Cytokines in SARS-CoV-2 Infection

A special issue of Viruses (ISSN 1999-4915). This special issue belongs to the section "SARS-CoV-2 and COVID-19".

Deadline for manuscript submissions: closed (29 March 2023) | Viewed by 20944

Special Issue Editors

Institute of Medical Microbiology, Jinan University, Guangzhou, China
Interests: viral pathogenesis; host innate immunity; anti-viral therapy
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Microbiology, Kohsar University Murree, Murree, Pakistan
Interests: microbiology; virology; immunology; host pathogen interaction

E-Mail Website
Guest Editor
Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 510275, China
Interests: influenza virus; coronavirus; universal vaccine; monoclonal antibody; neutralizing antibody; cross-reactive antibody; broad spectrum antibody; antibody affinity maturation; BCR ; single cell sequence; hemagglutinin, neuraminidase
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleague,

SARS-CoV-2 causing COVID-19 appears to act on the activation and maturation of chemokines and cytokines, which in turn evokes the immune system and triggers cytokine release syndrome (CRS) and cytokine storm. Most COVID-19 patients have high levels of cytokines with severe symptoms linked to multiple organ dysfunction syndrome (MODS), acute respiratory distress syndrome (ARDS), and even death. Therapeutic inventations are used for the inhibition of cytokines in order to avoid a cytokine storm.

This Special Issue welcomes studies focusing on (1) mechanisms of cytokines production and virus-related pathogenesis upon SARS-CoV-2 infection, and (2) therapeutic strategies for COVID-19 targeting cytokines or inflammatory response.

We are looking forward to submissions of both original research and review articles that are involved in these themes.

Dr. Zhen Luo
Dr. Muhammad Adnan Shereen
Prof. Dr. Yao-Qing Chen
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Viruses is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • SARS-CoV-2
  • COVID-19
  • cytokine and chemokines
  • cytokine Storm
  • therapeutic inventations

Published Papers (10 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

8 pages, 280 KiB  
Communication
Association of Polymorphisms of IL-6 Pathway Genes (IL6, IL6R and IL6ST) with COVID-19 Severity in an Amazonian Population
by Fabíola Brasil Barbosa Rodrigues, Rosilene da Silva, Erika Ferreira dos Santos, Mioni Thieli Figueiredo Magalhães de Brito, Andréa Luciana Soares da Silva, Mauro de Meira Leite, Flávia Póvoa da Costa, Maria de Nazaré do Socorro de Almeida Viana, Kevin Matheus Lima de Sarges, Marcos Henrique Damasceno Cantanhede, Adriana de Oliveira Lameira Veríssimo, Mayara da Silva Carvalho, Daniele Freitas Henriques, Carla Pinheiro da Silva, Igor Brasil Costa, Juliana Abreu Lima Nunes, Iran Barros Costa, Giselle Maria Rachid Viana, Maria Alice Freitas Queiroz, Sandra Souza Lima, Jeferson da Costa Lopes, Maria Karoliny da Silva Torres, Izaura Maria Vieira Cayres Vallinoto, Carlos David Araújo Bichara, Antonio Carlos Rosário Vallinoto and Eduardo José Melo dos Santosadd Show full author list remove Hide full author list
Viruses 2023, 15(5), 1197; https://doi.org/10.3390/v15051197 - 19 May 2023
Cited by 2 | Viewed by 1168
Abstract
Interleukin-6 has been recognized as a major role player in COVID-19 severity, being an important regulator of the cytokine storm. Hence, the evaluation of the influence of polymorphisms in key genes of the IL-6 pathway, namely IL6, IL6R, and IL6ST, may provide valuable [...] Read more.
Interleukin-6 has been recognized as a major role player in COVID-19 severity, being an important regulator of the cytokine storm. Hence, the evaluation of the influence of polymorphisms in key genes of the IL-6 pathway, namely IL6, IL6R, and IL6ST, may provide valuable prognostic/predictive markers for COVID-19. The present cross-sectional study genotyped three SNPs (rs1800795, rs2228145, and rs7730934) at IL6. IL6R and IL6ST genes, respectively, in 227 COVID-19 patients (132 hospitalized and 95 non-hospitalized). Genotype frequencies were compared between these groups. As a control group, published data on gene and genotype frequencies were gathered from published studies before the pandemic started. Our major results point to an association of the IL6 C allele with COVID-19 severity. Moreover, IL-6 plasmatic levels were higher among IL6 CC genotype carriers. Additionally, the frequency of symptoms was higher at IL6 CC and IL6R CC genotypes. In conclusion, the data suggest an important role of IL6 C allele and IL6R CC genotype on COVID-19 severity, in agreement with indirect evidence from the literature about the association of these genotypes with mortality rates, pneumonia, and heightening of protein plasmatic levels pro-inflammatory driven effects. Full article
(This article belongs to the Special Issue Cytokines in SARS-CoV-2 Infection)
23 pages, 2882 KiB  
Article
Cytokine Response Following SARS-CoV-2 Antigen Stimulation in Patients with Predominantly Antibody Deficiencies
by Zane Lucane, Baiba Slisere, Gita Gersone, Sindija Papirte, Linda Gailite, Peteris Tretjakovs and Natalja Kurjane
Viruses 2023, 15(5), 1146; https://doi.org/10.3390/v15051146 - 10 May 2023
Cited by 1 | Viewed by 1574
Abstract
Predominantly antibody deficiencies (PADs) are inborn disorders characterized by immune dysregulation and increased susceptibility to infections. Response to vaccination, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), may be impaired in these patients, and studies on responsiveness correlates, including cytokine signatures to antigen [...] Read more.
Predominantly antibody deficiencies (PADs) are inborn disorders characterized by immune dysregulation and increased susceptibility to infections. Response to vaccination, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), may be impaired in these patients, and studies on responsiveness correlates, including cytokine signatures to antigen stimulation, are sparse. In this study, we aimed to describe the spike-specific cytokine response following whole-blood stimulation with SARS-CoV-2 spike peptides in patients with PAD (n = 16 with common variable immunodeficiency and n = 15 with selective IgA deficiency) and its relationship with the occurrence of coronavirus disease 2019 (COVID-19) during up to 10-month follow-up period. Spike-induced antibody and cytokine production was measured using ELISA (anti-spike IgG, IFN-γ) and xMAP technology (interleukin-1β (IL-1β), IL-4, IL-6, IL-10, IL-15, IL-17A, IL-21, TNF-α, TGF-β1). No difference was found in the production of cytokines between patients with PAD and controls. Anti-spike IgG and cytokine levels did not predict contraction of COVID-19. The only cytokine that distinguished between vaccinated and naturally infected unvaccinated PAD patients was IFN-γ (median 0.64 (IQR = 1.08) in vaccinated vs. 0.10 (IQR = 0.28) in unvaccinated). This study describes the spike-specific cytokine response to SARS-CoV-2 antigens, which is not predictive of contracting COVID-19 during the follow-up. Full article
(This article belongs to the Special Issue Cytokines in SARS-CoV-2 Infection)
Show Figures

Figure 1

13 pages, 2589 KiB  
Article
SARS-CoV-2-Induced TSLP Is Associated with Duration of Hospital Stay in COVID-19 Patients
by Luke Gerla, Subhabrata Moitra, Desmond Pink, Natasha Govindasamy, Marc Duchesne, Eileen Reklow, Angela Hillaby, Amy May, John D. Lewis, Lyle Melenka, Tom C. Hobman, Irvin Mayers and Paige Lacy
Viruses 2023, 15(2), 556; https://doi.org/10.3390/v15020556 - 17 Feb 2023
Cited by 4 | Viewed by 2323
Abstract
Thymic stromal lymphopoietin (TSLP) is an epithelium-derived pro-inflammatory cytokine involved in lung inflammatory responses. Previous studies show conflicting observations in blood TSLP in COVID-19, while none report SARS-CoV-2 inducing TSLP expression in bronchial epithelial cells. Our objective in this study was to determine [...] Read more.
Thymic stromal lymphopoietin (TSLP) is an epithelium-derived pro-inflammatory cytokine involved in lung inflammatory responses. Previous studies show conflicting observations in blood TSLP in COVID-19, while none report SARS-CoV-2 inducing TSLP expression in bronchial epithelial cells. Our objective in this study was to determine whether TSLP levels increase in COVID-19 patients and if SARS-CoV-2 induces TSLP expression in bronchial epithelial cells. Plasma cytokine levels were measured in patients hospitalized with confirmed COVID-19 and age- and sex-matched healthy controls. Demographic and clinical information from COVID-19 patients was collected. We determined associations between plasma TSLP and clinical parameters using Poisson regression. Cultured human nasal (HNEpC) and bronchial epithelial cells (NHBEs), Caco-2 cells, and patient-derived bronchial epithelial cells (HBECs) obtained from elective bronchoscopy were infected in vitro with SARS-CoV-2, and secretion as well as intracellular expression of TSLP was detected by immunofluorescence. Increased TSLP levels were detected in the plasma of hospitalized COVID-19 patients (603.4 ± 75.4 vs 997.6 ± 241.4 fg/mL, mean ± SEM), the levels of which correlated with duration of stay in hospital (β: 0.11; 95% confidence interval (CI): 0.01–0.21). In cultured NHBE and HBECs but not HNEpCs or Caco-2 cells, TSLP levels were significantly elevated after 24 h post-infection with SARS-CoV-2 (p < 0.001) in a dose-dependent manner. Plasma TSLP in COVID-19 patients significantly correlated with duration of hospitalization, while SARS-CoV-2 induced TSLP secretion from bronchial epithelial cells in vitro. Based on our findings, TSLP may be considered an important therapeutic target for COVID-19 treatment. Full article
(This article belongs to the Special Issue Cytokines in SARS-CoV-2 Infection)
Show Figures

Figure 1

15 pages, 4240 KiB  
Article
Circulating Interleukin-8 Dynamics Parallels Disease Course and Is Linked to Clinical Outcomes in Severe COVID-19
by Ranit D’Rozario, Deblina Raychaudhuri, Purbita Bandopadhyay, Jafar Sarif, Priyanka Mehta, Chinky Shiu Chen Liu, Bishnu Prasad Sinha, Jayasree Roy, Ritwik Bhaduri, Monidipa Das, Sanghamitra Bandyopadhyay, Shekhar Ranjan Paul, Shilpak Chatterjee, Rajesh Pandey, Yogiraj Ray and Dipyaman Ganguly
Viruses 2023, 15(2), 549; https://doi.org/10.3390/v15020549 - 16 Feb 2023
Cited by 5 | Viewed by 1741
Abstract
Severe COVID-19 frequently features a systemic deluge of cytokines. Circulating cytokines that can stratify risks are useful for more effective triage and management. Here, we ran a machine-learning algorithm on a dataset of 36 plasma cytokines in a cohort of severe COVID-19 to [...] Read more.
Severe COVID-19 frequently features a systemic deluge of cytokines. Circulating cytokines that can stratify risks are useful for more effective triage and management. Here, we ran a machine-learning algorithm on a dataset of 36 plasma cytokines in a cohort of severe COVID-19 to identify cytokine/s useful for describing the dynamic clinical state in multiple regression analysis. We performed RNA-sequencing of circulating blood cells collected at different time-points. From a Bayesian Information Criterion analysis, a combination of interleukin-8 (IL-8), Eotaxin, and Interferon-γ (IFNγ) was found to be significantly linked to blood oxygenation over seven days. Individually testing the cytokines in receiver operator characteristics analyses identified IL-8 as a strong stratifier for clinical outcomes. Circulating IL-8 dynamics paralleled disease course. We also revealed key transitions in immune transcriptome in patients stratified for circulating IL-8 at three time-points. The study identifies plasma IL-8 as a key pathogenic cytokine linking systemic hyper-inflammation to the clinical outcomes in COVID-19. Full article
(This article belongs to the Special Issue Cytokines in SARS-CoV-2 Infection)
Show Figures

Figure 1

16 pages, 3311 KiB  
Article
Downregulation of the Protein C Signaling System Is Associated with COVID-19 Hypercoagulability—A Single-Cell Transcriptomics Analysis
by Bruna Rafaela Santos Silva, Carlos Poblete Jara, Davi Sidarta-Oliveira, Licio A. Velloso, William H. Velander and Eliana P. Araújo
Viruses 2022, 14(12), 2753; https://doi.org/10.3390/v14122753 - 09 Dec 2022
Cited by 3 | Viewed by 1872
Abstract
Because of the interface between coagulation and the immune response, it is expected that COVID-19-associated coagulopathy occurs via activated protein C signaling. The objective was to explore putative changes in the expression of the protein C signaling network in the liver, peripheral blood [...] Read more.
Because of the interface between coagulation and the immune response, it is expected that COVID-19-associated coagulopathy occurs via activated protein C signaling. The objective was to explore putative changes in the expression of the protein C signaling network in the liver, peripheral blood mononuclear cells, and nasal epithelium of patients with COVID-19. Single-cell RNA-sequencing data from patients with COVID-19 and healthy subjects were obtained from the COVID-19 Cell Atlas database. A functional protein–protein interaction network was constructed for the protein C gene. Patients with COVID-19 showed downregulation of protein C and components of the downstream protein C signaling cascade. The percentage of hepatocytes expressing protein C was lower. Part of the liver cell clusters expressing protein C presented increased expression of ACE2. In PBMC, there was increased ACE2, inflammatory, and pro-coagulation transcripts. In the nasal epithelium, PROC, ACE2, and PROS1 were expressed by the ciliated cell cluster, revealing co-expression of ACE-2 with transcripts encoding proteins belonging to the coagulation and immune system interface. Finally, there was upregulation of coagulation factor 3 transcript in the liver and PBMC. Protein C could play a mechanistic role in the hypercoagulability syndrome affecting patients with severe COVID-19. Full article
(This article belongs to the Special Issue Cytokines in SARS-CoV-2 Infection)
Show Figures

Figure 1

13 pages, 732 KiB  
Article
Evaluation of Cardiac Biomarkers and Expression Analysis of IL-1, IL-6, IL-10, IL-17, and IL-25 among COVID-19 Patients from Pakistan
by Razi Ullah, Jadoon Khan, Nosheen Basharat, Danqun Huo, Ahmad Ud Din and Guixue Wang
Viruses 2022, 14(10), 2149; https://doi.org/10.3390/v14102149 - 29 Sep 2022
Cited by 3 | Viewed by 2001
Abstract
Coronavirus disease 19 (COVID-19) is caused by viral infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Where upregulation of several important biomarkers and multiple organ dysfunction occurs, this study aimed to evaluate the association of cardiac biomarkers and CS induced acute lung [...] Read more.
Coronavirus disease 19 (COVID-19) is caused by viral infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Where upregulation of several important biomarkers and multiple organ dysfunction occurs, this study aimed to evaluate the association of cardiac biomarkers and CS induced acute lung damage with disease severity and mortality in survival of COVID-19 patients. A total of 500 COVID-19 patients with elevated cardiac biomarkers were studied for the analysis of myocardial abnormality through cardiac enzymes, inflammatory biomarkers, and the expression analysis of various cytokines, including IL-1, IL-6, IL-10, IL-17, and IL-25 genes. The elevation of various cardiac enzymes including LDH (87%), CK (78.4%), TNI (80.4%), CK-MB (83%), and D-dimer (80.8%) were found correlated (p < 0.001) with COVID-19 infection. Cardiac enzyme elevation was highly associated with an increased level of inflammatory biomarkers such as CRP (14.2%), SAA (11.4%) and erythrocyte sedimentation rate (ESR) (7.8%) (p = 0.001 for all). The quantitative expression analysis of IL-10, 1L-17, and 1L-25 were found to be high, while those of IL-1 and IL-6 were moderately elevated. The death-to-live ratio of COVID-19 patients was 457:43 indicating that the patients having elevated levels of both CKMB, D-dimer, CK and IL-1, IL-6, IL-10 and D-dimer, Troponin, CK and IL-1, IL-10 had high fatality rate (73% and 12% respectively). The current finding concludes that the evaluation of cardiac biomarkers with cytokine storm plays a significant role in COVID-19-associated anatomical organ damage, myocardial injury, and mortality. Physicians should pay special attention to cardiac biomarkers in patients with old age, inflammation, and comorbidities among COVID-19 infections. Full article
(This article belongs to the Special Issue Cytokines in SARS-CoV-2 Infection)
Show Figures

Figure 1

13 pages, 2020 KiB  
Article
High Expression of HERV-K (HML-2) Might Stimulate Interferon in COVID-19 Patients
by Yaolin Guo, Caiqin Yang, Yongjian Liu, Tianyi Li, Hanping Li, Jingwan Han, Lei Jia, Xiaolin Wang, Bohan Zhang, Jingyun Li and Lin Li
Viruses 2022, 14(5), 996; https://doi.org/10.3390/v14050996 - 07 May 2022
Cited by 10 | Viewed by 2806
Abstract
Background. Interferon is a marker of host antiviral immunity, which is disordered in COVID-19 patients. ERV can affect the secretion of interferon through the cGAS-STING pathway. In this study, we explored whether IFN-I and HERV-K (HML-2) were activated in COVID-19 patients and whether [...] Read more.
Background. Interferon is a marker of host antiviral immunity, which is disordered in COVID-19 patients. ERV can affect the secretion of interferon through the cGAS-STING pathway. In this study, we explored whether IFN-I and HERV-K (HML-2) were activated in COVID-19 patients and whether there was an interaction between them. Methods. We collected blood samples from COVID-19 patients and healthy controls. We first detected the expression of HERV-K (HML-2) gag, env, and pol genes and IFN-I-related genes between patients and healthy people by qPCR, synchronously detected VERO cells infected with SARS-CoV-2. Then, the chromosome distributions of highly expressed HERV-K (HML-2) gag, env, and pol genes were mapped by the next-generation sequencing results, and GO analysis was performed on the related genes. Results. We found that the HERV-K (HML-2) gag, env, and pol genes were highly expressed in COVID-19 patients and VERO cells infected with SARS-CoV-2. The interferon-related genes IFNB1, ISG15, and IFIT1 were also activated in COVID-19 patients, and GO analysis showed that HERV-K (HML-2) can regulate the secretion of interferon. Conclusions. The high expression of HERV-K (HML-2) might activate the increase of interferon in COVID-19 patients, proving that HERV-K does not only play a negative role in the human body. Full article
(This article belongs to the Special Issue Cytokines in SARS-CoV-2 Infection)
Show Figures

Figure 1

Review

Jump to: Research, Other

22 pages, 1538 KiB  
Review
Airway Epithelial-Derived Immune Mediators in COVID-19
by Tony J. F. Guo, Gurpreet K. Singhera, Janice M. Leung and Delbert R. Dorscheid
Viruses 2023, 15(8), 1655; https://doi.org/10.3390/v15081655 - 29 Jul 2023
Viewed by 1669
Abstract
The airway epithelium, which lines the conducting airways, is central to the defense of the lungs against inhaled particulate matter and pathogens such as SARS-CoV-2, the virus that causes COVID-19. Recognition of pathogens results in the activation of an innate and intermediate immune [...] Read more.
The airway epithelium, which lines the conducting airways, is central to the defense of the lungs against inhaled particulate matter and pathogens such as SARS-CoV-2, the virus that causes COVID-19. Recognition of pathogens results in the activation of an innate and intermediate immune response which involves the release of cytokines and chemokines by the airway epithelium. This response can inhibit further viral invasion and influence adaptive immunity. However, severe COVID-19 is characterized by a hyper-inflammatory response which can give rise to clinical presentations including lung injury and lead to acute respiratory distress syndrome, viral pneumonia, coagulopathy, and multi-system organ failure. In response to SARS-CoV-2 infection, the airway epithelium can mount a maladaptive immune response which can delay viral clearance, perpetuate excessive inflammation, and contribute to the pathogenesis of severe COVID-19. In this article, we will review the barrier and immune functions of the airway epithelium, how SARS-CoV-2 can interact with the epithelium, and epithelial-derived cytokines and chemokines and their roles in COVID-19 and as biomarkers. Finally, we will discuss these immune mediators and their potential as therapeutic targets in COVID-19. Full article
(This article belongs to the Special Issue Cytokines in SARS-CoV-2 Infection)
Show Figures

Figure 1

14 pages, 1365 KiB  
Review
Type 2 Immunity and Its Impact on COVID-19 Infection in the Airways
by Prabuddha S. Pathinayake, Nikhil T. Awatade and Peter A. B. Wark
Viruses 2023, 15(2), 402; https://doi.org/10.3390/v15020402 - 31 Jan 2023
Cited by 3 | Viewed by 2678
Abstract
Type 2 immune responses are characterized by elevated type 2 cytokines and blood eosinophilia. Emerging evidence suggests that people with chronic type 2 inflammatory lung diseases are not particularly susceptible to SARS-CoV-2 infection. Intriguingly, recent in vitro, ex vivo research demonstrates type 2 [...] Read more.
Type 2 immune responses are characterized by elevated type 2 cytokines and blood eosinophilia. Emerging evidence suggests that people with chronic type 2 inflammatory lung diseases are not particularly susceptible to SARS-CoV-2 infection. Intriguingly, recent in vitro, ex vivo research demonstrates type 2 cytokines, particularly IL-13, reduce the risk of SARS-CoV-2 infection in the airway epithelium. IL-13 treatment in airway epithelial cells followed by SARS-CoV-2 diminished viral entry, replication, spread, and cell death. IL-13 reduces the expression of the angiotensin-converting enzyme 2 (ACE2) receptor in the airway epithelium and transmembrane serine protease 2 (TMPRSS2), particularly in ciliated cells. It also alters the cellular composition toward a secretory-cell-rich phenotype reducing total ciliated cells and, thus, reducing viral tropism. IL-13 enhances Muc5ac mucin and glycocalyx secretion in the periciliary layer, which acts as a physical barrier to restrict virus attachment. Moreover, type 2 airway immune cells, such as M2 alveolar macrophages, CD4+ tissue-resident memory T cells, and innate lymphoid 2 cells, may also rescue type 2 airways from SARS-CoV-2-induced adverse effects. In this review, we discuss recent findings that demonstrate how type 2 immunity alters immune responses against SARS-CoV-2 and its consequences on COVID-19 pathogenesis. Full article
(This article belongs to the Special Issue Cytokines in SARS-CoV-2 Infection)
Show Figures

Figure 1

Other

Jump to: Research, Review

11 pages, 1669 KiB  
Brief Report
Proinflammatory Responses in SARS-CoV-2 and Soluble Spike Glycoprotein S1 Subunit Activated Human Macrophages
by Kim Chiok, Kevin Hutchison, Lindsay Grace Miller, Santanu Bose and Tanya A. Miura
Viruses 2023, 15(3), 754; https://doi.org/10.3390/v15030754 - 15 Mar 2023
Cited by 6 | Viewed by 1802
Abstract
Critically ill COVID-19 patients display signs of generalized hyperinflammation. Macrophages trigger inflammation to eliminate pathogens and repair tissue, but this process can also lead to hyperinflammation and resulting exaggerated disease. The role of macrophages in dysregulated inflammation during SARS-CoV-2 infection is poorly understood. [...] Read more.
Critically ill COVID-19 patients display signs of generalized hyperinflammation. Macrophages trigger inflammation to eliminate pathogens and repair tissue, but this process can also lead to hyperinflammation and resulting exaggerated disease. The role of macrophages in dysregulated inflammation during SARS-CoV-2 infection is poorly understood. We inoculated and treated human macrophage cell line THP-1 with SARS-CoV-2 and purified, glycosylated, soluble SARS-CoV-2 spike protein S1 subunit (S1) to clarify the role of macrophages in pro-inflammatory responses. Soluble S1 upregulated TNF-α and CXCL10 mRNAs, and induced secretion of TNF-α from THP-1 macrophages. While THP-1 macrophages did not support productive SARS-CoV-2 replication or viral entry, virus exposure resulted in upregulation of both TNF-α and CXCL10 genes. Our study shows that extracellular soluble S1 protein is a key viral component inducing pro-inflammatory responses in macrophages, independent of virus replication. Thus, virus- or soluble S1-activated macrophages may become sources of pro-inflammatory mediators contributing to hyperinflammation in COVID-19 patients. Full article
(This article belongs to the Special Issue Cytokines in SARS-CoV-2 Infection)
Show Figures

Figure 1

Back to TopTop