1
|
Kang N, Chawla A, Hillman H, Tippalagama R, Kim C, Mikulski Z, Seumois G, Vijayanand P, Scriba TJ, De Silva AD, Balmaseda A, Harris E, Weiskopf D, Sette A, Arlehamn CL, Peters B, Burel JG. A novel method for characterizing cell-cell interactions at single-cell resolution reveals unique signatures in blood T cell-monocyte complexes during infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.20.612103. [PMID: 39386643 PMCID: PMC11463634 DOI: 10.1101/2024.09.20.612103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Communication between immune cells through direct contact is a critical feature of immune responses. Here, we developed a novel high-throughput method to study the transcriptome and adaptive immune receptor repertoire of single cells forming complexes without needing bioinformatic deconvolution. We found that T cells and monocytes forming complexes in blood during active tuberculosis (TB) and dengue hold unique transcriptomic signatures indicative of TCR/MCH-II immune synapses. Additionally, T cells in complexes showed enrichment for effector phenotypes, imaging and transcriptomic features of active TCR signaling, and increased immune activity at diagnosis compared to after anti-TB therapy. We also found evidence for bidirectional RNA exchange between T cells and monocytes, since complexes were markedly enriched for "dual-expressing" cells (i.e., co-expressing T cell and monocyte genes). Thus, studying immune cell complexes at a single-cell resolution offers novel perspectives on immune synaptic interactions occurring in blood during infection.
Collapse
Affiliation(s)
- Ningxin Kang
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
| | - Ashu Chawla
- Bioinformatics Core, La Jolla Institute for Immunology, CA 92037, United States
| | - Hannah Hillman
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
| | - Rashmi Tippalagama
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
| | - Cheryl Kim
- Flow Cytometry Core, La Jolla Institute for Immunology, CA 92037, United States
| | - Zbigniew Mikulski
- Microscopy Core, La Jolla Institute for Immunology, CA 92037, United States
| | - Grégory Seumois
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, CA, United States
| | - Pandurangan Vijayanand
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, South Africa
| | - Aruna D De Silva
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
- Faculty of Medicine, General Sir John Kotelawala Defence University, Sri Lanka
| | | | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA 94720-3370, USA
| | - Daniela Weiskopf
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | | | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego (UCSD), La Jolla, CA 92037, USA
| | - Julie G Burel
- Center for Vaccine Innovation, La Jolla Institute for Immunology, CA 92037, United States
| |
Collapse
|
2
|
Obare LM, Simmons J, Oakes J, Zhang X, Nochowicz C, Priest S, Bailin SS, Warren CM, Mashayekhi M, Beasley HK, Shao J, Meenderink LM, Sheng Q, Stolze J, Gangula R, Absi T, Su YR, Neikirk K, Chopra A, Gabriel CL, Temu T, Pakala S, Wilfong EM, Gianella S, Phillips EJ, Harrison DG, Hinton A, Kalams SA, Kirabo A, Mallal SA, Koethe JR, Wanjalla CN. CD3 + T-cell: CD14 +monocyte complexes are dynamic and increased with HIV and glucose intolerance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.24.538020. [PMID: 37162990 PMCID: PMC10168203 DOI: 10.1101/2023.04.24.538020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
An increased risk of cardiometabolic disease accompanies persistent systemic inflammation. Yet, the innate and adaptive immune system features in persons who develop these conditions remain poorly defined. Doublets, or cell-cell complexes, are routinely eliminated from flow cytometric and other immune phenotyping analyses, which limits our understanding of their relationship to disease states. Using well-characterized clinical cohorts, including participants with controlled HIV as a model for chronic inflammation and increased immune cell interactions, we show that circulating CD14+ monocytes complexed to CD3+ T cells are dynamic, biologically relevant, and increased in individuals with diabetes after adjusting for confounding factors. The complexes form functional immune synapses with increased expression of proinflammatory cytokines and greater glucose utilization. Furthermore, in persons with HIV, the CD3+T-cell: CD14+monocyte complexes had more HIV copies compared to matched CD14+ monocytes or CD4+ T cells alone. Our results demonstrate that circulating CD3+T-cell:CD14+monocyte pairs represent dynamic cellular interactions that may contribute to inflammation and cardiometabolic disease pathogenesis and may originate or be maintained, in part, by chronic viral infections. These findings provide a foundation for future studies investigating mechanisms linking T cellmonocyte cell-cell complexes to developing immune-mediated diseases, including HIV and diabetes.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua Simmons
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jared Oakes
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xiuqi Zhang
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cindy Nochowicz
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephen Priest
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Samuel S. Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mona Mashayekhi
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, USA
| | - Leslie M. Meenderink
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - Joey Stolze
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - Rama Gangula
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yan Ru Su
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Curtis L. Gabriel
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tecla Temu
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Suman Pakala
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erin M. Wilfong
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara Gianella
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth J. Phillips
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David G. Harrison
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Spyros A. Kalams
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - John R. Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
3
|
Madruga MP, Grun LK, Santos LSMD, Friedrich FO, Antunes DB, Rocha MEF, Silva PL, Dorneles GP, Teixeira PC, Oliveira TF, Romão PRT, Santos L, Moreira JCF, Michaelsen VS, Cypel M, Antunes MOB, Jones MH, Barbé-Tuana FM, Bauer ME. Excess of body weight is associated with accelerated T-cell senescence in hospitalized COVID-19 patients. Immun Ageing 2024; 21:17. [PMID: 38454515 PMCID: PMC10921685 DOI: 10.1186/s12979-024-00423-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/28/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Several risk factors have been involved in the poor clinical progression of coronavirus disease-19 (COVID-19), including ageing, and obesity. SARS-CoV-2 may compromise lung function through cell damage and paracrine inflammation; and obesity has been associated with premature immunosenescence, microbial translocation, and dysfunctional innate immune responses leading to poor immune response against a range of viruses and bacterial infections. Here, we have comprehensively characterized the immunosenescence, microbial translocation, and immune dysregulation established in hospitalized COVID-19 patients with different degrees of body weight. RESULTS Hospitalised COVID-19 patients with overweight and obesity had similarly higher plasma LPS and sCD14 levels than controls (all p < 0.01). Patients with obesity had higher leptin levels than controls. Obesity and overweight patients had similarly higher expansions of classical monocytes and immature natural killer (NK) cells (CD56+CD16-) than controls. In contrast, reduced proportions of intermediate monocytes, mature NK cells (CD56+CD16+), and NKT were found in both groups of patients than controls. As expected, COVID-19 patients had a robust expansion of plasmablasts, contrasting to lower proportions of major T-cell subsets (CD4 + and CD8+) than controls. Concerning T-cell activation, overweight and obese patients had lower proportions of CD4+CD38+ cells than controls. Contrasting changes were reported in CD25+CD127low/neg regulatory T cells, with increased and decreased proportions found in CD4+ and CD8+ T cells, respectively. There were similar proportions of T cells expressing checkpoint inhibitors across all groups. We also investigated distinct stages of T-cell differentiation (early, intermediate, and late-differentiated - TEMRA). The intermediate-differentiated CD4 + T cells and TEMRA cells (CD4+ and CD8+) were expanded in patients compared to controls. Senescent T cells can also express NK receptors (NKG2A/D), and patients had a robust expansion of CD8+CD57+NKG2A+ cells than controls. Unbiased immune profiling further confirmed the expansions of senescent T cells in COVID-19. CONCLUSIONS These findings suggest that dysregulated immune cells, microbial translocation, and T-cell senescence may partially explain the increased vulnerability to COVID-19 in subjects with excess of body weight.
Collapse
Affiliation(s)
- Mailton Prestes Madruga
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Lucas Kich Grun
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Letícya Simone Melo Dos Santos
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | | | - Douglas Bitencourt Antunes
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Marcella Elesbão Fogaça Rocha
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Pedro Luis Silva
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Gilson P Dorneles
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Paula Coelho Teixeira
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Tiago Franco Oliveira
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Pedro R T Romão
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Lucas Santos
- Centro de Estudos em Estresse Oxidativo - Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (IB-UFRGS), Porto Alegre, RS, Brazil
| | - José Claudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo - Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (IB-UFRGS), Porto Alegre, RS, Brazil
| | - Vinicius Schenk Michaelsen
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Marcelo Cypel
- Toronto General Hospital Research Institute, Department of Surgery, University Health Network, University of Toronto, Toronto, Canada
| | - Marcos Otávio Brum Antunes
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Marcus Herbert Jones
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Florencia María Barbé-Tuana
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Moisés Evandro Bauer
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil.
| |
Collapse
|
4
|
Zhao J, Zhang H, Jiang L, Cheng F, Li W, Wang Z, Liu H, Li S, Jiang Y, Li M, Li Y, Liu S, Fang M, Zhou X, Ye X, Zhao S, Zheng Y, Meng S. Increased antibody titers but induced T cell AICD and apoptosis response in COVID-19 convalescents by inactivated vaccine booster. Microbiol Spectr 2024; 12:e0243523. [PMID: 38319108 PMCID: PMC10913726 DOI: 10.1128/spectrum.02435-23] [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: 06/10/2023] [Accepted: 01/12/2024] [Indexed: 02/07/2024] Open
Abstract
It is urgently needed to evaluate the necessity and benefits of booster vaccination against the coronavirus 2 of the severe acute respiratory syndrome (SARS-CoV-2) Omicron to facilitate clinical decision-making for 2019 coronavirus disease (COVID-19) convalescents. We conducted a multicenter, prospective clinical trial (registration number: ChiCTR2100045810) in the first patients with COVID-19 from 28 January 2020 to 20 February 2020 to assess the long-term durability of neutralizing antibodies against live Omicron BA.5 and further assess the efficiency and safety of CoronaVac in the convalescent group. A total of 96 COVID-19 convalescents were enrolled in this study. Neutralizing antibody titers in convalescents were significantly reduced in 9-10 months. A dose-refreshing vaccination in 28 convalescents with an antibody titer below 96 significantly induced neutralizing antibodies against live Omicron by 4.84-fold. Meanwhile, the abundance of naive T cells increased dramatically, and TEMRA and TEM cells gradually decreased after vaccination. Activation-induced cell death and apoptosis-related genes were significantly elevated after vaccination in all T-cell subtypes. One-dose booster vaccination was effective in inducing a robust antibody response against SARS-CoV-2 Omicron in COVID-19 convalescents with low antibody titers. However, vaccine-mediated T-cell consumption and regeneration patterns may be detrimental to the antiviral response.IMPORTANCEThe globally dominant coronavirus 2 of the severe acute respiratory syndrome (SARS-CoV-2) Omicron variant raises the possibility of repeat infections among 2019 coronavirus disease (COVID-19) convalescents with low neutralizing antibody titers. The importance of this multicenter study lies in its evaluation of the long-term durability of neutralizing antibodies in COVID-19 convalescents and the efficacy of a booster vaccination against the live Omicron. The findings suggest that a one-dose booster vaccination is effective in inducing a robust antibody response against SARS-CoV-2 Omicron in convalescents with low antibody titers. However, the study also highlights the potential detrimental effects on the antiviral response due to vaccine-mediated T-cell consumption and regeneration patterns. These results are crucial for facilitating clinical decision-making for COVID-19 convalescents and informing public health policies regarding booster vaccinations.
Collapse
Affiliation(s)
- Jingmin Zhao
- Department of Pathology and Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Han Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lina Jiang
- Department of Pathology and Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Fang Cheng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- Department of Infectious Diseases, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zihao Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hongyang Liu
- Department of Pathology and Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Shaohua Li
- Department of Pathology and Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yiyun Jiang
- Department of Pathology and Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Meiling Li
- Department of Pathology and Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yan Li
- Department of Pathology and Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Shuhong Liu
- Department of Pathology and Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Min Fang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuyu Zhou
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin Ye
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shousong Zhao
- Department of Infectious Diseases, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yuxuan Zheng
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Songdong Meng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
5
|
May L, Chu CF, Zielinski CE. Single-Cell RNA Sequencing Reveals HIF1A as a Severity-Sensitive Immunological Scar in Circulating Monocytes of Convalescent Comorbidity-Free COVID-19 Patients. Cells 2024; 13:300. [PMID: 38391913 PMCID: PMC10886588 DOI: 10.3390/cells13040300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
COVID-19, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is characterized by a wide range of clinical symptoms and a poorly predictable disease course. Although in-depth transcriptomic investigations of peripheral blood samples from COVID-19 patients have been performed, the detailed molecular mechanisms underlying an asymptomatic, mild or severe disease course, particularly in patients without relevant comorbidities, remain poorly understood. While previous studies have mainly focused on the cellular and molecular dissection of ongoing COVID-19, we set out to characterize transcriptomic immune cell dysregulation at the single-cell level at different time points in patients without comorbidities after disease resolution to identify signatures of different disease severities in convalescence. With single-cell RNA sequencing, we reveal a role for hypoxia-inducible factor 1-alpha (HIF1A) as a severity-sensitive long-term immunological scar in circulating monocytes of convalescent COVID-19 patients. Additionally, we show that circulating complexes formed by monocytes with either T cells or NK cells represent a characteristic cellular marker in convalescent COVID-19 patients irrespective of their preceding symptom severity. Together, these results provide cellular and molecular correlates of recovery from COVID-19 and could help in immune monitoring and in the design of new treatment strategies.
Collapse
Affiliation(s)
- Lilly May
- Leibniz Institute for Natural Products Research and Infection Biology, Department of Infection Immunology, 07745 Jena, Germany; (L.M.); (C.-F.C.)
- Center for Translational Cancer Research (TranslaTUM) & Institute of Virology, Technical University of Munich, 81675 Munich, Germany
| | - Chang-Feng Chu
- Leibniz Institute for Natural Products Research and Infection Biology, Department of Infection Immunology, 07745 Jena, Germany; (L.M.); (C.-F.C.)
- Center for Translational Cancer Research (TranslaTUM) & Institute of Virology, Technical University of Munich, 81675 Munich, Germany
| | - Christina E. Zielinski
- Leibniz Institute for Natural Products Research and Infection Biology, Department of Infection Immunology, 07745 Jena, Germany; (L.M.); (C.-F.C.)
- Center for Translational Cancer Research (TranslaTUM) & Institute of Virology, Technical University of Munich, 81675 Munich, Germany
- Department of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
| |
Collapse
|
6
|
Rubinstein A, Kudryavtsev I, Malkova A, Mammedova J, Isakov D, Isakova-Sivak I, Kudlay D, Starshinova A. Sarcoidosis-related autoimmune inflammation in COVID-19 convalescent patients. Front Med (Lausanne) 2023; 10:1271198. [PMID: 38179278 PMCID: PMC10765615 DOI: 10.3389/fmed.2023.1271198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Currently, there are a large number of reports about the development of autoimmune conditions after COVID-19. Also, there have been cases of sarcoid-like granulomas in convalescents as a part of the post-COVID-19 syndrome. Since one of the etiological theories of sarcoidosis considers it to be an autoimmune disease, we decided to study changes in the adaptive humoral immune response in sarcoidosis and SARS-CoV-2 infection and to find out whether COVID-19 can provoke the development of sarcoidosis. This review discusses histological changes in lymphoid organs in sarcoidosis and COVID-19, changes in B cell subpopulations, T-follicular helper cells (Tfh), and T-follicular regulatory cells (Tfr), and analyzes various autoantibodies detected in these pathologies. Based on the data studied, we concluded that SARS-CoV-2 infection may cause the development of autoimmune pathologies, in particular contributing to the onset of sarcoidosis in convalescents.
Collapse
Affiliation(s)
- Artem Rubinstein
- Almazov National Medical Research Centre, Saint Petersburg, Russia
- Institution of Experimental Medicine, Saint Petersburg, Russia
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, Saint Petersburg, Russia
- Institution of Experimental Medicine, Saint Petersburg, Russia
- Far Eastern Federal University, Vladivostok, Russia
| | - Annа Malkova
- Ariel University Faculty of Natural Sciences, Ariel, Israel
| | | | - Dmitry Isakov
- First Saint Petersburg State I. Pavlov Medical University, Saint Petersburg, Russia
| | | | - Dmitry Kudlay
- Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- NRC Institute of Immunology, Moscow, Russia
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Moscow, Russia
| | - Anna Starshinova
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| |
Collapse
|
7
|
Pang K, Dong S, Zhu Y, Zhu X, Zhou Q, Gu B, Jin W, Zhang R, Fu Y, Yu B, Sun D, Duanmu Z, Wei X. Advanced flow cytometry for biomedical applications. JOURNAL OF BIOPHOTONICS 2023; 16:e202300135. [PMID: 37263969 DOI: 10.1002/jbio.202300135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/03/2023]
Abstract
Flow cytometry (FC) is a versatile tool with excellent capabilities to detect and measure multiple characteristics of a population of cells or particles. Notable advancements in in vivo photoacoustic FC, coherent Raman FC, microfluidic FC, and so on, have been achieved in the last two decades, which endows FC with new functions and expands its applications in basic research and clinical practice. Advanced FC broadens the tools available to researchers to conduct research involving cancer detection, microbiology (COVID-19, HIV, bacteria, etc.), and nucleic acid analysis. This review presents an overall picture of advanced flow cytometers and provides not only a clear understanding of their mechanisms but also new insights into their practical applications. We identify the latest trends in this area and aim to raise awareness of advanced techniques of FC. We hope this review expands the applications of FC and accelerates its clinical translation.
Collapse
Affiliation(s)
- Kai Pang
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Sihan Dong
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Yuxi Zhu
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Xi Zhu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Quanyu Zhou
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Bobo Gu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Jin
- International Cancer Institute, Peking University, Beijing, China
| | - Rui Zhang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Yuting Fu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Bingchen Yu
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Da Sun
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Zheng Duanmu
- School of Instrument Science and Opto-Electronics Engineering of Beijing Information Science & Technology University, Beijing, China
| | - Xunbin Wei
- International Cancer Institute, Peking University, Beijing, China
| |
Collapse
|
8
|
Mina Y, Enose-Akahata Y, Hammoud DA, Videckis AJ, Narpala SR, O'Connell SE, Carroll R, Lin BC, McMahan CC, Nair G, Reoma LB, McDermott AB, Walitt B, Jacobson S, Goldstein DS, Smith BR, Nath A. Deep Phenotyping of Neurologic Postacute Sequelae of SARS-CoV-2 Infection. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/4/e200097. [PMID: 37147136 PMCID: PMC10162706 DOI: 10.1212/nxi.0000000000200097] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/04/2023] [Indexed: 05/07/2023]
Abstract
BACKGROUND AND OBJECTIVES SARS-CoV-2 infection has been associated with a syndrome of long-term neurologic sequelae that is poorly characterized. We aimed to describe and characterize in-depth features of neurologic postacute sequelae of SARS-CoV-2 infection (neuro-PASC). METHODS Between October 2020 and April 2021, 12 participants were seen at the NIH Clinical Center under an observational study to characterize ongoing neurologic abnormalities after SARS-CoV-2 infection. Autonomic function and CSF immunophenotypic analysis were compared with healthy volunteers (HVs) without prior SARS-CoV-2 infection tested using the same methodology. RESULTS Participants were mostly female (83%), with a mean age of 45 ± 11 years. The median time of evaluation was 9 months after COVID-19 (range 3-12 months), and most (11/12, 92%) had a history of only a mild infection. The most common neuro-PASC symptoms were cognitive difficulties and fatigue, and there was evidence for mild cognitive impairment in half of the patients (MoCA score <26). The majority (83%) had a very disabling disease, with Karnofsky Performance Status ≤80. Smell testing demonstrated different degrees of microsmia in 8 participants (66%). Brain MRI scans were normal, except 1 patient with bilateral olfactory bulb hypoplasia that was likely congenital. CSF analysis showed evidence of unique intrathecal oligoclonal bands in 3 cases (25%). Immunophenotyping of CSF compared with HVs showed that patients with neuro-PASC had lower frequencies of effector memory phenotype both for CD4+ T cells (p < 0.0001) and for CD8+ T cells (p = 0.002), an increased frequency of antibody-secreting B cells (p = 0.009), and increased frequency of cells expressing immune checkpoint molecules. On autonomic testing, there was evidence for decreased baroreflex-cardiovagal gain (p = 0.009) and an increased peripheral resistance during tilt-table testing (p < 0.0001) compared with HVs, without excessive plasma catecholamine responses. DISCUSSION CSF immune dysregulation and neurocirculatory abnormalities after SARS-CoV-2 infection in the setting of disabling neuro-PASC call for further evaluation to confirm these changes and explore immunomodulatory treatments in the context of clinical trials.
Collapse
Affiliation(s)
- Yair Mina
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Yoshimi Enose-Akahata
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Dima A Hammoud
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Anthony J Videckis
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Sandeep R Narpala
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Sarah E O'Connell
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Robin Carroll
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Bob C Lin
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Cynthia Chen McMahan
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Govind Nair
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Lauren B Reoma
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Adrian B McDermott
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Brian Walitt
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Steven Jacobson
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - David S Goldstein
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Bryan R Smith
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Avindra Nath
- From the National Institute of Neurological Disorders and Stroke (Y.M., Y.E.-A., A.J.V., C.C.M., G.N., L.B.R., B.W., S.J., D.S.G., B.R.S., A.N.), National Institutes of Health, Bethesda, MD; Sackler Faculty of Medicine (Y.M.), Tel-Aviv University, Israel; Center for Infectious Disease Imaging (D.A.H.), Radiology and Imaging Sciences, Clinical Center, National Institutes of Health; and Vaccine Immunology Program (S.R.N., S.E.O.C., R.C., B.C.L., A.B.M.), Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
9
|
Gil-Manso S, Herrero-Quevedo D, Carbonell D, Martínez-Bonet M, Bernaldo-de-Quirós E, Kennedy-Batalla R, Gallego-Valle J, López-Esteban R, Blázquez-López E, Miguens-Blanco I, Correa-Rocha R, Gomez-Verdejo V, Pion M. Multidimensional analysis of immune cells from COVID-19 patients identified cell subsets associated with the severity at hospital admission. PLoS Pathog 2023; 19:e1011432. [PMID: 37311004 PMCID: PMC10263360 DOI: 10.1371/journal.ppat.1011432] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/18/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND SARS-CoV-2 emerged as a new coronavirus causing COVID-19, and it has been responsible for more than 760 million cases and 6.8 million deaths worldwide until March 2023. Although infected individuals could be asymptomatic, other patients presented heterogeneity and a wide range of symptoms. Therefore, identifying those infected individuals and being able to classify them according to their expected severity could help target health efforts more effectively. METHODOLOGY/PRINCIPAL FINDINGS Therefore, we wanted to develop a machine learning model to predict those who will develop severe disease at the moment of hospital admission. We recruited 75 individuals and analysed innate and adaptive immune system subsets by flow cytometry. Also, we collected clinical and biochemical information. The objective of the study was to leverage machine learning techniques to identify clinical features associated with disease severity progression. Additionally, the study sought to elucidate the specific cellular subsets involved in the disease following the onset of symptoms. Among the several machine learning models tested, we found that the Elastic Net model was the better to predict the severity score according to a modified WHO classification. This model was able to predict the severity score of 72 out of 75 individuals. Besides, all the machine learning models revealed that CD38+ Treg and CD16+ CD56neg HLA-DR+ NK cells were highly correlated with the severity. CONCLUSIONS/SIGNIFICANCE The Elastic Net model could stratify the uninfected individuals and the COVID-19 patients from asymptomatic to severe COVID-19 patients. On the other hand, these cellular subsets presented here could help to understand better the induction and progression of the symptoms in COVID-19 individuals.
Collapse
Affiliation(s)
- Sergio Gil-Manso
- Advanced ImmunoRegulation Group, Gregorio Marañón Health Research Institute (IiSGM), General University Hospital Gregorio Marañón, Madrid, Spain
| | - Diego Herrero-Quevedo
- Signal Processing and Communications Department, University Carlos III de Madrid, Leganés, Madrid, Spain
| | - Diego Carbonell
- Department of Hematology, General University Hospital Gregorio Marañón (HGUGM), Madrid, Spain
- Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Marta Martínez-Bonet
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IiSGM), General University Hospital Gregorio Marañón, Madrid, Spain
| | - Esther Bernaldo-de-Quirós
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IiSGM), General University Hospital Gregorio Marañón, Madrid, Spain
| | - Rebeca Kennedy-Batalla
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IiSGM), General University Hospital Gregorio Marañón, Madrid, Spain
| | - Jorge Gallego-Valle
- Advanced ImmunoRegulation Group, Gregorio Marañón Health Research Institute (IiSGM), General University Hospital Gregorio Marañón, Madrid, Spain
| | - Rocío López-Esteban
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IiSGM), General University Hospital Gregorio Marañón, Madrid, Spain
| | - Elena Blázquez-López
- Gastroenterology—Digestive Service, General University Hospital Gregorio Marañón, Network of Hepatic and Digestive Diseases (CIBEREHD), Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Iria Miguens-Blanco
- Emergency Department, General University Hospital Gregorio Marañón, Madrid, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IiSGM), General University Hospital Gregorio Marañón, Madrid, Spain
| | - Vanessa Gomez-Verdejo
- Signal Processing and Communications Department, University Carlos III de Madrid, Leganés, Madrid, Spain
| | - Marjorie Pion
- Advanced ImmunoRegulation Group, Gregorio Marañón Health Research Institute (IiSGM), General University Hospital Gregorio Marañón, Madrid, Spain
| |
Collapse
|
10
|
Gonçalves GS, Correa-Silva S, Zheng Y, Avelar I, Montenegro MM, Ferreira AEF, Bain V, Fink TT, Suguita P, Astley C, Lindoso L, Martins F, Matsuo OM, Ferreira JCOA, Firigato I, de Toledo Gonçalves F, Fernanda B Pereira M, Artur A da Silva C, Carneiro-Sampaio M, Marques HHS, Palmeira P. Circulating sTREM-1 as a predictive biomarker of pediatric multisystemic inflammatory syndrome (MIS-C). Cytokine 2023; 161:156084. [PMID: 36403563 PMCID: PMC9671781 DOI: 10.1016/j.cyto.2022.156084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/19/2022] [Accepted: 10/29/2022] [Indexed: 11/19/2022]
Abstract
The exacerbation of the inflammatory response caused by SARS-CoV-2 in adults promotes the production of soluble mediators that could act as diagnostic and prognostic biomarkers for COVID-19. Among the potential biomarkers, the soluble triggering receptor expressed on myeloid cell-1 (sTREM-1) has been described as a predictor of inflammation severity. The aim was to evaluate sTREM-1 and cytokine serum concentrations in pediatric patients during the acute and convalescent phases of COVID-19. This was a prospective study that included 53 children/adolescents with acute COVID-19 (Acute-CoV group); 54 who recovered from COVID-19 (Post-CoV group) and 54 controls (Control group). Preexisting chronic conditions were present in the three groups, which were defined as follows: immunological diseases, neurological disorders, and renal and hepatic failures. The three groups were matched by age, sex, and similar preexisting chronic conditions. No differences in sTREM-1 levels were detected among the groups or when the groups were separately analyzed by preexisting chronic conditions. However, sTREM-1 analysis in the seven multisystemic inflammatory syndrome children (MIS-C) within the Acute-Cov group showed that sTREM-1 concentrations were higher in MIS-C vs non-MIS-C acute patients. Then, the receiver operating curve analysis (ROC) performed with MIS-C acute patients revealed a significant AUC of 0.870, and the sTREM-1 cutoff value of > 5781 pg/mL yielded a sensitivity of 71.4 % and a specificity of 91.3 % for disease severity, and patients with sTREM-1 levels above this cutoff presented an elevated risk for MIS-C development in 22.85-fold (OR = 22.85 [95 % CI 1.64-317.5], p = 0.02). The cytokine analyses in the acute phase revealed that IL-6, IL-8, and IL-10 concentrations were elevated regardless of whether the patient developed MIS-C, and those levels decreased in the convalescent phase, even when compared with controls. Spearman correlation analysis generated positive indexes between sTREM-1 and IL-12 and TNF-α concentrations, only within the Acute-CoV group. Our findings revealed that sTREM-1 in pediatric patients has good predictive accuracy as an early screening tool for surveillance of MIS-C cases, even in patients with chronic underlying conditions.
Collapse
Affiliation(s)
- Guilherme S Gonçalves
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Simone Correa-Silva
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Universidade Paulista, UNIP, Sao Paulo, SP, Brazil.
| | - Yingying Zheng
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Isabela Avelar
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Marília M Montenegro
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Arthur E F Ferreira
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Vera Bain
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Thais T Fink
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Priscila Suguita
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Camilla Astley
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Livia Lindoso
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Fernanda Martins
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Olivia M Matsuo
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Juliana C O A Ferreira
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Isabela Firigato
- Laboratorio de Imunohematologia e Hematologia Forense (LIM-40), Departamento de Medicina Legal, Bioética, Medicina do Trabalho e Medicina Física e Reabilitação, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Fernanda de Toledo Gonçalves
- Laboratorio de Imunohematologia e Hematologia Forense (LIM-40), Departamento de Medicina Legal, Bioética, Medicina do Trabalho e Medicina Física e Reabilitação, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Maria Fernanda B Pereira
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Clovis Artur A da Silva
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Magda Carneiro-Sampaio
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Heloisa H S Marques
- Departamento de Pediatria, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Patricia Palmeira
- Laboratorio de Pediatria Clinica (LIM-36), Departamento de Pediatria, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
11
|
Ponciano-Gómez A, Valle-Solis MI, Campos-Aguilar M, Jijón-Lorenzo R, Herrera-Cogco EDLC, Ramos-Alor R, Bazán-Mendez CI, Cervantes GAPG, Ávila-García R, Aguilar AG, Texale MGS, Tapia-Sánchez WD, Duarte-Martínez CL, Olivas-Quintero S, Sigrist-Flores SC, Gallardo-Ortíz IA, Villalobos-Molina R, Méndez-Cruz AR, Jimenez-Flores R, Santos-Argumedo L, Luna-Arias JP, Romero-Ramírez H, Rosales-García VH, Avendaño-Borromeo B. High baseline expression of IL-6 and IL-10 decreased CCR7 B cells in individuals with previous SARS-CoV-2 infection during BNT162b2 vaccination. Front Immunol 2022; 13:946770. [PMID: 36052060 PMCID: PMC9425053 DOI: 10.3389/fimmu.2022.946770] [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: 05/18/2022] [Accepted: 07/22/2022] [Indexed: 01/09/2023] Open
Abstract
The current pandemic generated by SARS-CoV-2 has led to mass vaccination with different biologics that have shown wide variations among human populations according to the origin and formulation of the vaccine. Studies evaluating the response in individuals with a natural infection before vaccination have been limited to antibody titer analysis and evaluating a few humoral and cellular response markers, showing a more rapid and intense humoral response than individuals without prior infection. However, the basis of these differences has not been explored in depth. In the present work, we analyzed a group of pro and anti-inflammatory cytokines, antibody titers, and cell populations in peripheral blood of individuals with previous SARS-CoV-2 infection using BNT162b2 biologic. Our results suggest that higher antibody concentration in individuals with an earlier disease could be generated by higher production of plasma cells to the detriment of the presence of memory B cells in the bloodstream, which could be related to the high baseline expression of cytokines (IL-6 and IL-10) before vaccination.
Collapse
Affiliation(s)
- Alberto Ponciano-Gómez
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, Mexico
| | - Martha Iris Valle-Solis
- Secretaría de Salud de Veracruz, Servicios de Salud de Veracruz, SESVER, Xalapa Veracruz, Mexico
| | - Myriam Campos-Aguilar
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, Mexico
| | - Rafael Jijón-Lorenzo
- Secretaría de Salud de Veracruz, Servicios de Salud de Veracruz, SESVER, Xalapa Veracruz, Mexico
| | | | - Roberto Ramos-Alor
- Secretaría de Salud de Veracruz, Servicios de Salud de Veracruz, SESVER, Xalapa Veracruz, Mexico
| | | | | | - Ricardo Ávila-García
- Secretaría de Salud de Veracruz, Servicios de Salud de Veracruz, SESVER, Xalapa Veracruz, Mexico
| | - Abdiel González Aguilar
- Secretaría de Salud de Veracruz, Servicios de Salud de Veracruz, SESVER, Xalapa Veracruz, Mexico
| | | | - Wilfrido David Tapia-Sánchez
- Laboratorio de Citometría de Flujo y Hematología, Diagnóstico Molecular de Leucemias y Terapia Celular (DILETEC), Gustavo A. Madero, Ciudad de Mexico, Mexico
| | - Carlos Leonardo Duarte-Martínez
- Laboratorio de Citometría de Flujo y Hematología, Diagnóstico Molecular de Leucemias y Terapia Celular (DILETEC), Gustavo A. Madero, Ciudad de Mexico, Mexico
| | - Sandra Olivas-Quintero
- Department of Health Sciences, Autonomus University of Occident, Culiacan, Sinaloa, Mexico
| | - Santiago Cristobal Sigrist-Flores
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, Mexico
| | - Itzell Alejandrina Gallardo-Ortíz
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, Mexico
| | - Rafael Villalobos-Molina
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, Mexico
| | - Adolfo Rene Méndez-Cruz
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, Mexico
| | - Rafael Jimenez-Flores
- Laboratorio de Inmunología, Unidad de Morfología y Función, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, Mexico
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico
| | - Juan Pedro Luna-Arias
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico
| | - Hector Romero-Ramírez
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico
| | - Victor Hugo Rosales-García
- Laboratorio de Citometría de Flujo y Hematología, Diagnóstico Molecular de Leucemias y Terapia Celular (DILETEC), Gustavo A. Madero, Ciudad de Mexico, Mexico
- Laboratorios Nacionales de Servicios Experimentales, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | | |
Collapse
|