID patients.
Collapse
Affiliation(s)
- Julia C. Berentschot
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Hemmo A. Drexhage
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | | | | | - Marion P. G. Koopmans
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jolanda J. C. Voermans
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Rudi W. Hendriks
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Nicole M. A. Nagtzaam
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Maaike de Bie
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Majanka H. Heijenbrok-Kal
- Department of Rehabilitation Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Rijndam Rehabilitation, Rotterdam, Netherlands
| | - L. Martine Bek
- Department of Rehabilitation Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Gerard M. Ribbers
- Department of Rehabilitation Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Rijndam Rehabilitation, Rotterdam, Netherlands
| | | | - Joachim G. J. V. Aerts
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Merel E. Hellemons
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
17
|
Hopkins FR, Nordgren J, Fernandez-Botran R, Enocsson H, Govender M, Svanberg C, Svensson L, Hagbom M, Nilsdotter-Augustinsson Å, Nyström S, Sjöwall C, Sjöwall J, Larsson M. Pentameric C-reactive protein is a better prognostic biomarker and remains elevated for longer than monomeric CRP in hospitalized patients with COVID-19. Front Immunol 2023; 14:1259005. [PMID: 37724104 PMCID: PMC10505432 DOI: 10.3389/fimmu.2023.1259005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/11/2023] [Indexed: 09/20/2023] Open
Abstract
The differing roles of the pentameric (p) and monomeric (m) C-reactive protein (CRP) isoforms in viral diseases are not fully understood, which was apparent during the COVID-19 pandemic regarding the clinical course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Herein, we investigated the predictive value of the pCRP and mCRP isoforms for COVID-19 severity in hospitalized patients and evaluated how the levels of the protein isoforms changed over time during and after acute illness. This study utilized samples from a well-characterized cohort of Swedish patients with SARS-CoV-2 infection, the majority of whom had known risk factors for severe COVID-19 and required hospitalization. The levels of pCRP were significantly raised in patients with severe COVID-19 and in contrast to mCRP the levels were significantly associated with disease severity. Additionally, the pCRP levels remained elevated for at least six weeks post inclusion, which was longer compared to the two weeks for mCRP. Our data indicates a low level of inflammation lasting for at least six weeks following COVID-19, which might indicate that the disease has an adverse effect on the immune system even after the viral infection is resolved. It is also clear that the current standard method of testing pCRP levels upon hospitalization is a useful marker for predicting disease severity and mCRP testing would not add any clinical relevance for patients with COVID-19.
Collapse
Affiliation(s)
- Francis R. Hopkins
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johan Nordgren
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Rafael Fernandez-Botran
- Department of Pathology & Laboratory Medicine, University of Louisville, Louisville, KY, United States
| | - Helena Enocsson
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Melissa Govender
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Cecilia Svanberg
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lennart Svensson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Division of Infectious Diseases, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Marie Hagbom
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Åsa Nilsdotter-Augustinsson
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Infectious Diseases, Vrinnevi Hospital, Norrköping, Sweden
| | - Sofia Nyström
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Clinical Immunology and Transfusion Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Christopher Sjöwall
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johanna Sjöwall
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Infectious Diseases, Vrinnevi Hospital, Norrköping, Sweden
| | - Marie Larsson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
18
|
Rabady S, Hoffmann K, Aigner M, Altenberger J, Brose M, Costa U, Denk-Linnert DM, Gruber S, Götzinger F, Helbok R, Hüfner K, Koczulla R, Kurz K, Lamprecht B, Leis S, Löffler J, Müller CA, Rittmannsberger H, Rommer PS, Sator P, Strenger V, Struhal W, Untersmayr E, Vonbank K, Wancata J, Weber T, Wendler M, Zwick RH. [S1 guidelines for the management of postviral conditions using the example of post-COVID-19]. Wien Klin Wochenschr 2023; 135:525-598. [PMID: 37555900 PMCID: PMC10504206 DOI: 10.1007/s00508-023-02242-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2023] [Indexed: 08/10/2023]
Abstract
These S1 guidelines are an updated and expanded version of the S1 guidelines on long COVID differential diagnostic and management strategies. They summarize the state of knowledge on postviral conditions like long/post COVID at the time of writing. Due to the dynamic nature of knowledge development, they are intended to be "living guidelines". The focus is on practical applicability at the level of primary care, which is understood to be the appropriate place for initial access and for primary care and treatment. The guidelines provide recommendations on the course of treatment, differential diagnostics of the most common symptoms that can result from infections like with SARS-CoV-2, treatment options, patient management and care, reintegration and rehabilitation. The guidelines have been developed through an interdisciplinary and interprofessional process and provide recommendations on interfaces and possibilities for collaboration.
Collapse
Affiliation(s)
- Susanne Rabady
- Department Allgemeine Gesundheitsstudien, Kompetenzzentrum für Allgemein- und Familienmedizin, Karl Landsteiner Privatuniversität für Gesundheitswissenschaften, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Österreich.
| | - Kathryn Hoffmann
- Leiterin der Abteilung Primary Care Medicine, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich
| | - Martin Aigner
- Abteilung für Psychiatrie und psychotherapeutische Medizin, Karl Landsteiner Privatuniversität für Gesundheitswissenschaften, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Österreich
| | - Johann Altenberger
- Pensionsversicherungsanstalt, Rehabilitationszentrum Großgmain, Salzburger Str. 520, 5084, Großgmain, Österreich
| | - Markus Brose
- Department Allgemeine Gesundheitsstudien, Kompetenzzentrum für Allgemein- und Familienmedizin, Karl Landsteiner Privatuniversität für Gesundheitswissenschaften, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Österreich
| | - Ursula Costa
- Ergotherapie und Handlungswissenschaft, fhg - Zentrum für Gesundheitsberufe Tirol GmbH/fh, Innrain 98, 6020, Innsbruck, Österreich
| | - Doris-Maria Denk-Linnert
- Klinische Abteilung für Allgemeine Hals‑, Nasen- und Ohrenkrankheiten, Klin. Abteilung Phoniatrie-Logopädie, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich
| | - Samuel Gruber
- Department Allgemeine Gesundheitsstudien, Kompetenzzentrum für Allgemein- und Familienmedizin, Karl Landsteiner Privatuniversität für Gesundheitswissenschaften, Dr. Karl-Dorrek-Str. 30, 3500, Krems, Österreich
| | - Florian Götzinger
- Abteilung für Kinderheilkunde, Klinik Ottakring, Montleartstr. 37, 1160, Wien, Österreich
| | - Raimund Helbok
- Universitätsklinik für Neurologie, Johannes Kepler Universität Linz, Standort Neuromed Campus & Med Campus Kepler Universitätsklinikum GmbH, 4020, Linz, Österreich
| | - Katharina Hüfner
- Dep. für Psychiatrie, Psychotherapie, Psychosomatik und Medizinische Psychologie, Universitätsklinik für Psychiatrie II, Medizinische Universität Innsbruck, Anichstr. 35, 6020, Innsbruck, Österreich
| | - Rembert Koczulla
- Fachbereich Medizin, Klinik für Pneumologie Marburg, Baldingerstr., 35035, Marburg, Deutschland
| | - Katharina Kurz
- Innere Medizin II, Medizinische Universität Innsbruck, Anichstr. 35, 6020, Innsbruck, Österreich
| | - Bernd Lamprecht
- Universitätsklinik für Innere Medizin mit Schwerpunkt Pneumologie, Kepler Universitätsklinikum, 4020, Linz, Österreich
| | - Stefan Leis
- Universitätsklinik für Neurologie der PMU, MME Universitätsklinikum Salzburg Christian-Doppler-Klinik, Ignaz-Harrer-Str. 79, 5020, Salzburg, Österreich
| | - Judith Löffler
- Innere Medizin II, Medizinische Universität Innsbruck, Anichstr. 35, 6020, Innsbruck, Österreich
| | - Christian A Müller
- Klinische Abteilung für Allgemeine Hals‑, Nasen- und Ohrenkrankheiten, Klin. Abteilung für Allgemeine HNO, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich
| | | | - Paulus S Rommer
- Universitätsklinik für Neurologie, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich
| | - Paul Sator
- Dermatologische Abteilung, Klinik Hietzing, Wolkersbergenstr. 1, 1130, Wien, Österreich
| | - Volker Strenger
- Klinische Abteilung für Allgemeinpädiatrie, Universitätsklinik für Kinder- und Jugendheilkunde, Medizinische Universität Graz, 8036, Graz, Österreich
| | - Walter Struhal
- Klinische Abteilung für Neurologie, Universitätsklinikum Tulln, Karl Landsteiner Privatuniversität für Gesundheitswissenschaften, Alter Ziegelweg 10, 3430, Tulln an der Donau, Österreich
| | - Eva Untersmayr
- Institut für Pathophysiologie und Allergieforschung Zentrum für Pathophysiologie, Infektiologie und Immunologie, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich
| | - Karin Vonbank
- Klinische Abteilung für Pulmologie, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich
| | - Johannes Wancata
- Klinische Abteilung für Sozialpsychiatrie, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090, Wien, Österreich
| | - Thomas Weber
- Kardiologische Abteilung Klinikum Wels-Grieskirchen, Grieskirchnerstr. 42, 4600, Wels, Österreich
| | | | - Ralf-Harun Zwick
- Ludwig Boltzmann Institute for Rehabilitation Research, Kurbadstr. 14, 1100, Wien, Österreich
| |
Collapse
|
19
|
Nelson MC, Manos CK, Flanagan E, Prahalad S. COVID-19 after rituximab therapy in cSLE patients. Ther Adv Vaccines Immunother 2023; 11:25151355231181242. [PMID: 37362155 PMCID: PMC10285438 DOI: 10.1177/25151355231181242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Childhood-onset systemic lupus erythematosus (cSLE) is an autoimmune disease associated with significant morbidity and mortality. Rituximab is a B-cell depleting therapy utilized in the treatment of SLE. In adults, rituximab has been associated with increased risk of adverse outcomes in patients who develop coronavirus disease 2019 (COVID-19). We aimed to assess the impact of prior rituximab treatment on clinical outcomes from Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection in children with SLE. To describe the impact of rituximab on outcomes from SARS-CoV-2 infection, we conducted a retrospective study of pediatric SLE patients in our center diagnosed with COVID-19 who had previously received rituximab between February 2019 and October 2022. Patients' clinical characteristics, disease activity, and outcomes were assessed. Of the eight subjects assessed, five required hospitalizations for COVID-19, four required ICU admission, and two were seen in the emergency department for their symptoms. One patient ultimately expired from her illness. The median time between rituximab administration and COVID-19 diagnosis was 3 months. We assessed the clinical outcomes, including the need of ICU admission and fatal outcome, of COVID-19 in our cSLE patient population after rituximab administration. Approximately 60% of our patients required hospitalization for their illness, and seven out of eight patients required healthcare utilization to include hospitalization and/or emergency department visits.
Collapse
Affiliation(s)
| | - Cynthia K. Manos
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USAChildren’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Elaine Flanagan
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USAChildren’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Sampath Prahalad
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USAChildren’s Healthcare of Atlanta, Atlanta, GA, USADepartment of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
20
|
Vavilova JD, Ustiuzhanina MO, Boyko AA, Streltsova MA, Kust SA, Kanevskiy LM, Iskhakov RN, Sapozhnikov AM, Gubernatorova EO, Drutskaya MS, Bychinin MV, Novikova ON, Sotnikova AG, Yusubalieva GM, Baklaushev VP, Kovalenko EI. Alterations in the CD56 - and CD56 + T Cell Subsets during COVID-19. Int J Mol Sci 2023; 24:9047. [PMID: 37240393 PMCID: PMC10219320 DOI: 10.3390/ijms24109047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/18/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
The effectiveness of the antiviral immune response largely depends on the activation of cytotoxic T cells. The heterogeneous group of functionally active T cells expressing the CD56 molecule (NKT-like cells), that combines the properties of T lymphocytes and NK cells, is poorly studied in COVID-19. This work aimed to analyze the activation and differentiation of both circulating NKT-like cells and CD56- T cells during COVID-19 among intensive care unit (ICU) patients, moderate severity (MS) patients, and convalescents. A decreased proportion of CD56+ T cells was found in ICU patients with fatal outcome. Severe COVID-19 was accompanied by a decrease in the proportion of CD8+ T cells, mainly due to the CD56- cell death, and a redistribution of the NKT-like cell subset composition with a predominance of more differentiated cytotoxic CD8+ T cells. The differentiation process was accompanied by an increase in the proportions of KIR2DL2/3+ and NKp30+ cells in the CD56+ T cell subset of COVID-19 patients and convalescents. Decreased percentages of NKG2D+ and NKG2A+ cells and increased PD-1 and HLA-DR expression levels were found in both CD56- and CD56+ T cells, and can be considered as indicators of COVID-19 progression. In the CD56- T cell fraction, increased CD16 levels were observed in MS patients and in ICU patients with lethal outcome, suggesting a negative role for CD56-CD16+ T cells in COVID-19. Overall, our findings suggest an antiviral role of CD56+ T cells in COVID-19.
Collapse
Affiliation(s)
- Julia D. Vavilova
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (J.D.V.); (S.A.K.); (A.M.S.)
| | - Maria O. Ustiuzhanina
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (J.D.V.); (S.A.K.); (A.M.S.)
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
| | - Anna A. Boyko
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (J.D.V.); (S.A.K.); (A.M.S.)
| | - Maria A. Streltsova
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (J.D.V.); (S.A.K.); (A.M.S.)
| | - Sofya A. Kust
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (J.D.V.); (S.A.K.); (A.M.S.)
| | - Leonid M. Kanevskiy
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (J.D.V.); (S.A.K.); (A.M.S.)
| | - Rustam N. Iskhakov
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (J.D.V.); (S.A.K.); (A.M.S.)
| | - Alexander M. Sapozhnikov
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (J.D.V.); (S.A.K.); (A.M.S.)
| | - Ekaterina O. Gubernatorova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Marina S. Drutskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Division of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sirius, Krasnodarsky Krai, 354349 Sochi, Russia
| | - Mikhail V. Bychinin
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
| | - Oksana N. Novikova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
| | - Anna G. Sotnikova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladimir P. Baklaushev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elena I. Kovalenko
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (J.D.V.); (S.A.K.); (A.M.S.)
| |
Collapse
|
21
|
Xiang M, Wu X, Jing H, Novakovic VA, Shi J. The intersection of obesity and (long) COVID-19: Hypoxia, thrombotic inflammation, and vascular endothelial injury. Front Cardiovasc Med 2023; 10:1062491. [PMID: 36824451 PMCID: PMC9941162 DOI: 10.3389/fcvm.2023.1062491] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
The role of hypoxia, vascular endothelial injury, and thrombotic inflammation in worsening COVID-19 symptoms has been generally recognized. Damaged vascular endothelium plays a crucial role in forming in situ thrombosis, pulmonary dysfunction, and hypoxemia. Thrombotic inflammation can further aggravate local vascular endothelial injury and affect ventilation and blood flow ratio. According to the results of many studies, obesity is an independent risk factor for a variety of severe respiratory diseases and contributes to high mechanical ventilation rate, high mortality, and slow recovery in COVID-19 patients. This review will explore the mechanisms by which obesity may aggravate the acute phase of COVID-19 and delay long COVID recovery by affecting hypoxia, vascular endothelial injury, and thrombotic inflammation. A systematic search of PubMed database was conducted for papers published since January 2020, using the medical subject headings of "COVID-19" and "long COVID" combined with the following keywords: "obesity," "thrombosis," "endothelial injury," "inflammation," "hypoxia," "treatment," and "anticoagulation." In patients with obesity, the accumulation of central fat restricts the expansion of alveoli, exacerbating the pulmonary dysfunction caused by SARS-CoV-2 invasion, inflammatory damage, and lung edema. Abnormal fat secretion and immune impairment further aggravate the original tissue damage and inflammation diffusion. Obesity weakens baseline vascular endothelium function leading to an early injury and pre-thrombotic state after infection. Enhanced procoagulant activity and microthrombi promote early obstruction of the vascular. Obesity also prolongs the duration of symptoms and increases the risk of sequelae after hospital discharge. Persistent viral presence, long-term inflammation, microclots, and hypoxia may contribute to the development of persistent symptoms, suggesting that patients with obesity are uniquely susceptible to long COVID. Early interventions, including supplemental oxygen, comprehensive antithrombotic therapy, and anti-inflammatory drugs, show effectiveness in many studies in the prevention of serious hypoxia, thromboembolic events, and systemic inflammation, and are therefore recommended to reduce intensive care unit admission, mortality, and sequelae.
Collapse
Affiliation(s)
- Mengqi Xiang
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Xiaoming Wu
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Haijiao Jing
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Valerie A. Novakovic
- Department of Research, Veterans Affairs Boston Healthcare System and Harvard Medical School, Boston, MA, United States
| | - Jialan Shi
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
- Department of Research, Veterans Affairs Boston Healthcare System and Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
22
|
Hopkins FR, Govender M, Svanberg C, Nordgren J, Waller H, Nilsdotter-Augustinsson Å, Henningsson AJ, Hagbom M, Sjöwall J, Nyström S, Larsson M. Major alterations to monocyte and dendritic cell subsets lasting more than 6 months after hospitalization for COVID-19. Front Immunol 2023; 13:1082912. [PMID: 36685582 PMCID: PMC9846644 DOI: 10.3389/fimmu.2022.1082912] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction After more than two years the Coronavirus disease-19 (COVID-19) pandemic continues to burden healthcare systems and economies worldwide, and it is evident that the effects on the immune system can persist for months post-infection. The activity of myeloid cells such as monocytes and dendritic cells (DC) is essential for correct mobilization of the innate and adaptive responses to a pathogen. Impaired levels and responses of monocytes and DC to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is likely to be a driving force behind the immune dysregulation that characterizes severe COVID-19. Methods Here, we followed a cohort of COVID-19 patients hospitalized during the early waves of the pandemic for 6-7 months. The levels and phenotypes of circulating monocyte and DC subsets were assessed to determine both the early and long-term effects of the SARS-CoV-2 infection. Results We found increased monocyte levels that persisted for 6-7 months, mostly attributed to elevated levels of classical monocytes. Myeloid derived suppressor cells were also elevated over this period. While most DC subsets recovered from an initial decrease, we found elevated levels of cDC2/cDC3 at the 6-7 month timepoint. Analysis of functional markers on monocytes and DC revealed sustained reduction in program death ligand 1 (PD-L1) expression but increased CD86 expression across almost all cell types examined. Finally, C-reactive protein (CRP) correlated positively to the levels of intermediate monocytes and negatively to the recovery of DC subsets. Conclusion By exploring the myeloid compartments, we show here that alterations in the immune landscape remain more than 6 months after severe COVID-19, which could be indicative of ongoing healing and/or persistence of viral antigens.
Collapse
Affiliation(s)
- Francis R. Hopkins
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Melissa Govender
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Cecilia Svanberg
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johan Nordgren
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Hjalmar Waller
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Åsa Nilsdotter-Augustinsson
- Division of Infection and Inflammation, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Infectious Diseases, Linköping University, Linköping, Sweden
| | - Anna J. Henningsson
- Division of Infection and Inflammation, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Division of Clinical Microbiology, Department of Laboratory Medicine in Jönköping, Ryhov County Hospital, Jönköping, Sweden
| | - Marie Hagbom
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johanna Sjöwall
- Division of Infection and Inflammation, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Infectious Diseases, Linköping University, Linköping, Sweden
| | - Sofia Nyström
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Linköping University, Linköping, Sweden
| | - Marie Larsson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|