1
|
Asghariazar V, Eterafi M, Matin S, Fouladi N, Abolhasani R, Falsafi M, Fathi A, Safarzadeh E. Evaluation of Dendritic Cell Subpopulations Frequency in COVID-19 Patients and their Correlation with Disease Severity. IRANIAN JOURNAL OF IMMUNOLOGY : IJI 2025; 22:70-82. [PMID: 40130356 DOI: 10.22034/iji.2025.104236.2887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Background COVID-19 (2019) clearly demonstrates an imbalanced immune response. Variations in the function and subtypes of dendritic cells (DCs) may have effects on immune responses in COVID-19 patients and contribute to immunopathology. Objective To assess the phenotype and frequency of Plasmacytoid dendritic cells (pDCs), Conventional DCs (cDCs), and double-positive DCs in COVID-19 patients admitted to the ICU and non-ICU compared to the healthy control group. Methods The study included 10 healthy individuals and 25 COVID-19 patients. In the second week of their illness, Peripheral blood mononuclear cells (PBMCs) were isolated from the patients and labeled with targeted antibodies for HLA-DR, CD123, and CD11c. The samples were then analyzed using flow cytometry. The COVID-19 patients were divided into two ICU and non-ICU groups and were closely monitored throughout the study. Results In comparison to healthy controls, COVID-19 patients exhibited a significantly lower pDCs ratio (P=0.04). Patients were categorized into two groups: (A) the ICU group (n=11; 44%) and (B) the non-ICU group (n=14; 56%). The frequency of pDC was significantly lower in ICU patients than in non-ICU patients (P<0.01). Although not statistically significant, ICU patients had a lower frequency of cDCs and double positive DCs compared to non-ICU patients. Additionally, a significant association between the age of COVID-19 patients and cDC levels was observed (p=0.049). Conclusion SARS-CoV-2 can evade attacks from the immune response by reducing the number of DCs and suppressing their function of DCs, ultimately resulting in weakened development of both innate and adaptive immunity.
Collapse
Affiliation(s)
- Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Eterafi
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Somaieh Matin
- Department of Internal Medicine, Emam Khomeini Hospital, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nasrin Fouladi
- School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
- Social Determinants of Health Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Rozita Abolhasani
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Monireh Falsafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Afshin Fathi
- Pediatric Hematology and Oncology Department, Ardabil University of Medical Science, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
2
|
Vu Manh TP, Gouin C, De Wolf J, Jouneau L, Pascale F, Bevilacqua C, Ar Gouilh M, Da Costa B, Chevalier C, Glorion M, Hannouche L, Urien C, Estephan J, Magnan A, Le Guen M, Marquant Q, Descamps D, Dalod M, Schwartz-Cornil I, Sage E. SARS-CoV2 infection in whole lung primarily targets macrophages that display subset-specific responses. Cell Mol Life Sci 2024; 81:351. [PMID: 39147987 PMCID: PMC11335275 DOI: 10.1007/s00018-024-05322-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 08/17/2024]
Abstract
Deciphering the initial steps of SARS-CoV-2 infection, that influence COVID-19 outcomes, is challenging because animal models do not always reproduce human biological processes and in vitro systems do not recapitulate the histoarchitecture and cellular composition of respiratory tissues. To address this, we developed an innovative ex vivo model of whole human lung infection with SARS-CoV-2, leveraging a lung transplantation technique. Through single-cell RNA-seq, we identified that alveolar and monocyte-derived macrophages (AMs and MoMacs) were initial targets of the virus. Exposure of isolated lung AMs, MoMacs, classical monocytes and non-classical monocytes (ncMos) to SARS-CoV-2 variants revealed that while all subsets responded, MoMacs produced higher levels of inflammatory cytokines than AMs, and ncMos contributed the least. A Wuhan lineage appeared to be more potent than a D614G virus, in a dose-dependent manner. Amidst the ambiguity in the literature regarding the initial SARS-CoV-2 cell target, our study reveals that AMs and MoMacs are dominant primary entry points for the virus, and suggests that their responses may conduct subsequent injury, depending on their abundance, the viral strain and dose. Interfering on virus interaction with lung macrophages should be considered in prophylactic strategies.
Collapse
Affiliation(s)
- Thien-Phong Vu Manh
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13009, Marseille, France.
| | - Carla Gouin
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Julien De Wolf
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| | - Luc Jouneau
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, UVSQ, BREED, 78350, Jouy-en-Josas, France
| | - Florentina Pascale
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| | - Claudia Bevilacqua
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Meriadeg Ar Gouilh
- Department of Virology, Univ Caen Normandie, Dynamicure INSERM UMR 1311, CHU Caen, 14000, Caen, France
| | - Bruno Da Costa
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | | | - Matthieu Glorion
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| | - Laurent Hannouche
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13009, Marseille, France
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Céline Urien
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Jérôme Estephan
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Antoine Magnan
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Pulmonology, Foch Hospital, 92150, Suresnes, France
| | - Morgan Le Guen
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Anesthesiology, Foch Hospital, 92150, Suresnes, France
| | - Quentin Marquant
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Pulmonology, Foch Hospital, 92150, Suresnes, France
- Delegation to Clinical Research and Innovation, Foch Hospital, 92150, Suresnes, France
| | - Delphyne Descamps
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Marc Dalod
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13009, Marseille, France
| | | | - Edouard Sage
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| |
Collapse
|
3
|
Bukreieva T, Kyryk V, Nikulina V, Svitina H, Vega A, Chybisov O, Shablii I, Mankovska O, Lobyntseva G, Nemtinov P, Skrypkina I, Shablii V. Dynamic changes in radiological parameters, immune cells, selected miRNAs, and cytokine levels in peripheral blood of patients with severe COVID‑19. Biomed Rep 2023; 18:33. [PMID: 37034572 PMCID: PMC10074022 DOI: 10.3892/br.2023.1615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
The present study aimed to investigate the dynamic changes in peripheral blood leucocyte subpopulations, cytokine and miRNA levels, and changes in computed tomography (CT) scores in patients with severe coronavirus disease 2019 (COVID-19) (n=14) and age-matched non-COVID-19 volunteers (n=17), which were included as a reference control group. All data were collected on the day of patient admission (day 0) and on the 7th, 14th and 28th days of follow-up while CT of the lungs was performed on weeks 2, 8, 24 and 48. On day 0, lymphopenia and leucopenia were detected in most patients with COVID-19, as well as an increase in the percentage of banded neutrophils, B cells, and CD4+ Treg cells, and a decrease in the content of PD-1low T cells, classical, plasmacytoid, and regulatory dendritic cells. On day 7, the percentage of T and natural killer cells decreased with a concurrent increase in B cells, but returned to the initial level after treatment discharge. The content of different T and dendritic cell subsets among CD45+ cells increased during two weeks and remained elevated, suggesting the activation of an adaptive immune response. The increase of PD-1-positive subpopulations of T and non-T cells and regulatory CD4 T cells in patients with COVID-19 during the observation period suggests the development of an inflammation control mechanism. The levels of interferon γ-induced protein 10 (IP-10), tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 decreased on day 7, but increased again on days 14 and 28. C-reactive protein and granulocyte colony-stimulating factor (G-CSF) levels decreased gradually throughout the observation period. The relative expression levels of microRNA (miR)-21-5p, miR-221-3p, miR-27a-3p, miR-146a-5p, miR-133a-3p, and miR-126-3p were significantly higher at the beginning of hospitalization compared to non-COVID-19 volunteers. The plasma levels of all miRs, except for miR-126-3p, normalized within one week of treatment. At week 48, CT scores were most prominently correlated with the content of lymphocytes, senescent memory T cells, CD127+ T cells and CD57+ T cells, and increased concentrations of G-CSF, IP-10, and macrophage inflammatory protein-1α.
Collapse
Affiliation(s)
- Tetiana Bukreieva
- Laboratory of Biosynthesis of Nucleic Acids, Department of Functional Genomics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
- Placenta Stem Cell Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine
| | - Vitalii Kyryk
- Laboratory of Cell and Tissue Cultures, Department of Cell and Tissue Technologies, State Institute of Genetic and Regenerative Medicine, National Academy of Medical Sciences of Ukraine, Kyiv 04114, Ukraine
- Laboratory of Pathophysiology and Immunology, D.F. Chebotarev State Institute of Gerontology of The National Academy of Medical Sciences of Ukraine, Kyiv 04114, Ukraine
| | - Viktoriia Nikulina
- Placenta Stem Cell Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine
| | - Hanna Svitina
- Laboratory of Biosynthesis of Nucleic Acids, Department of Functional Genomics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
- Placenta Stem Cell Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine
| | - Alyona Vega
- Department of Infectious Diseases, Shupyk National Healthcare University of Ukraine, Kyiv 04112, Ukraine
| | - Oleksii Chybisov
- Endoscopic Unit, CNE Kyiv City Clinical Hospital No. 4, Kyiv 03110, Ukraine
| | - Iuliia Shablii
- Laboratory of Biosynthesis of Nucleic Acids, Department of Functional Genomics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
| | - Oksana Mankovska
- Department of Molecular Oncogenetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
| | - Galyna Lobyntseva
- Placenta Stem Cell Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine
| | - Petro Nemtinov
- Placenta Stem Cell Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine
| | - Inessa Skrypkina
- Laboratory of Biosynthesis of Nucleic Acids, Department of Functional Genomics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
| | - Volodymyr Shablii
- Laboratory of Biosynthesis of Nucleic Acids, Department of Functional Genomics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv 03143, Ukraine
- Placenta Stem Cell Laboratory, Cryobank, Institute of Cell Therapy, Kyiv 03126, Ukraine
| |
Collapse
|
4
|
Nasrollahi H, Talepoor AG, Saleh Z, Eshkevar Vakili M, Heydarinezhad P, Karami N, Noroozi M, Meri S, Kalantar K. Immune responses in mildly versus critically ill COVID-19 patients. Front Immunol 2023; 14:1077236. [PMID: 36793739 PMCID: PMC9923185 DOI: 10.3389/fimmu.2023.1077236] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
The current coronavirus pandemic (COVID-19), caused by SARS-CoV-2, has had devastating effects on the global health and economic system. The cellular and molecular mediators of both the innate and adaptive immune systems are critical in controlling SARS-CoV-2 infections. However, dysregulated inflammatory responses and imbalanced adaptive immunity may contribute to tissue destruction and pathogenesis of the disease. Important mechanisms in severe forms of COVID-19 include overproduction of inflammatory cytokines, impairment of type I IFN response, overactivation of neutrophils and macrophages, decreased frequencies of DC cells, NK cells and ILCs, complement activation, lymphopenia, Th1 and Treg hypoactivation, Th2 and Th17 hyperactivation, as well as decreased clonal diversity and dysregulated B lymphocyte function. Given the relationship between disease severity and an imbalanced immune system, scientists have been led to manipulate the immune system as a therapeutic approach. For example, anti-cytokine, cell, and IVIG therapies have received attention in the treatment of severe COVID-19. In this review, the role of immunity in the development and progression of COVID-19 is discussed, focusing on molecular and cellular aspects of the immune system in mild vs. severe forms of the disease. Moreover, some immune- based therapeutic approaches to COVID-19 are being investigated. Understanding key processes involved in the disease progression is critical in developing therapeutic agents and optimizing related strategies.
Collapse
Affiliation(s)
- Hamid Nasrollahi
- Radio-Oncology Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Atefe Ghamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Saleh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Eshkevar Vakili
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paria Heydarinezhad
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Karami
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Noroozi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seppo Meri
- Department of Bacteriology and Immunology, University of Helsinki and Diagnostic Center of the Helsinki University Hospital, Helsinki, Finland
| | - Kurosh Kalantar
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
5
|
Abstract
The worldwide coronavirus disease 2019 pandemic was sparked by the severe acute respiratory syndrome caused by coronavirus 2 (SARS-CoV-2) that first surfaced in December 2019 (COVID-19). The effects of COVID-19 differ substantially not just between patients individually but also between populations with different ancestries. In humans, the human leukocyte antigen (HLA) system coordinates immune regulation. Since HLA molecules are a major component of antigen-presenting pathway, they play an important role in determining susceptibility to infectious disease. It is likely that differential susceptibility to SARS-CoV-2 infection and/or disease course in COVID-19 in different individuals could be influenced by the variations in the HLA genes which are associated with various immune responses to SARS-CoV-2. A growing number of studies have identified a connection between HLA variation and diverse COVID-19 outcomes. Here, we review research investigating the impact of HLA on individual responses to SARS-CoV-2 infection and/or progression, also discussing the significance of MHC-related immunological patterns and its use in vaccine design.
Collapse
Affiliation(s)
- Anshika Srivastava
- grid.266102.10000 0001 2297 6811University of California San Francisco, San Francisco, CA USA
| | - Jill A. Hollenbach
- grid.266102.10000 0001 2297 6811University of California San Francisco, San Francisco, CA USA
| |
Collapse
|
6
|
Gupta GS. The Lactate and the Lactate Dehydrogenase in Inflammatory Diseases and Major Risk Factors in COVID-19 Patients. Inflammation 2022; 45:2091-2123. [PMID: 35588340 PMCID: PMC9117991 DOI: 10.1007/s10753-022-01680-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/04/2022] [Accepted: 05/03/2022] [Indexed: 12/15/2022]
Abstract
Lactate dehydrogenase (LDH) is a terminating enzyme in the metabolic pathway of anaerobic glycolysis with end product of lactate from glucose. The lactate formation is crucial in the metabolism of glucose when oxygen is in inadequate supply. Lactate can also be formed and utilised by different cell types under fully aerobic conditions. Blood LDH is the marker enzyme, which predicts mortality in many conditions such as ARDS, serious COVID-19 and cancer patients. Lactate plays a critical role in normal physiology of humans including an energy source, a signaling molecule and a pH regulator. Depending on the pH, lactate exists as the protonated acidic form (lactic acid) at low pH or as sodium salt (sodium lactate) at basic pH. Lactate can affect the immune system and act as a signaling molecule, which can provide a "danger" signal for life. Several reports provide evidence that the serum lactate represents a chemical marker of severity of disease similar to LDH under inflammatory conditions. Since the mortality rate is much higher among COVID-19 patients, associated with high serum LDH, this article is aimed to review the LDH as a therapeutic target and lactate as potential marker for monitoring treatment response of inflammatory diseases. Finally, the review summarises various LDH inhibitors, which offer potential applications as therapeutic agents for inflammatory diseases, associated with high blood LDH. Both blood LDH and blood lactate are suggested as risk factors for the mortality of patients in serious inflammatory diseases.
Collapse
Affiliation(s)
- G S Gupta
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
7
|
Milenkovic D, Rodriguez‐Mateos A, Lucosz M, Istas G, Declerck K, Sansone R, Deenen R, Köhrer K, Corral‐Jara KF, Altschmied J, Haendeler J, Kelm M, Berghe WV, Heiss C. Flavanol Consumption in Healthy Men Preserves Integrity of Immunological-Endothelial Barrier Cell Functions: Nutri(epi)genomic Analysis. Mol Nutr Food Res 2022; 66:e2100991. [PMID: 35094491 PMCID: PMC9787825 DOI: 10.1002/mnfr.202100991] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/16/2022] [Indexed: 12/30/2022]
Abstract
SCOPE While cocoa flavanol (CF) consumption improves cardiovascular risk biomarkers, molecular mechanisms underlying their protective effects are not understood. OBJECTIVE To investigate nutri(epi)genomic effects of CF and identify regulatory networks potential mediating vascular health benefits. METHODS AND RESULTS Twenty healthy middle-aged men consume CF (bi-daily 450 mg) or control drinks for 1 month. Microarray analysis identifies 2235 differentially expressed genes (DEG) involved in processes regulating immune response, cell adhesion, or cytoskeleton organization. Distinct patterns of DEG correlate with CF-related changes in endothelial function, arterial stiffness, and blood pressure. DEG profile negatively correlates with expression profiles of cardiovascular disease patients. CF modulated DNA methylation profile of genes implicates in cell adhesion, actin cytoskeleton organization, or cell signaling. In silico docking analyses indicate that CF metabolites have the potential of binding to cell signaling proteins and transcription factors. Incubation of plasma obtained after CF consumption decrease monocyte to endothelial adhesion and dose-dependently increase nitric oxide-dependent chemotaxis of circulating angiogenic cells further validating the biological functions of CF metabolites. CONCLUSION In healthy humans, CF consumption may mediate vascular protective effects by modulating gene expression and DNA methylation towards a cardiovascular protective effect, in agreement with clinical results, by preserving integrity of immunological-endothelial barrier functions.
Collapse
Affiliation(s)
- Dragan Milenkovic
- Department of NutritionUniversity of California DavisDavisCA95616USA
- INRAEUNHUniversité Clermont AuvergneClermont‐FerrandF‐63000France
| | - Ana Rodriguez‐Mateos
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
- Department of Nutritional SciencesSchool of Life Course and Population SciencesFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Margarete Lucosz
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - Geoffrey Istas
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
- Department of Nutritional SciencesSchool of Life Course and Population SciencesFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Ken Declerck
- PPESDepartment of Biomedical SciencesUniversity of Antwerp (UA)WilrijkBelgium
| | - Roberto Sansone
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - René Deenen
- Biological and Medical Research Center (BMFZ)Heinrich Heine UniversityDüsseldorfGermany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ)Heinrich Heine UniversityDüsseldorfGermany
| | | | - Joachim Altschmied
- Environmentally‐induced Cardiovascular DegenerationClinical Chemistry and Laboratory DiagnosticsMedical FacultyUniversity Hospital and Heinrich‐Heine UniversityDüsseldorfGermany
- IUF‐Leibniz Research Institute for Environmental MedicineDüsseldorfGermany
| | - Judith Haendeler
- Environmentally‐induced Cardiovascular DegenerationClinical Chemistry and Laboratory DiagnosticsMedical FacultyUniversity Hospital and Heinrich‐Heine UniversityDüsseldorfGermany
| | - Malte Kelm
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - Wim Vanden Berghe
- PPESDepartment of Biomedical SciencesUniversity of Antwerp (UA)WilrijkBelgium
| | - Christian Heiss
- Division of CardiologyPulmonology, and Vascular MedicineMedical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
- Clinical Medicine SectionDepartment of Clinical and Experimental MedicineFaculty of Health and Medical SciencesUniversity of SurreyGuildfordUK
- Department of Vascular MedicineSurrey and Sussex NHS Healthcare TrustEast Surrey HospitalRedhillUK
| |
Collapse
|
8
|
Budhraja A, Basu A, Gheware A, Abhilash D, Rajagopala S, Pakala S, Sumit M, Ray A, Subramaniam A, Mathur P, Nambirajan A, Kumar S, Gupta R, Wig N, Trikha A, Guleria R, Sarkar C, Gupta I, Jain D. Molecular signature of postmortem lung tissue from COVID-19 patients suggests distinct trajectories driving mortality. Dis Model Mech 2022; 15:dmm049572. [PMID: 35438176 PMCID: PMC9194484 DOI: 10.1242/dmm.049572] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/07/2022] [Indexed: 12/19/2022] Open
Abstract
To elucidate the molecular mechanisms that manifest lung abnormalities during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, we performed whole-transcriptome sequencing of lung autopsies from 31 patients with severe COVID-19 and ten uninfected controls. Using metatranscriptomics, we identified the existence of two distinct molecular signatures of lethal COVID-19. The dominant 'classical' signature (n=23) showed upregulation of the unfolded protein response, steroid biosynthesis and complement activation, supported by massive metabolic reprogramming leading to characteristic lung damage. The rarer signature (n=8) that potentially represents 'cytokine release syndrome' (CRS) showed upregulation of cytokines such as IL1 and CCL19, but absence of complement activation. We found that a majority of patients cleared SARS-CoV-2 infection, but they suffered from acute dysbiosis with characteristic enrichment of opportunistic pathogens such as Staphylococcus cohnii in 'classical' patients and Pasteurella multocida in CRS patients. Our results suggest two distinct models of lung pathology in severe COVID-19 patients, which can be identified through complement activation, presence of specific cytokines and characteristic microbiome. These findings can be used to design personalized therapy using in silico identified drug molecules or in mitigating specific secondary infections.
Collapse
Affiliation(s)
- Anshul Budhraja
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Anubhav Basu
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Atish Gheware
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Dasari Abhilash
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Seesandra Rajagopala
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Suman Pakala
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Madhuresh Sumit
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Animesh Ray
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Arulselvi Subramaniam
- Department of Laboratory Medicine, JPNATC, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Purva Mathur
- Department of Laboratory Medicine, JPNATC, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Aruna Nambirajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sachin Kumar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Ritu Gupta
- Laboratory Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital (IRCH), All India Institute of Medical Sciences, New Delhi 110029, India
| | - Naveet Wig
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Anjan Trikha
- Department of Anaesthesiology, Critical Care and Pain Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Randeep Guleria
- Department of Pulmonary Medicine and Sleep Disorders, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Ishaan Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| |
Collapse
|
9
|
Matsunaga A, Tsuzuki S, Morioka S, Ohmagari N, Ishizaka Y. Long COVID: current status in Japan and knowledge about its molecular background. Glob Health Med 2022; 4:83-93. [PMID: 35586759 PMCID: PMC9066464 DOI: 10.35772/ghm.2022.01013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/23/2022] [Accepted: 04/04/2022] [Indexed: 06/15/2023]
Abstract
Even after recovering from coronavirus disease 2019 (COVID-19), patients can experience prolonged complaints, referred to as "long COVID". Similar to reports in Caucasians, a follow-up study in Japan revealed that fatigue, dyspnea, cough, anosmia/dysgeusia, and dyssomnia are common symptoms. Although the precise mode of long COVID remains elusive, multiple etiologies such as direct organ damage by infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), autoimmunity, prolonged inflammatory reactions, and psychiatric impairment seem to be involved. Notably, SARS-CoV-2 is neurotropic, and viral RNA and proteins are continuously detectable in multiple organs, including the brain. Viral proteins exert a number of different toxic effects on cells, suggesting that persistent infection is a key element for understanding long COVID. Here, we first reviewed the current status of long COVID in Japan, and then summarized literature that help us understand the molecular background of the symptoms. Finally, we discuss the feasibility of vaccination as a treatment for patients with long COVID.
Collapse
Affiliation(s)
- Akihiro Matsunaga
- Department of Intractable Diseases, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinya Tsuzuki
- AMR Clinical Reference Center, National Center for Global Health and Medicine Hospital, Tokyo, Japan
| | - Shinichiro Morioka
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yukihito Ishizaka
- Department of Intractable Diseases, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Programmed cell death: the pathways to severe COVID-19? Biochem J 2022; 479:609-628. [PMID: 35244141 PMCID: PMC9022977 DOI: 10.1042/bcj20210602] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023]
Abstract
Two years after the emergence of SARS-CoV-2, our understanding of COVID-19 disease pathogenesis is still incomplete. Despite unprecedented global collaborative scientific efforts and rapid vaccine development, an uneven vaccine roll-out and the emergence of novel variants of concern such as omicron underscore the critical importance of identifying the mechanisms that contribute to this disease. Overt inflammation and cell death have been proposed to be central drivers of severe pathology in COVID-19 patients and their pathways and molecular components therefore present promising targets for host-directed therapeutics. In our review, we summarize the current knowledge on the role and impact of diverse programmed cell death (PCD) pathways on COVID-19 disease. We dissect the complex connection of cell death and inflammatory signaling at the cellular and molecular level and identify a number of critical questions that remain to be addressed. We provide rationale for targeting of cell death as potential COVID-19 treatment and provide an overview of current therapeutics that could potentially enter clinical trials in the near future.
Collapse
|
11
|
Abstract
Considerable research effort has been made worldwide to decipher the immune response triggered upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, identify the drivers of severe and fatal COVID-19, and understand what leads to the prolongation of symptoms after disease resolution. We review the results of almost 2 years of COVID-19 immunology research and discuss definitive findings and remaining questions regarding our understanding of COVID-19 pathophysiology. We discuss emerging understanding of differences in immune responses seen in those with and without Long Covid syndrome, also known as post-acute sequelae of SARS-CoV-2. We hope that the knowledge gained from this COVID-19 research will be applied in studies of inflammatory processes involved in critical and chronic illnesses, which remain a major unmet need.
Collapse
Affiliation(s)
- Miriam Merad
- Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Catherine A Blish
- Department of Medicine and Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Federica Sallusto
- Institute of Microbiology, ETH Zürich, 8093 Zürich, Switzerland.,Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
12
|
Yıldırım SK, Erbağcı E, Demirel Öğüt N. Evaluation of patients with telogen effluvium during the pandemic: May the monocytesberesponsible for post COVID-19 telogen effluvium? J Cosmet Dermatol 2022; 21:1809-1815. [PMID: 35201647 PMCID: PMC9115203 DOI: 10.1111/jocd.14883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/18/2022] [Indexed: 11/27/2022]
Abstract
Introduction Telogen effluvium (TE) is one of the causes of non‐scarring hair loss that occurred commonly 2–3 months after a triggering factor. It was reported that the incidence of TE increased during the COVID‐19 (coronavirus disease 2019) pandemic. However, to date, there is no study evaluating the status of COVID‐19 before the onset of hair loss in patients with TE. The aim of this study is to evaluate the patients with TE whether they had COVID‐19 or not before the onset of their hair loss and to compare the demographic and clinical characteristics and laboratory parameters of those with and without a history of COVID‐19. Method We conducted an observational cohort study of TE patients. The diagnosis of TE depended on anamnesis and physical examination of the patients. Also, hair pull test was performed. Demographic data and the results of COVID‐19 real‐time polymerase chain reaction (RT‐PCR) were recorded from the electronic medical records. Results Totally, 181 patients with TE were included in the study. Sixty‐four of patients (35.4%) had been diagnosed with COVID‐19 before the hair loss started. The median duration of development of hair loss was 2 months (range 1–11 months, IQR 3) after COVID‐19 diagnosis. In this group, 87.5% of patients (n = 56) had acute TE and 12.5% of patients (n = 8) had chronic TE. The rate of acute TE and the use of vitamin supplements were ignificantly higher (p < 0.001 and p = 0.027, respectively) and the monocyte count in peripheral blood was lower (p = 0.041) in the group diagnosed with COVID‐19. Discussion and Conclusion It was stated that monocytes and macrophages infected by SARS‐CoV‐2 can produce pro‐inflammatory cytokines that play a crucial role in the development of COVID‐19‐related complications. Also, it was suggested that the number of monocytes tends to be lower in the late recovery stage. The lower monocyte count in patients with a history of COVID‐19 in our study may be related to evaluating the patients in the late period of recovery and the migration of circulating monocytes to hair follicles. The history of COVID‐19 must be questioned in patients with TE. It should be kept in mind that hair loss that develops after COVID‐19 may be presented as chronic TE form too. The exact mechanisms of hair loss induced by COVID‐19 are not fully explained; the roles of monocytes on the hair follicles may be one of the responsible mechanisms.
Collapse
Affiliation(s)
- Sema Koç Yıldırım
- Department of Dermatology and Venereology, Uşak University Training and Research Hospital, Uşak, Turkey
| | - Ece Erbağcı
- Department of Dermatology and Venereology, Uşak University Training and Research Hospital, Uşak, Turkey
| | - Neslihan Demirel Öğüt
- Department of Dermatology and Venereology, Uşak University Training and Research Hospital, Uşak, Turkey
| |
Collapse
|
13
|
Salazar F, Bignell E, Brown GD, Cook PC, Warris A. Pathogenesis of Respiratory Viral and Fungal Coinfections. Clin Microbiol Rev 2022; 35:e0009421. [PMID: 34788127 PMCID: PMC8597983 DOI: 10.1128/cmr.00094-21] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Individuals suffering from severe viral respiratory tract infections have recently emerged as "at risk" groups for developing invasive fungal infections. Influenza virus is one of the most common causes of acute lower respiratory tract infections worldwide. Fungal infections complicating influenza pneumonia are associated with increased disease severity and mortality, with invasive pulmonary aspergillosis being the most common manifestation. Strikingly, similar observations have been made during the current coronavirus disease 2019 (COVID-19) pandemic. The copathogenesis of respiratory viral and fungal coinfections is complex and involves a dynamic interplay between the host immune defenses and the virulence of the microbes involved that often results in failure to return to homeostasis. In this review, we discuss the main mechanisms underlying susceptibility to invasive fungal disease following respiratory viral infections. A comprehensive understanding of these interactions will aid the development of therapeutic modalities against newly identified targets to prevent and treat these emerging coinfections.
Collapse
Affiliation(s)
- Fabián Salazar
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Elaine Bignell
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Gordon D. Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Peter C. Cook
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
14
|
Ramírez-de-Arellano A, Gutiérrez-Franco J, Sierra-Diaz E, Pereira-Suárez AL. The role of estradiol in the immune response against COVID-19. Hormones (Athens) 2021; 20:657-667. [PMID: 34142358 PMCID: PMC8210971 DOI: 10.1007/s42000-021-00300-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the pathogen agent causing coronavirus disease (COVID)-19, which was declared a global pandemic in 2020. The spike protein of this virus and the angiotensin-converter enzyme (ACE)-2 in host cells in humans play a vital role in infection and in COVID-19 pathogenesis. Estradiol is known to modulate the actions of immune cells, and, therefore, the antiviral mechanisms of these cells could also be modified by this hormone stimulus. Even though estradiol is not considered a protective factor, evidence shows that women with high levels of this hormone have a lower risk of developing severe symptoms and an even a lower incidence of death. Understanding the mechanism of action of estradiol with regard to viral infections and COVID-19 is essential for the improvement of therapeutic strategies. This review aims to describe the effects that estradiol exerts on immune cells during viral infections and COVID-19.
Collapse
Affiliation(s)
- Adrián Ramírez-de-Arellano
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Guadalajara, Jalisco, 44 340, México
| | - Jorge Gutiérrez-Franco
- Unidad Académica de Ciencias Químico Biológicas Y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, 63 000, México
| | - Erick Sierra-Diaz
- Departamento de Salud Pública, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, División de Epidemiología, Unidad Medica de Alta Especialidad, Hospital de Especialidades, Centro Médico Nacional de Occidente, Guadalajara, Jalisco, 44340, México
| | - Ana Laura Pereira-Suárez
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Guadalajara, Jalisco, 44 340, México.
- Departamento de Microbiología Y Patología, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP, 44340, Guadalajara, Jalisco, México.
| |
Collapse
|
15
|
Bose P, Sunita P, Pattanayak SP. Molecular Insights into the Crosstalk Between Immune Inflammation Nexus and SARS-CoV-2 Virus. Curr Microbiol 2021; 78:3813-3828. [PMID: 34550435 PMCID: PMC8456397 DOI: 10.1007/s00284-021-02657-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023]
Abstract
COVID-19, a type of viral pneumonia caused by severe acute respiratory syndrome coronavirus 2 has challenged the world as global pandemic. It has marked the identification of third generation of extremely pathogenic zoonotic coronaviruses of twenty-first century posing threat to humans and mainly targeting the lower respiratory tract. In this review, we focused on not only the structure and virology of SARS-COV-2 but have discussed in detail the molecular immunopathogenesis of this novel virus highlighting its interaction with immune system and the role of compromised or dysregulated immune response towards disease severity. We attempted to correlate the crosstalk between unregulated inflammatory outcomes with disrupted host immunity which may play a potential role towards fatal acute respiratory distress syndrome that claims to be life-threatening in COVID-19. Exploration and investigation of molecular host-virus interactions will provide a better understanding on the mechanism of fatal COVID-19 infection and also enlighten the escape routes from the same.
Collapse
Affiliation(s)
- Pritha Bose
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Priyashree Sunita
- Government Pharmacy Institute, Govt. of Jharkhand, Bariatu, Ranchi, Jharkhand, 834009, India
| | - Shakti P Pattanayak
- Department of Pharmacy, School of Health Sciences, Central University of South Bihar, Govt. of India, Gaya, 824236, India.
| |
Collapse
|
16
|
Vázquez-Jiménez A, Avila-Ponce De León UE, Matadamas-Guzman M, Muciño-Olmos EA, Martínez-López YE, Escobedo-Tapia T, Resendis-Antonio O. On Deep Landscape Exploration of COVID-19 Patients Cells and Severity Markers. Front Immunol 2021; 12:705646. [PMID: 34603282 PMCID: PMC8481922 DOI: 10.3389/fimmu.2021.705646] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
COVID-19 is a disease with a spectrum of clinical responses ranging from moderate to critical. To study and control its effects, a large number of researchers are focused on two substantial aims. On the one hand, the discovery of diverse biomarkers to classify and potentially anticipate the disease severity of patients. These biomarkers could serve as a medical criterion to prioritize attention to those patients with higher prone to severe responses. On the other hand, understanding how the immune system orchestrates its responses in this spectrum of disease severities is a fundamental issue required to design new and optimized therapeutic strategies. In this work, using single-cell RNAseq of bronchoalveolar lavage fluid of nine patients with COVID-19 and three healthy controls, we contribute to both aspects. First, we presented computational supervised machine-learning models with high accuracy in classifying the disease severity (moderate and severe) in patients with COVID-19 starting from single-cell data from bronchoalveolar lavage fluid. Second, we identified regulatory mechanisms from the heterogeneous cell populations in the lungs microenvironment that correlated with different clinical responses. Given the results, patients with moderate COVID-19 symptoms showed an activation/inactivation profile for their analyzed cells leading to a sequential and innocuous immune response. In comparison, severe patients might be promoting cytotoxic and pro-inflammatory responses in a systemic fashion involving epithelial and immune cells without the possibility to develop viral clearance and immune memory. Consequently, we present an in-depth landscape analysis of how transcriptional factors and pathways from these heterogeneous populations can regulate their expression to promote or restrain an effective immune response directly linked to the patients prognosis.
Collapse
Affiliation(s)
- Aarón Vázquez-Jiménez
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Ugo Enrique Avila-Ponce De León
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biológicas, UNAM, Mexico City, Mexico
| | - Meztli Matadamas-Guzman
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, UNAM, Mexico City, Mexico
| | - Erick Andrés Muciño-Olmos
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, UNAM, Mexico City, Mexico
| | - Yoscelina E. Martínez-López
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Doctorado en Ciencias Médicas y de la Salud, UNAM, Mexico City, Mexico
| | - Thelma Escobedo-Tapia
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Programa de Maestría y Doctorado en Ciencias Bioquímicas, UNAM, Mexico City, Mexico
| | - Osbaldo Resendis-Antonio
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Coordinación de la Investigación Científica - Red de Apoyo a la Investigación, UNAM, Mexico City, Mexico
| |
Collapse
|
17
|
Niles MA, Gogesch P, Kronhart S, Ortega Iannazzo S, Kochs G, Waibler Z, Anzaghe M. Macrophages and Dendritic Cells Are Not the Major Source of Pro-Inflammatory Cytokines Upon SARS-CoV-2 Infection. Front Immunol 2021; 12:647824. [PMID: 34122407 PMCID: PMC8187925 DOI: 10.3389/fimmu.2021.647824] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
The exact role of innate immune cells upon infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and their contribution to the formation of the corona virus-induced disease (COVID)-19 associated cytokine storm is not yet fully understood. We show that human in vitro differentiated myeloid dendritic cells (mDC) as well as M1 and M2 macrophages are susceptible to infection with SARS-CoV-2 but are not productively infected. Furthermore, infected mDC, M1-, and M2 macrophages show only slight changes in their activation status. Surprisingly, none of the infected innate immune cells produced the pro-inflammatory cytokines interleukin (IL)-6, tumor necrosis factor (TNF)-α, or interferon (IFN)-α. Moreover, even in co-infection experiments using different stimuli, as well as non-influenza (non-flu) or influenza A (flu) viruses, only very minor IL-6 production was induced. In summary, we conclude that mDC and macrophages are unlikely the source of the first wave of cytokines upon infection with SARS-CoV-2.
Collapse
Affiliation(s)
- Marc A. Niles
- Section “Product Testing of Immunological Biomedicines”, Paul-Ehrlich-Institut, Langen, Germany
| | - Patricia Gogesch
- Section “Product Testing of Immunological Biomedicines”, Paul-Ehrlich-Institut, Langen, Germany
| | - Stefanie Kronhart
- Section “Product Testing of Immunological Biomedicines”, Paul-Ehrlich-Institut, Langen, Germany
| | - Samira Ortega Iannazzo
- Section “Product Testing of Immunological Biomedicines”, Paul-Ehrlich-Institut, Langen, Germany
| | - Georg Kochs
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Zoe Waibler
- Section “Product Testing of Immunological Biomedicines”, Paul-Ehrlich-Institut, Langen, Germany
| | - Martina Anzaghe
- Section “Product Testing of Immunological Biomedicines”, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
18
|
Wang W, Lei W, Jiang L, Gao S, Hu S, Zhao ZG, Niu CY, Zhao ZA. Therapeutic mechanisms of mesenchymal stem cells in acute respiratory distress syndrome reveal potentials for Covid-19 treatment. J Transl Med 2021; 19:198. [PMID: 33971907 PMCID: PMC8107778 DOI: 10.1186/s12967-021-02862-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/30/2021] [Indexed: 02/07/2023] Open
Abstract
The mortality rate of critically ill patients with acute respiratory distress syndrome (ARDS) is 30.9% to 46.1%. The emergence of the coronavirus disease 2019 (Covid-19) has become a global issue with raising dire concerns. Patients with severe Covid-19 may progress toward ARDS. Mesenchymal stem cells (MSCs) can be derived from bone marrow, umbilical cord, adipose tissue and so on. The easy accessibility and low immunogenicity enable MSCs for allogeneic administration, and thus they were widely used in animal and clinical studies. Accumulating evidence suggests that mesenchymal stem cell infusion can ameliorate ARDS. However, the underlying mechanisms of MSCs need to be discussed. Recent studies showed MSCs can modulate immune/inflammatory cells, attenuate endoplasmic reticulum stress, and inhibit pulmonary fibrosis. The paracrine cytokines and exosomes may account for these beneficial effects. In this review, we summarize the therapeutic mechanisms of MSCs in ARDS, analyzed the most recent animal experiments and Covid-19 clinical trial results, discussed the adverse effects and prospects in the recent studies, and highlight the potential roles of MSC therapy for Covid-19 patients with ARDS.
Collapse
Affiliation(s)
- Wendi Wang
- Institute of Microcirculation, Hebei North University, 11 Diamond South-road, Keji Building, Room 213, Zhangjiakou, 075000, Hebei, China.,Department of Pathophysiology of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China.,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Lina Jiang
- Institute of Microcirculation, Hebei North University, 11 Diamond South-road, Keji Building, Room 213, Zhangjiakou, 075000, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China
| | - Siqi Gao
- Institute of Microcirculation, Hebei North University, 11 Diamond South-road, Keji Building, Room 213, Zhangjiakou, 075000, Hebei, China.,Department of Pathophysiology of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China.,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China.,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Zi-Gang Zhao
- Institute of Microcirculation, Hebei North University, 11 Diamond South-road, Keji Building, Room 213, Zhangjiakou, 075000, Hebei, China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China. .,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China.
| | - Chun-Yu Niu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China. .,Basic Medical College, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
| | - Zhen-Ao Zhao
- Institute of Microcirculation, Hebei North University, 11 Diamond South-road, Keji Building, Room 213, Zhangjiakou, 075000, Hebei, China. .,Department of Pathophysiology of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050017, Hebei, China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Zhangjiakou, 075000, Hebei, China. .,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, 075000, Hebei, China.
| |
Collapse
|
19
|
Degauque N, Haziot A, Brouard S, Mooney N. Endothelial cell, myeloid, and adaptive immune responses in SARS-CoV-2 infection. FASEB J 2021; 35:e21577. [PMID: 33831263 PMCID: PMC8250117 DOI: 10.1096/fj.202100024r] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is an emerging respiratory pathogen that has rapidly spread in human populations. Severe forms of infection associate cytokine release syndrome and acute lung injury due to hyperinflammatory responses even though virus clearance is achieved. Key components of inflammation include immune cell recruitment in infected tissues, a step which is under the control of endothelial cells. Here, we review endothelial cell responses in inflammation and infection due to SARS-CoV-2 together with phenotypic and functional alterations of monocytes, T and B lymphocytes with which they interact. We surmise that endothelial cells function as an integrative and active platform for the various cells recruited, where fine tuning of immune responses takes place and which provides opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Nicolas Degauque
- Centre De Recherche En Transplantation Et ImmunologieUMR1064, INSERM, Université De NantesNantesFrance
- Institut De Transplantation Urologie Néphrologie (ITUN)CHU NantesNantesFrance
- Laboratoire d’ImmunologieCHU NantesNantesFrance
| | - Alain Haziot
- INSERM U976Institut de Recherche Saint LouisParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisParisFrance
| | - Sophie Brouard
- Centre De Recherche En Transplantation Et ImmunologieUMR1064, INSERM, Université De NantesNantesFrance
- Institut De Transplantation Urologie Néphrologie (ITUN)CHU NantesNantesFrance
- Laboratoire d’ImmunologieCHU NantesNantesFrance
| | - Nuala Mooney
- INSERM U976Institut de Recherche Saint LouisParisFrance
- Institut de Recherche Saint‐LouisUniversité de ParisParisFrance
| |
Collapse
|
20
|
Saichi M, Ladjemi MZ, Korniotis S, Rousseau C, Ait Hamou Z, Massenet-Regad L, Amblard E, Noel F, Marie Y, Bouteiller D, Medvedovic J, Pène F, Soumelis V. Single-cell RNA sequencing of blood antigen-presenting cells in severe COVID-19 reveals multi-process defects in antiviral immunity. Nat Cell Biol 2021; 23:538-551. [PMID: 33972731 DOI: 10.1038/s41556-021-00681-2] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 04/06/2021] [Indexed: 02/03/2023]
Abstract
COVID-19 can lead to life-threatening respiratory failure, with increased inflammatory mediators and viral load. Here, we perform single-cell RNA-sequencing to establish a high-resolution map of blood antigen-presenting cells (APCs) in 15 patients with moderate or severe COVID-19 pneumonia, at day 1 and day 4 post admission to intensive care unit or pulmonology department, as well as in 4 healthy donors. We generated a unique dataset of 81,643 APCs, including monocytes and rare dendritic cell (DC) subsets. We uncovered multi-process defects in antiviral immune defence in specific APCs from patients with severe disease: (1) increased pro-apoptotic pathways in plasmacytoid DCs (pDCs, key effectors of antiviral immunity), (2) a decrease of the innate sensors TLR9 and DHX36 in pDCs and CLEC9a+ DCs, respectively, (3) downregulation of antiviral interferon-stimulated genes in monocyte subsets and (4) a decrease of major histocompatibility complex (MHC) class II-related genes and MHC class II transactivator activity in cDC1c+ DCs, suggesting viral inhibition of antigen presentation. These novel mechanisms may explain patient aggravation and suggest strategies to restore the defective immune defence.
Collapse
Affiliation(s)
| | - Maha Zohra Ladjemi
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, Paris, France
- Service de Médecine Intensive & Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris. Centre & Université de Paris, Paris, France
| | | | - Christophe Rousseau
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, Paris, France
| | - Zakaria Ait Hamou
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, Paris, France
- Service de Médecine Intensive & Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris. Centre & Université de Paris, Paris, France
| | - Lucile Massenet-Regad
- Université de Paris, INSERM U976, Paris, France
- Université Paris-Saclay, Saint-Aubin, France
| | - Elise Amblard
- Université de Paris, INSERM U976, Paris, France
- Université de Paris, Centre de Recherches Interdisciplinaires, Paris, France
| | | | - Yannick Marie
- Institut du Cerveau (ICM), Plateforme de Génotypage Séquençage, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie, Paris, France
| | - Delphine Bouteiller
- Institut du Cerveau (ICM), Plateforme de Génotypage Séquençage, Paris, France
| | | | - Frédéric Pène
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, Paris, France
- Service de Médecine Intensive & Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris. Centre & Université de Paris, Paris, France
| | - Vassili Soumelis
- Université de Paris, INSERM U976, Paris, France.
- AP-HP, Hôpital Saint-Louis, Laboratoire d'Immunologie-Histocompatibilité, Paris, France.
| |
Collapse
|
21
|
Teran-Navarro H, Salcines-Cuevas D, Calderon-Gonzalez R, Tobes R, Calvo-Montes J, Pérez-Del Molino Bernal IC, Yañez-Diaz S, Fresno M, Alvarez-Dominguez C. A Comparison Between Recombinant Listeria GAPDH Proteins and GAPDH Encoding mRNA Conjugated to Lipids as Cross-Reactive Vaccines for Listeria, Mycobacterium, and Streptococcus. Front Immunol 2021; 12:632304. [PMID: 33953709 PMCID: PMC8092121 DOI: 10.3389/fimmu.2021.632304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/15/2021] [Indexed: 12/29/2022] Open
Abstract
Cross-reactive vaccines recognize common molecular patterns in pathogens and are able to confer broad spectrum protection against different infections. Antigens common to pathogenic bacteria that induce broad immune responses, such as the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) of the genera Listeria, Mycobacterium, or Streptococcus, whose sequences present more than 95% homology at the N-terminal GAPDH1-22 peptide, are putative candidates for universal vaccines. Here, we explore vaccine formulations based on dendritic cells (DC) loaded with two molecular forms of Listeria monocytogenes GAPDH (LM-GAPDH), such as mRNA carriers or recombinant proteins, and compare them with the same molecular forms of three other antigens used in experimental vaccines, listeriolysin O of Listeria monocytogeness, Ag85A of Mycobacterium marinum, and pneumolysin of Streptococcus pneumoniae. DC loaded with LM-GAPDH recombinant proteins proved to be the safest and most immunogenic vaccine vectors, followed by mRNA encoding LM-GAPDH conjugated to lipid carriers. In addition, macrophages lacked sufficient safety as vaccines for all LM-GAPDH molecular forms. The ability of DC loaded with LM-GAPDH recombinant proteins to induce non-specific DC activation explains their adjuvant potency and their capacity to trigger strong CD4+ and CD8+ T cell responses explains their high immunogenicity. Moreover, their capacity to confer protection in vaccinated mice against challenges with L. monocytogenes, M. marinum, or S. pneumoniae validated their efficiency as cross-reactive vaccines. Cross-protection appears to involve the induction of high percentages of GAPDH1-22 specific CD4+ and CD8+ T cells stained for intracellular IFN-γ, and significant levels of peptide-specific antibodies in vaccinated mice. We concluded that DC vaccines loaded with L. monocytogenes GAPDH recombinant proteins are cross-reactive vaccines that seem to be valuable tools in adult vaccination against Listeria, Mycobacterium, and Streptococcus taxonomic groups.
Collapse
Affiliation(s)
| | - David Salcines-Cuevas
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
- Grupo de Oncología y Nanovacunas, Santander, Spain
| | - Ricardo Calderon-Gonzalez
- Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
- Grupo de Oncología y Nanovacunas, Santander, Spain
| | | | - Jorge Calvo-Montes
- Microbiology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | | | - Sonsoles Yañez-Diaz
- Dermatology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
- Facultad de Medicina, Universidad de Cantabria, Santander, Spain
| | - Manuel Fresno
- DIOMUNE S.L., Parque Científico de Madrid, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Alvarez-Dominguez
- Facultad de Educación y Facultad de Ciencias de la Salud, Universidad Internacional de La Rioja, Logroño, Spain
| |
Collapse
|
22
|
Li J, Liu HH, Yin XD, Li CC, Wang J. COVID-19 illness and autoimmune diseases: recent insights. Inflamm Res 2021; 70:407-428. [PMID: 33640999 PMCID: PMC7914392 DOI: 10.1007/s00011-021-01446-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/25/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The aim of this review is to explore whether patients with autoimmune diseases (AIDs) were at high risk of infection during the COVID-19 epidemic and how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic affected immune system. METHODS A systematic literature search was performed using the foreign databases (NCBI, web of science, EBSCO, ELSEVIER ScienceDirect) and Chinese databases (WanFang, CNKI (China National Knowledge Infrastructure), VIP, CBM) to locate all relevant publications (up to January 10, 2021). The search strategies used Medical Search Headings (MeSH) headings and keywords for "COVID-19" or "SARS-CoV-2" or "coronavirus" and "autoimmune disease". RESULTS This review evaluates the effect of SARS-CoV-2 on the immune system through ACE-2 receptor binding as the main pathway for cell attachment and invasion. It is speculated that SARS-COV-2 infection can activate lymphocytes and inflammatory response, which may play a role in the clinical onset of AIDs and also patients were treated with immunomodulatory drugs during COVID-19 outbreak. Preliminary studies suggested that the risk of developing severe forms of COVID-19 in patients with AIDs treated with immunomodulators or biologics might not increase. A large number of samples are needed for further verification, leading to an excessive immune response to external stimuli. CONCLUSION The relationship between autoimmune diseases and SARS-CoV-2 infection is complex. During the COVID-19 epidemic, individualized interventions for AIDs should be provided such as Internet-based service.
Collapse
Affiliation(s)
- Juan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, People's Republic of China
- Medical Data Processing Center of School of Public Health of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Hong-Hui Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, People's Republic of China
- Medical Data Processing Center of School of Public Health of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Xiao-Dong Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, People's Republic of China
- Medical Data Processing Center of School of Public Health of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Cheng-Cheng Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, People's Republic of China
- Medical Data Processing Center of School of Public Health of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Jing Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, People's Republic of China.
- Medical Data Processing Center of School of Public Health of Anhui Medical University, Anhui Medical University, Hefei, China.
| |
Collapse
|
23
|
AbdelMassih AF, Menshawey R, Hozaien R, Kamel A, Mishriky F, Husseiny RJ, Hanoura AM, Yacoub E, AlShehry N, Menshawey E, El-Husseiny N, Yasser R, Arsanyous M, Nathan L, Seyam M, Massoud D, Ali N, Kassim A, AmanAllah M, Elsayed R, Sheashaa H, Husseiny Y, Hassan NH, Badr K, Elkhateb A, Fouad V, Elfishawy M, Medhat O, Mustafa M, Khalil N, Elsayed R, Nada Y, Elshawarbi P, Abdelmoneim N, Gamal N, Messiha M, Ghazy M, Abdelfatah E, Nasry F, Gayed R, Eesa M, Luis M, Eskandar E, Yacoub S, Saud A, Rajab M, Abdelaziz M, Elgamal N, Jaber H, Tayssir S, Michael M, Sabry A, Shehata J, Abdelaziz R, Rateb S, El-Maghraby A, Mahjoub Y, Amr A, Mabrouk A, Kelada P, Ragab S, Eltaher B, Hassan Galal R, Aly OM, Aly T, AbdelHaleem R, ElShaarawy A, Mohamed O. The potential use of lactate blockers for the prevention of COVID-19 worst outcome, insights from exercise immunology. Med Hypotheses 2021; 148:110520. [PMID: 33561624 PMCID: PMC7840393 DOI: 10.1016/j.mehy.2021.110520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 12/16/2022]
Abstract
Following the decline in Physical Activity (PA) due to COVID-19 restrictions in the form of government mandated lockdowns and closures of public spaces, the modulatory effect of physical exercise on immunity is being heavily revisited. In an attempt to comprehend the wide discrepancy in patient response to COVID-19 and the factors that potentially modulate it, we summarize the findings relating PA to inflammation and immunity. A distinction is drawn between moderate intensity and high intensity physical exercise based on the high lactate production observed in the latter. We hypothesize that, the lactate production associated with high intensity anaerobic exercise is implicated in the modulation of several components of the innate and adaptive immunity. In this review, we also summarize these immunomodulatory effects of lactate. These include increasing serum IL-6 levels, the main mediator of cytokine storms, as well as affecting NK cells, Macrophages, Dendritic cells and cytotoxic T-lymphocytes. The implications of high lactate levels in athletic performance are highlighted where athletes should undergo endurance training to increase VO2 max and minimize lactate production. Tumor models of hypoxia were also reported where lactate levels are elevated leading to increased invasiveness and angiogenesis. Accordingly, the novel lactate blocking strategy employed in cancer treatment is evaluated for its potential benefit in COVID-19 in addition to the readily available beta-blockers as an antagonist to lactate. Finally, we suggest the diagnostic/prognostic purpose of the elevated lactate levels that can be determined through sweat lactate testing. It is the detrimental effect of lactate on immunity and its presence in sweat that qualify it to be used as a potential non-invasive marker of poor COVID-19 outcome.
Collapse
Affiliation(s)
- Antoine Fakhry AbdelMassih
- Pediatric Cardiology Unit, Pediatrics' Department, Faculty of Medicine, Cairo University, Egypt; Pediatric Cardio-Oncology Department, Children Cancer Hospital of Egypt (57357), Egypt.
| | - Rahma Menshawey
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Rafeef Hozaien
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Aya Kamel
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Fady Mishriky
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Reem J Husseiny
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | | | - Elaria Yacoub
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Nada AlShehry
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Esraa Menshawey
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Nadine El-Husseiny
- Faculty of Dentistry, Cairo University, Egypt; Pixagon Graphic Design Agency, Cairo, Egypt
| | - Reem Yasser
- Department of Pharmaceutical and Pharmacological Sciences, Faculty of Medicine, Padova University, Padova, Italy
| | - Mariem Arsanyous
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Lauren Nathan
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Mahmoud Seyam
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Doaa Massoud
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Nada Ali
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Assem Kassim
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Mostafa AmanAllah
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Rokaya Elsayed
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Hesham Sheashaa
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Yousef Husseiny
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, New Giza University, Egypt
| | - Nourhan Hatem Hassan
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Kirollos Badr
- Faculty of Pharmacy, Future University, Cairo, Egypt
| | - Amr Elkhateb
- Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Verina Fouad
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Mayada Elfishawy
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Omar Medhat
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Mai Mustafa
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Noha Khalil
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Rawan Elsayed
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Youssef Nada
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Passant Elshawarbi
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Noha Abdelmoneim
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Nada Gamal
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Mariam Messiha
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Marihan Ghazy
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Emmy Abdelfatah
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Febronia Nasry
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Ramy Gayed
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Marian Eesa
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Merna Luis
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Alexandria University, Egypt
| | - Estfana Eskandar
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Shenoda Yacoub
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Alaa Saud
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Maram Rajab
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Mariam Abdelaziz
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Nadine Elgamal
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Hutaf Jaber
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Sara Tayssir
- Pediatric Residency Program, Faculty of Medicine, Cairo University, Egypt
| | - Mark Michael
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Ahmed Sabry
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Joseph Shehata
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Rania Abdelaziz
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Sherry Rateb
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Ahmed El-Maghraby
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Yara Mahjoub
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Alaa Amr
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Amin Mabrouk
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Peter Kelada
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Shahd Ragab
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Basant Eltaher
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Ain Shams University, Egypt
| | - Rahma Hassan Galal
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Omnya Mahmoud Aly
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Taquwa Aly
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Rana AbdelHaleem
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Areeg ElShaarawy
- Research Accessibility Team, Student and Internship Research Program, Faculty of Medicine, Cairo University, Egypt
| | - Omnia Mohamed
- Sports Medicine, Faculty of Physiotherapy, Cairo University, Egypt
| |
Collapse
|
24
|
Zingaropoli MA, Nijhawan P, Carraro A, Pasculli P, Zuccalà P, Perri V, Marocco R, Kertusha B, Siccardi G, Del Borgo C, Curtolo A, Ajassa C, Iannetta M, Ciardi MR, Mastroianni CM, Lichtner M. Increased sCD163 and sCD14 Plasmatic Levels and Depletion of Peripheral Blood Pro-Inflammatory Monocytes, Myeloid and Plasmacytoid Dendritic Cells in Patients With Severe COVID-19 Pneumonia. Front Immunol 2021; 12:627548. [PMID: 33777012 PMCID: PMC7993197 DOI: 10.3389/fimmu.2021.627548] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
Background Emerging evidence argues that monocytes, circulating innate immune cells, are principal players in COVID-19 pneumonia. The study aimed to investigate the role of soluble (s)CD163 and sCD14 plasmatic levels in predicting disease severity and characterize peripheral blood monocytes and dendritic cells (DCs), in patients with COVID-19 pneumonia (COVID-19 subjects). Methods On admission, in COVID-19 subjects sCD163 and sCD14 plasmatic levels, and peripheral blood monocyte and DC subsets were compared to healthy donors (HDs). According to clinical outcome, COVID-19 subjects were divided into ARDS and non-ARDS groups. Results Compared to HDs, COVID-19 subjects showed higher sCD163 (p<0.0001) and sCD14 (p<0.0001) plasmatic levels. We observed higher sCD163 plasmatic levels in the ARDS group compared to the non-ARDS one (p=0.002). The cut-off for sCD163 plasmatic level greater than 2032 ng/ml was predictive of disease severity (AUC: 0.6786, p=0.0022; sensitivity 56.7% [CI: 44.1–68.4] specificity 73.8% [CI: 58.9–84.7]). Positive correlation between plasmatic levels of sCD163, LDH and IL-6 and between plasmatic levels of sCD14, D-dimer and ferritin were found. Compared to HDs, COVID-19 subjects showed lower percentages of non-classical (p=0.0012) and intermediate monocytes (p=0.0447), slanDCs (p<0.0001), myeloid DCs (mDCs, p<0.0001), and plasmacytoid DCs (pDCs, p=0.0014). Compared to the non-ARDS group, the ARDS group showed lower percentages of non-classical monocytes (p=0.0006), mDCs (p=0.0346), and pDCs (p=0.0492). Conclusions The increase in sCD163 and sCD14 plasmatic levels, observed on hospital admission in COVID-19 subjects, especially in those who developed ARDS, and the correlations of these monocyte/macrophage activation markers with typical inflammatory markers of COVID-19 pneumonia, underline their potential use to assess the risk of progression of the disease. In an early stage of the disease, the assessment of sCD163 plasmatic levels could have clinical utility in predicting the severity of COVID-19 pneumonia.
Collapse
Affiliation(s)
| | - Parni Nijhawan
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Anna Carraro
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Patrizia Pasculli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Paola Zuccalà
- Infectious Diseases Unit, SM Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Valentina Perri
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Raffaella Marocco
- Infectious Diseases Unit, SM Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Blerta Kertusha
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Guido Siccardi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Cosmo Del Borgo
- Infectious Diseases Unit, SM Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Ambrogio Curtolo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Camilla Ajassa
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Marco Iannetta
- Department of System Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | | | - Miriam Lichtner
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.,Infectious Diseases Unit, SM Goretti Hospital, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
25
|
Tomić S, Đokić J, Stevanović D, Ilić N, Gruden-Movsesijan A, Dinić M, Radojević D, Bekić M, Mitrović N, Tomašević R, Mikić D, Stojanović D, Čolić M. Reduced Expression of Autophagy Markers and Expansion of Myeloid-Derived Suppressor Cells Correlate With Poor T Cell Response in Severe COVID-19 Patients. Front Immunol 2021; 12:614599. [PMID: 33692788 PMCID: PMC7937809 DOI: 10.3389/fimmu.2021.614599] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Widespread coronavirus disease (COVID)-19 is causing pneumonia, respiratory and multiorgan failure in susceptible individuals. Dysregulated immune response marks severe COVID-19, but the immunological mechanisms driving COVID-19 pathogenesis are still largely unknown, which is hampering the development of efficient treatments. Here we analyzed ~140 parameters of cellular and humoral immune response in peripheral blood of 41 COVID-19 patients and 16 age/gender-matched healthy donors by flow-cytometry, quantitative PCR, western blot and ELISA, followed by integrated correlation analyses with ~30 common clinical and laboratory parameters. We found that lymphocytopenia in severe COVID-19 patients (n=20) strongly affects T, NK and NKT cells, but not B cells and antibody production. Unlike increased activation of ICOS-1+ CD4+ T cells in mild COVID-19 patients (n=21), T cells in severe patients showed impaired activation, low IFN-γ production and high functional exhaustion, which correlated with significantly down-regulated HLA-DR expression in monocytes, dendritic cells and B cells. The latter phenomenon was followed by lower interferon responsive factor (IRF)-8 and autophagy-related genes expressions, and the expansion of myeloid derived suppressor cells (MDSC). Intriguingly, PD-L1-, ILT-3-, and IDO-1-expressing monocytic MDSC were the dominant producers of IL-6 and IL-10, which correlated with the increased inflammation and accumulation of regulatory B and T cell subsets in severe COVID-19 patients. Overall, down-regulated IRF-8 and autophagy-related genes expression, and the expansion of MDSC subsets could play critical roles in dysregulating T cell response in COVID-19, which could have large implications in diagnostics and design of novel therapeutics for this disease.
Collapse
Affiliation(s)
- Sergej Tomić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Jelena Đokić
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Dejan Stevanović
- Clinical Hospital Center Zemun, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nataša Ilić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Alisa Gruden-Movsesijan
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Miroslav Dinić
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Dušan Radojević
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marina Bekić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Nebojša Mitrović
- Clinical Hospital Center Zemun, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ratko Tomašević
- Clinical Hospital Center Zemun, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragan Mikić
- Clinics for Infectious and Tropical Diseases, Military Medical Academy, Belgrade, Serbia
| | - Dragoš Stojanović
- Clinical Hospital Center Zemun, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Miodrag Čolić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
- Department for Medical Sciences, Serbian Academy of Sciences and Arts, Belgrade, Serbia
| |
Collapse
|
26
|
Neeland MR, Bannister S, Clifford V, Dohle K, Mulholland K, Sutton P, Curtis N, Steer AC, Burgner DP, Crawford NW, Tosif S, Saffery R. Innate cell profiles during the acute and convalescent phase of SARS-CoV-2 infection in children. Nat Commun 2021; 12:1084. [PMID: 33597531 PMCID: PMC7889848 DOI: 10.1038/s41467-021-21414-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022] Open
Abstract
Children have mild severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) confirmed disease (COVID-19) compared to adults and the immunological mechanisms underlying this difference remain unclear. Here, we report acute and convalescent innate immune responses in 48 children and 70 adults infected with, or exposed to, SARS-CoV-2. We find clinically mild SARS-CoV-2 infection in children is characterised by reduced circulating subsets of monocytes (classical, intermediate, non-classical), dendritic cells and natural killer cells during the acute phase. In contrast, SARS-CoV-2-infected adults show reduced proportions of non-classical monocytes only. We also observe increased proportions of CD63+ activated neutrophils during the acute phase to SARS-CoV-2 in infected children. Children and adults exposed to SARS-CoV-2 but negative on PCR testing display increased proportions of low-density neutrophils that we observe up to 7 weeks post exposure. This study characterises the innate immune response during SARS-CoV-2 infection and household exposure in children. Childhood infection with SARS CoV2 is associated with a milder course of infection but the immunopathogenesis of this remains unclear. Here the authors explore immunological differences in the innate immune system during acute and convalescent SARS CoV2 infection in the young.
Collapse
Affiliation(s)
- Melanie R Neeland
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia. .,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.
| | - Samantha Bannister
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Infectious Diseases Unit, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Vanessa Clifford
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Laboratory Services, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Kate Dohle
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Kim Mulholland
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Philip Sutton
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Nigel Curtis
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Infectious Diseases Unit, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Andrew C Steer
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Infectious Diseases Unit, The Royal Children's Hospital, Parkville, VIC, Australia
| | - David P Burgner
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Infectious Diseases Unit, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Nigel W Crawford
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Department of General Medicine, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Shidan Tosif
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.,Department of General Medicine, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Richard Saffery
- Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
27
|
Alamri A, Fisk D, Upreti D, Kung SKP. A Missing Link: Engagements of Dendritic Cells in the Pathogenesis of SARS-CoV-2 Infections. Int J Mol Sci 2021; 22:1118. [PMID: 33498725 PMCID: PMC7865603 DOI: 10.3390/ijms22031118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DC) connect the innate and adaptive arms of the immune system and carry out numerous roles that are significant in the context of viral disease. Their functions include the control of inflammatory responses, the promotion of tolerance, cross-presentation, immune cell recruitment and the production of antiviral cytokines. Based primarily on the available literature that characterizes the behaviour of many DC subsets during Severe acute respiratory syndrome (SARS) and coronavirus disease 2019 (COVID-19), we speculated possible mechanisms through which DC could contribute to COVID-19 immune responses, such as dissemination of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to lymph nodes, mounting dysfunctional inteferon responses and T cell immunity in patients. We highlighted gaps of knowledge in our understanding of DC in COVID-19 pathogenesis and discussed current pre-clinical development of therapies for COVID-19.
Collapse
Affiliation(s)
- Abdulaziz Alamri
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E OT5, Canada; (A.A.); (D.F.)
| | - Derek Fisk
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E OT5, Canada; (A.A.); (D.F.)
| | - Deepak Upreti
- Surgery, Faculty of Health Sciences, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada;
| | - Sam K. P. Kung
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E OT5, Canada; (A.A.); (D.F.)
| |
Collapse
|
28
|
Carsetti R, Zaffina S, Piano Mortari E, Terreri S, Corrente F, Capponi C, Palomba P, Mirabella M, Cascioli S, Palange P, Cuccaro I, Milito C, Zumla A, Maeurer M, Camisa V, Vinci MR, Santoro A, Cimini E, Marchioni L, Nicastri E, Palmieri F, Agrati C, Ippolito G, Porzio O, Concato C, Onetti Muda A, Raponi M, Quintarelli C, Quinti I, Locatelli F. Different Innate and Adaptive Immune Responses to SARS-CoV-2 Infection of Asymptomatic, Mild, and Severe Cases. Front Immunol 2020; 11:610300. [PMID: 33391280 PMCID: PMC7772470 DOI: 10.3389/fimmu.2020.610300] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is a novel coronavirus, not encountered before by humans. The wide spectrum of clinical expression of SARS-CoV-2 illness suggests that individual immune responses to SARS-CoV-2 play a crucial role in determining the clinical course after first infection. Immunological studies have focused on patients with moderate to severe disease, demonstrating excessive inflammation in tissues and organ damage. In order to understand the basis of the protective immune response in COVID-19, we performed a longitudinal follow-up, flow-cytometric and serological analysis of innate and adaptive immunity in 64 adults with a spectrum of clinical presentations: 28 healthy SARS-CoV-2-negative contacts of COVID-19 cases; 20 asymptomatic SARS-CoV-2-infected cases; eight patients with Mild COVID-19 disease and eight cases of Severe COVID-19 disease. Our data show that high frequency of NK cells and early and transient increase of specific IgA, IgM and, to a lower extent, IgG are associated with asymptomatic SARS-CoV-2 infection. By contrast, monocyte expansion and high and persistent levels of IgA and IgG, produced relatively late in the course of the infection, characterize severe disease. Modest increase of monocytes and different kinetics of antibodies are detected in mild COVID-19. The importance of innate NK cells and the short-lived antibody response of asymptomatic individuals and patients with mild disease suggest that only severe COVID-19 may result in protective memory established by the adaptive immune response.
Collapse
Affiliation(s)
- Rita Carsetti
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Salvatore Zaffina
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Eva Piano Mortari
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Sara Terreri
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Francesco Corrente
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Claudia Capponi
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Patrizia Palomba
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Mattia Mirabella
- Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Simona Cascioli
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paolo Palange
- Department of Public Health and Infectious Diseases Pulmonary Division, Policlinico Umberto I Hospital, Rome, Italy
| | - Ilaria Cuccaro
- Department of Public Health and Infectious Diseases Pulmonary Division, Policlinico Umberto I Hospital, Rome, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alimuddin Zumla
- Center for Clinical Microbiology, Division of Infection and Immunity, University College London, London, United Kingdom
- NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, United Kingdom
| | - Markus Maeurer
- Immunotherapy Programme, Champalimaud Foundation, Lisbon, Portugal
- Med Clinic, University of Mainz, Mainz, Germany
| | - Vincenzo Camisa
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Maria Rosaria Vinci
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Annapaola Santoro
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Eleonora Cimini
- Cellular Immunology Laboratory, INMI L Spallanzani, IRCCS, Rome, Italy
| | | | | | | | - Chiara Agrati
- Cellular Immunology Laboratory, INMI L Spallanzani, IRCCS, Rome, Italy
| | | | - Ottavia Porzio
- Medical Laboratory Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carlo Concato
- Virology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Andrea Onetti Muda
- Department of Laboratories, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Massimiliano Raponi
- Health Directorate, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Concetta Quintarelli
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Pediatrics, Sapienza, University of Rome, Rome, Italy
| |
Collapse
|
29
|
Cañas CA. The triggering of post-COVID-19 autoimmunity phenomena could be associated with both transient immunosuppression and an inappropriate form of immune reconstitution in susceptible individuals. Med Hypotheses 2020; 145:110345. [PMID: 33080459 PMCID: PMC7556280 DOI: 10.1016/j.mehy.2020.110345] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/14/2020] [Accepted: 10/10/2020] [Indexed: 01/08/2023]
Abstract
With the progression of the COVID-19 pandemic, there have been different reports about the development of autoimmune diseases once the infection is controlled. After entering the respiratory epithelial cells, SARS-CoV-2-the virus that causes the disease-triggers a severe inflammatory state in some patients known as "cytokine storm" and the development of thrombotic phenomena-both conditions being associated with high mortality. Patients additionally present severe lymphopenia and, in some cases, complement consumption and autoantibody development. There is a normalization of lymphocytes once the infection is controlled. After this, autoimmune conditions of unknown etiology may occur. A hypothesis for the development of post-COVID-19 autoimmunity is proposed based on the consequences of both a transient immunosuppression (both of innate and acquired immunity) in which self-tolerance is lost and an inappropriate form of immune reconstitution that amplifies the process.
Collapse
Affiliation(s)
- Carlos A Cañas
- Unit of Rheumatology, Fundación Valle del Lili, Cra.98 No.18-49, Cali 760032, Colombia.
| |
Collapse
|
30
|
Kazancioglu S, Yilmaz FM, Bastug A, Sakallı A, Ozbay BO, Buyuktarakci C, Bodur H, Yilmaz G. Lymphocyte Subset Alteration and Monocyte CD4 Expression Reduction in Patients with Severe COVID-19. Viral Immunol 2020; 34:342-351. [PMID: 33264073 DOI: 10.1089/vim.2020.0166] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The spectrum of coronavirus disease 2019 (COVID-19) severity, related to cellular immune functions, has not been fully clarified yet. Therefore, this study aimed to investigate the alteration of peripheral blood cells in patients with COVID-19. The flow cytometric characterization of immune cell subset was performed on 69 COVID-19 patients and 21 healthy controls. These data were evaluated based on the disease severity. A total of 69 patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were classified as asymptomatic infection (n = 14), nonsevere (n = 39), and severe (n = 16) groups. Decreased lymphocytes and increased CD14 + 4- monocytes are found in patients with severe COVID-19. Decreased CD4 expression level was observed in the monocytes of patients with severe COVID-19. The total lymphocytes, B and T lymphocytes, CD4+ cells and CD8+ cells, and natural killer (NK) and natural killer T (NKT) cells were found to be decreased in patients with severe COVID-19. The CD4+/CD8+ ratio was not significantly different between patients with COVID-19 and healthy controls. The percentage of activated T cells (CD3+HLA-DR+) and B cells (CD19+CD38+) was lower in patients with severe COVID-19. Age and CD4- monocytes were independent predictors of disease severity. The SARS-CoV-2 infection may affect lymphocyte subsets, resulting in decreased T and B cells, monocytes, and NK and NKT cells. Decreased CD4 expression level by monocytes was significantly correlated with disease severity. Further studies on the host immune response to SARS-CoV-2 infection are necessary to predict the disease severity and protect against the virus.
Collapse
Affiliation(s)
- Sumeyye Kazancioglu
- Department of Infectious Diseases and Clinical Microbiology, and Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Fatma Meric Yilmaz
- Medical Biochemistry Laboratory, Ministry of Health Ankara City Hospital, Ankara, Turkey.,Faculty of Medicine, Medical Biochemistry Department, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Aliye Bastug
- Department of Infectious Diseases and Clinical Microbiology, Ankara City Hospital, Health Science University Turkey, Ankara, Turkey
| | - Arzu Sakallı
- Medical Biochemistry Laboratory, Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Bahadır Orkun Ozbay
- Department of Infectious Diseases and Clinical Microbiology, and Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Cansu Buyuktarakci
- Department of Infectious Diseases and Clinical Microbiology, and Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Hurrem Bodur
- Department of Infectious Diseases and Clinical Microbiology, Ankara City Hospital, Health Science University Turkey, Ankara, Turkey
| | - Gulsen Yilmaz
- Medical Biochemistry Laboratory, Ministry of Health Ankara City Hospital, Ankara, Turkey.,Faculty of Medicine, Medical Biochemistry Department, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
31
|
Tosif S, Neeland MR, Sutton P, Licciardi PV, Sarkar S, Selva KJ, Do LAH, Donato C, Quan Toh Z, Higgins R, Van de Sandt C, Lemke MM, Lee CY, Shoffner SK, Flanagan KL, Arnold KB, Mordant FL, Mulholland K, Bines J, Dohle K, Pellicci DG, Curtis N, McNab S, Steer A, Saffery R, Subbarao K, Chung AW, Kedzierska K, Burgner DP, Crawford NW. Immune responses to SARS-CoV-2 in three children of parents with symptomatic COVID-19. Nat Commun 2020; 11:5703. [PMID: 33177504 PMCID: PMC7658256 DOI: 10.1038/s41467-020-19545-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/20/2020] [Indexed: 12/27/2022] Open
Abstract
Compared to adults, children with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have predominantly mild or asymptomatic infections, but the underlying immunological differences remain unclear. Here, we describe clinical features, virology, longitudinal cellular, and cytokine immune profile, SARS-CoV-2-specific serology and salivary antibody responses in a family of two parents with PCR-confirmed symptomatic SARS-CoV-2 infection and their three children, who tested repeatedly SARS-CoV-2 PCR negative. Cellular immune profiles and cytokine responses of all children are similar to their parents at all timepoints. All family members have salivary anti-SARS-CoV-2 antibodies detected, predominantly IgA, that coincide with symptom resolution in 3 of 4 symptomatic members. Plasma from both parents and one child have IgG antibody against the S1 protein and virus-neutralizing activity detected. Using a systems serology approach, we demonstrate higher levels of SARS-CoV-2-specific antibody features of these family members compared to healthy controls. These data indicate that children can mount an immune response to SARS-CoV-2 without virological confirmation of infection, raising the possibility that immunity in children can prevent the establishment of SARS-CoV-2 infection. Relying on routine virological and serological testing may not identify exposed children, with implications for epidemiological and clinical studies across the life-span.
Collapse
Affiliation(s)
- Shidan Tosif
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia.
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia.
- Department of General Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia.
| | - Melanie R Neeland
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Philip Sutton
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Paul V Licciardi
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Sohinee Sarkar
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Kevin J Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lien Anh Ha Do
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Celeste Donato
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Zheng Quan Toh
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Rachel Higgins
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Carolien Van de Sandt
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Melissa M Lemke
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | - Christina Y Lee
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | | | - Katie L Flanagan
- Department of Infectious Diseases, Launceston General Hospital, Launceston, Tasmania, Australia
- School of Health Sciences and School of Medicine, University of Tasmania, Launceston, Tasmania, Australia
- Department of Immunology and Pathology, Monash University, Commercial Road, Melbourne, Victoria, Australia
- School of Health and Biomedical Science, RMIT University, Melbourne, Victoria, Australia
| | - Kelly B Arnold
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | - Francesca L Mordant
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kim Mulholland
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital, Melbourne, Australia
| | - Julie Bines
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Gastroenterology, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Kate Dohle
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Daniel G Pellicci
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital, Melbourne, Australia
| | - Sarah McNab
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Department of General Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Andrew Steer
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital, Melbourne, Australia
| | - Richard Saffery
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Melbourne, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - David P Burgner
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital, Melbourne, Australia
| | - Nigel W Crawford
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Department of General Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Xu G, Qi F, Li H, Yang Q, Wang H, Wang X, Liu X, Zhao J, Liao X, Liu Y, Liu L, Zhang S, Zhang Z. The differential immune responses to COVID-19 in peripheral and lung revealed by single-cell RNA sequencing. Cell Discov 2020; 6:73. [PMID: 33101705 PMCID: PMC7574992 DOI: 10.1038/s41421-020-00225-2] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/27/2020] [Indexed: 01/08/2023] Open
Abstract
Understanding the mechanism that leads to immune dysfunction in severe coronavirus disease 2019 (COVID-19) is crucial for the development of effective treatment. Here, using single-cell RNA sequencing, we characterized the peripheral blood mononuclear cells (PBMCs) from uninfected controls and COVID-19 patients and cells in paired broncho-alveolar lavage fluid (BALF). We found a close association of decreased dendritic cells (DCs) and increased monocytes resembling myeloid-derived suppressor cells (MDSCs), which correlated with lymphopenia and inflammation in the blood of severe COVID-19 patients. Those MDSC-like monocytes were immune-paralyzed. In contrast, monocyte-macrophages in BALFs of COVID-19 patients produced massive amounts of cytokines and chemokines, but secreted little interferons. The frequencies of peripheral T cells and NK cells were significantly decreased in severe COVID-19 patients, especially for innate-like T and various CD8+ T cell subsets, compared to healthy controls. In contrast, the proportions of various activated CD4+ T cell subsets among the T cell compartment, including Th1, Th2, and Th17-like cells were increased and more clonally expanded in severe COVID-19 patients. Patients' peripheral T cells showed no sign of exhaustion or augmented cell death, whereas T cells in BALFs produced higher levels of IFNG, TNF, CCL4, CCL5, etc. Paired TCR tracking indicated abundant recruitment of peripheral T cells to the severe patients' lung. Together, this study comprehensively depicts how the immune cell landscape is perturbed in severe COVID-19.
Collapse
Affiliation(s)
- Gang Xu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518112 China
| | - Furong Qi
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518112 China
| | - Hanjie Li
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055 China
| | - Qianting Yang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518112 China
| | - Haiyan Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518112 China
| | - Xin Wang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518112 China
| | - Xiaoju Liu
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515 China
| | - Juanjuan Zhao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518112 China
| | - Xuejiao Liao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518112 China
| | - Yang Liu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518112 China
| | - Lei Liu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518112 China
| | - Shuye Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518112 China
| |
Collapse
|
33
|
Abstract
The impact of host adaptive immune response on COVID-19 has now become a critical issue in absence of specific therapy and immunotherapies. In SARS CoV-2 infection, the immune response is thought to contribute both to the pathogenesis of the disease and to protection during its resolution. While mild cases develop an immune response that contributes to host protection, immunity of severely infected patients is a balance between harmful and protective immune responses. The severity of the disease has raised many questions about the kinetic, amplitude and the quality of adaptive immunity to the virus and its generation during the early phases of infection in severe, mild and asymptomatic patients. The role of antibody and CD4+ and CD8+ T cell responses have been studied and the development of an adaptive immunity seems to correlate with convalescence. The bioinformatics study of the T and B epitopes of coronaviruses has raised the question of the existence of cross-immunity between SARS-CoV-2 and other coronaviruses such as MERS-CoV and SARS-CoV. In this review, we discuss the adaptive immune responses and their potential roles in protection during COVID-19.
Collapse
Affiliation(s)
- Béhazine Combadière
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 91 boulevard de l'Hôpital, 75013 Paris, France
| |
Collapse
|
34
|
Liu T, Guo Y, Zhao J, He S, Bai Y, Wang N, Lin Y, Liu Q, Xu X. Systems Pharmacology and Verification of ShenFuHuang Formula in Zebrafish Model Reveal Multi-Scale Treatment Strategy for Septic Syndrome in COVID-19. Front Pharmacol 2020; 11:584057. [PMID: 33041827 PMCID: PMC7523021 DOI: 10.3389/fphar.2020.584057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) has affected millions of people worldwide. Critically ill COVID-19 patients develop viral septic syndrome, including inflammatory damage, immune dysfunction, and coagulation disorder. In this study, we investigated ShenFuHuang formula (SFH), a traditional Chinese medicine, which has been widely used as complementary therapy for clinical treatment of COVID-19 in Wuhan, to understand its pharmacological properties. Results of systems pharmacology identified 49 active compounds of SFH and their 69 potential targets, including GSK3β, ESR1, PPARG, PTGS2, AKR1B10, and MAPK14. Network analysis illustrated that the targets of SFH may be involved in viral disease, bacterial infection/mycosis, and metabolic disease. Moreover, signaling pathway analysis showed that Toll-like receptors, MAPK, PPAR, VEGF, NOD-like receptor, and NF-kappa B signaling pathways are highly connected with the potential targets of SFH. We further employed multiple zebrafish models to confirm the pharmacological effects of SFH. Results showed that SFH treatment significantly inhibited the inflammatory damage by reducing the generation of neutrophils in Poly (I:C)-induced viral infection model. Moreover, SFH treatment could improve the phagocytosis of macrophages and enhance the expression of immune genes in an immune deficiency model. Furthermore, SFH treatment exhibited promising anti-thrombosis effect in a thrombus model. This study provided additional evidence of SFH formula for treating COVID-19 patients with septic syndrome using multiple-scale estimation.
Collapse
Affiliation(s)
- Tengwen Liu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong
| | - Yuhong Guo
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Jingxia Zhao
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Shasha He
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Yunjing Bai
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Ning Wang
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Yan Lin
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Qingquan Liu
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| | - Xiaolong Xu
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China
| |
Collapse
|
35
|
Campana P, Parisi V, Leosco D, Bencivenga D, Della Ragione F, Borriello A. Dendritic Cells and SARS-CoV-2 Infection: Still an Unclarified Connection. Cells 2020; 9:E2046. [PMID: 32911691 PMCID: PMC7564940 DOI: 10.3390/cells9092046] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 01/19/2023] Open
Abstract
The ongoing pandemic due to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has so far infected about 2.42 × 107 (as at 27 August 2020) subjects with more than 820,000 deaths. It is the third zoonotic coronavirus-dependent outbreak in the last twenty years and represents a major infective threat for public health worldwide. A main aspect of the infection, in analogy to other viral infections, is the so-called "cytokine storm", an inappropriate molecular response to virus spread which plays major roles in tissue and organ damage. Immunological therapies, including vaccines and humanized monoclonal antibodies, have been proposed as major strategies for prevention and treatment of the disease. Accordingly, a detailed mechanistic knowledge of the molecular events with which the virus infects cells and induces an immunological response appears necessary. In this review, we will report details of the initial process of SARS-CoV-2 cellular entry with major emphasis on the maturation of the spike protein. Then, a particular focus will be devoted to describe the possible mechanisms by which dendritic cells, a major cellular component of innate and adaptive immune responses, may play a role in the spread of the virus in the human body and in the clinical evolution of the disease.
Collapse
Affiliation(s)
- Pasquale Campana
- Department of Translational Medical Sciences, University of Naples ‘Federico II’, Via Sergio Pansini 5, 80131 Naples, Italy; (P.C.); (V.P.); (D.L.)
| | - Valentina Parisi
- Department of Translational Medical Sciences, University of Naples ‘Federico II’, Via Sergio Pansini 5, 80131 Naples, Italy; (P.C.); (V.P.); (D.L.)
| | - Dario Leosco
- Department of Translational Medical Sciences, University of Naples ‘Federico II’, Via Sergio Pansini 5, 80131 Naples, Italy; (P.C.); (V.P.); (D.L.)
| | - Debora Bencivenga
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy;
| | - Fulvio Della Ragione
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy;
| | - Adriana Borriello
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via De Crecchio 7, 80138 Naples, Italy;
| |
Collapse
|
36
|
Lebeau G, Vagner D, Frumence É, Ah-Pine F, Guillot X, Nobécourt E, Raffray L, Gasque P. Deciphering SARS-CoV-2 Virologic and Immunologic Features. Int J Mol Sci 2020; 21:E5932. [PMID: 32824753 PMCID: PMC7460647 DOI: 10.3390/ijms21165932] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 and its associated pathology, COVID-19, have been of particular concerns these last months due to the worldwide burden they represent. The number of cases requiring intensive care being the critical point in this epidemic, a better understanding of the pathophysiology leading to these severe cases is urgently needed. Tissue lesions can be caused by the pathogen or can be driven by an overwhelmed immune response. Focusing on SARS-CoV-2, we and others have observed that this virus can trigger indeed an immune response that can be dysregulated in severe patients and leading to further injury to multiple organs. The purpose of the review is to bring to light the current knowledge about SARS-CoV-2 virologic and immunologic features. Thus, we address virus biology, life cycle, tropism for many organs and how ultimately it will affect several host biological and physiological functions, notably the immune response. Given that therapeutic avenues are now highly warranted, we also discuss the immunotherapies available to manage the infection and the clinical outcomes.
Collapse
Affiliation(s)
- Grégorie Lebeau
- Unité de Recherche Études Pharmaco-Immunologiques, Centre Hospitalier Universitaire La Réunion Site Félix Guyon, CS11021, 97400 Saint Denis de La Réunion, France; (D.V.); (É.F.); (X.G.); (P.G.)
- Laboratoire de Biologie, Secteur Laboratoire d’immunologie Clinique et Expérimentale de la Zone de l’océan Indien (LICE-OI), Centre Hospitalier Universitaire La Réunion Site Félix Guyon, CS11021, 97400 Saint Denis de La Réunion, France
| | - Damien Vagner
- Unité de Recherche Études Pharmaco-Immunologiques, Centre Hospitalier Universitaire La Réunion Site Félix Guyon, CS11021, 97400 Saint Denis de La Réunion, France; (D.V.); (É.F.); (X.G.); (P.G.)
- Unité Mixte de Recherche Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de La Réunion, INSERM UMR 1187, CNRS 9192, IRD 249, Platform CYROI, 2 rue Maxime Rivière, 97491 Sainte Clotilde, La Réunion, France
| | - Étienne Frumence
- Unité de Recherche Études Pharmaco-Immunologiques, Centre Hospitalier Universitaire La Réunion Site Félix Guyon, CS11021, 97400 Saint Denis de La Réunion, France; (D.V.); (É.F.); (X.G.); (P.G.)
- Laboratoire de Biologie, Secteur Laboratoire d’immunologie Clinique et Expérimentale de la Zone de l’océan Indien (LICE-OI), Centre Hospitalier Universitaire La Réunion Site Félix Guyon, CS11021, 97400 Saint Denis de La Réunion, France
| | - Franck Ah-Pine
- Service d’anatomo-Pathologie, Centre Hospitalier Universitaire Sud Réunion, 97410 Saint Pierre, France;
| | - Xavier Guillot
- Unité de Recherche Études Pharmaco-Immunologiques, Centre Hospitalier Universitaire La Réunion Site Félix Guyon, CS11021, 97400 Saint Denis de La Réunion, France; (D.V.); (É.F.); (X.G.); (P.G.)
- Service de Rhumatologie, Centre Hospitalier Universitaire La Réunion Site Félix Guyon, CS11021, 97400 Saint Denis de La Réunion, France
| | - Estelle Nobécourt
- Service d’endocrinologie Diabétologie, Centre Hospitalier Universitaire Sud Réunion, 97410 Saint Pierre, France;
- Université de Formation et de Recherche Santé, Université de la Réunion, 97400 Saint-Denis, France
| | - Loïc Raffray
- Service de Médecine Interne, Centre Hospitalier Universitaire La Réunion Site Félix Guyon, CS11021, 97400 Saint Denis de La Réunion, France;
| | - Philippe Gasque
- Unité de Recherche Études Pharmaco-Immunologiques, Centre Hospitalier Universitaire La Réunion Site Félix Guyon, CS11021, 97400 Saint Denis de La Réunion, France; (D.V.); (É.F.); (X.G.); (P.G.)
- Laboratoire de Biologie, Secteur Laboratoire d’immunologie Clinique et Expérimentale de la Zone de l’océan Indien (LICE-OI), Centre Hospitalier Universitaire La Réunion Site Félix Guyon, CS11021, 97400 Saint Denis de La Réunion, France
| |
Collapse
|
37
|
Abstract
The ongoing pandemic severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) causes a disproportionate number of severe cases and deaths in older adults. Severe SARS-CoV-2-associated disease (coronavirus disease 2019 (COVID-19)) was declared a pandemic by the World Health Organization in March 2020 and is characterized by cytokine storm, acute respiratory distress syndrome, and in some cases by systemic inflammation-related pathology. Currently, our knowledge of the determinants of severe COVID-19 is primarily observational. Here, I review emerging evidence to argue that monocytes, a circulating innate immune cell, are principal players in cytokine storm and associated pathologies in COVID-19. I also describe changes in monocyte function and phenotype that are characteristic of both aging and severe COVID-19, which suggests a potential mechanism underlying increased morbidity and mortality due to SARS-CoV-2 infection in older adults. The innate immune system is therefore a potentially important target for therapeutic treatment of COVID-19, but experimental studies are needed, and SARS-CoV-2 presents unique challenges for pre-clinical and mechanistic studies in vivo. The immediate establishment of colonies of SARS-CoV-2-susceptible animal models for aging studies, as well as strong collaborative efforts in the geroscience community, will be required in order to develop the therapies needed to combat severe COVID-19 in older adult populations.
Collapse
Affiliation(s)
- Brandt D Pence
- School of Health Studies, University of Memphis, Memphis, TN, 38152, USA.
- Center for Nutraceutical and Dietary Supplement Research, University of Memphis, Memphis, TN, 38152, USA.
- University of Memphis, 304 Elma Roane Fieldhouse, 495 Zach H. Curlin St., Memphis, TN, 38152, USA.
| |
Collapse
|