Surviving the Storm: Cytokine Biosignature in SARS-CoV-2 Severity Prediction
Abstract
:Simple Summary
Abstract
1. Introduction
2. Objectives of the Study
3. Material and Methods
4. Immune Response in SARS-CoV-2 Infection
5. Neutrophilia
6. Lymphopenia
7. Antibody-Mediated Effect of B Lymphocytes
8. Monocyte and Macrophage Dysregulation
9. Response of Interferon Type 1
10. Interleukin 6
11. Interleukin 7
12. Interleukin 10
13. Interleukin 12
14. Interleukin 2
15. Interleukin 17
16. Tumor Necrosis Factor α
17. Interferon γ
18. Interleukin 1β
19. Granulocyte-Macrophage-Colony-Stimulating Factor (GM-CSF) Signaling
20. C Reactive Protein (CRP)
21. D Dimer
22. Cytokine Storm
23. Predictive Factors and High-Risk Case of Cytokine Strom
24. The Complications of Cytokine Storm
25. The Current Therapeutic Options for Cytokine Storm and Time to Intervene
26. Limitations of this Paper
27. Conclusions
28. Recommendation
29. Article Highlights
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Acknowledgments
Conflicts of Interest
References
- World Health Organization. Coronavirus Disease (COVID-2019) Situation Reports; WHO Press, World Health Organization: Geneva, Switzerland, 2021; Available online: https://www.who.int/emergencies/diseases/novel-coronavirus-2019/situation-reports (accessed on 25 November 2021).
- Abdool Karim, S.S.; de Oliveira, T. New SARS-CoV-2 Variants—Clinical, Public Health, and Vaccine Implications. N. Engl. J. Med. 2021, 384, 1866–1868. [Google Scholar] [CrossRef] [PubMed]
- World Health Organization. Classification of Omicron (B.1.1.529): SARS-CoV-2 Variant of Concern; WHO Press, World Health Organization: Geneva, Switzerland, 2021; Available online: https://www.who.int/news/item/26-11-2021-classification-of-omicron-(b.1.1.529)-sars-cov-2-variant-of-concern (accessed on 28 November 2021).
- Centers for Disease Control and Prevention. SARS-CoV-2 Variant Classifications and Definitions; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2021. Available online: https://www.cdc.gov/coronavirus/2019-ncov/variants/variant-info.html (accessed on 9 March 2022).
- Fan, E.; Beitler, J.R.; Brochard, L.; Calfee, C.S.; Ferguson, N.D.; Slutsky, A.S. COVID-19-associated acute respiratory distress syndrome: Is a different approach to management warranted? Lancet Respir. Med. 2020, 8, 816–821. [Google Scholar] [CrossRef]
- Bradley, B.T.; Maioli, H.; Johnston, R.; Chaudhry, I.; Fink, S.L.; Xu, H. Histopathology and ultrastructural findings of fatal COVID-19 infections in Washington State: A case series. Lancet 2020, 396, 320–332. [Google Scholar] [CrossRef]
- Struyf, T.; Deeks, J.J.; Dinnes, J.; Takwoingi, Y.; Davenport, C.; Leeflang, M.M. Signs and symptoms to determine if a patient presenting in primary care or hospital outpatient settings has COVID-19 disease. Cochrane Database Syst. Rev. 2020, 7, 13665. [Google Scholar]
- Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020, 395, 507–513. [Google Scholar] [CrossRef] [Green Version]
- Salamanna, F.; Maglio, M.; Landini, M.P.; Fini, M. Body Localization of ACE-2: On the Trail of the Keyhole of SARS-CoV-2. Front. Med. 2020, 7, 594495. [Google Scholar] [CrossRef]
- Gheblawi, M.; Wang, K.; Viveiros, A.; Nguyen, Q.; Zhong, J.C.; Turner, A.J. Angiotensin-Converting Enzyme 2: SARS-CoV-2 Receptor and Regulator of the Renin-Angiotensin System: Celebrating the 20th Anniversary of the Discovery of ACE2. Circ. Res. 2020, 126, 1456–1474. [Google Scholar] [CrossRef]
- Zhang, H.; Li, H.B.; Lyu, J.R.; Lei, X.M.; Li, W.; Wu, G. Specific ACE2 expression in small intestinal enterocytes may cause gastrointestinal symptoms and injury after 2019-nCoV infection. Int. J. Infect. Dis. 2020, 96, 19–24. [Google Scholar] [CrossRef]
- Zhou, P.; Yang, X.L.; Wang, X.G.; Hu, B.; Zhang, L.; Zhang, W. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [Green Version]
- Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef]
- Matsuyama, S.; Nao, N.; Shirato, K.; Kawase, M.; Saito, S.; Takayama, I. Enhanced isolation of SARS-CoV-2 by TMPRSS2-expressing cells. Proc. Natl. Acad. Sci. USA 2020, 117, 7001–7003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef] [PubMed]
- Stetson, D.B.; Medzhitov, R. Type I interferons in host defense. Immunity 2006, 25, 373–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ye, Q.; Wang, B.; Mao, J. The pathogenesis and treatment of the ‘Cytokine Storm’ in COVID-19. J. Infect. 2020, 80, 607–613. [Google Scholar] [CrossRef]
- Henderson, L.A.; Canna, S.W.; Schulert, G.S.; Volpi, S.; Lee, P.Y.; Kernan, K.F. On the Alert for Cytokine Storm: Immunopathology in COVID-19. Arthritis Rheumatol. 2020, 72, 1059–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mehta, P.; McAuley, D.F.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet 2020, 395, 1033–1034. [Google Scholar] [CrossRef]
- Tufan, A.; Avanoğlu Güler, A.; Matucci-Cerinic, M. COVID-19, immune system response, hyper inflammation and repurposing antirheumatic drugs. Turk. J. Med. Sci. 2020, 50, 620–632. [Google Scholar] [CrossRef]
- Mangalmurti, N.; Hunter, C.A. Cytokine Storms: Understanding COVID-19. Immunity 2020, 53, 19–25. [Google Scholar] [CrossRef]
- Yang, L.; Xie, X.; Tu, Z.; Fu, J.; Xu, D.; Zhou, Y. The signal pathways and treatment of cytokine storm in COVID-19. Signal Transduct. Target. Ther. 2021, 6, 255. [Google Scholar] [CrossRef]
- Davila, M.L.; Riviere, I.; Wang, X.; Bartido, S.; Park, J.; Curran, K. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci. Transl. Med. 2014, 6, 224ra25. [Google Scholar] [CrossRef] [Green Version]
- Grupp, S.A.; Kalos, M.; Barrett, D.; Aplenc, R.; Porter, D.L.; Rheingold, S.R. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 2013, 368, 1509–1518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- London, N.R.; Zhu, W.; Bozza, F.A.; Smith, M.C.; Greif, D.M.; Sorensen, L.K. Targeting Robo4-dependent Slit signaling to survive the cytokine storm in sepsis and influenza. Sci. Transl. Med. 2010, 2, 23ra19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Imus, P.H.; Blackford, A.L.; Bettinotti, M.; Luznik, L.; Fuchs, E.J.; Huff, C.A. Severe Cytokine Release Syndrome after Haploidentical Peripheral Blood Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2019, 25, 2431–2437. [Google Scholar] [CrossRef] [PubMed]
- Wadia, P.P.; Tambur, A.R. Yin and yang of cytokine regulation in solid organ graft rejection and tolerance. Clin. Lab. Med. 2008, 28, 469–479. [Google Scholar] [CrossRef] [PubMed]
- Kumar, V. Toll-like receptors in sepsis-associated cytokine storm and their endogenous negative regulators as future immunomodulatory targets. Int. Immunopharmacol. 2020, 89, 107087. [Google Scholar] [CrossRef]
- Chousterman, B.G.; Swirski, F.K.; Weber, G.F. Cytokine storm and sepsis disease pathogenesis. Semin. Immunopathol. 2017, 39, 517–528. [Google Scholar] [CrossRef]
- IQWiG. The Innate and Adaptive Immune Systems; InformedHealth.org [Internet]; Institute for Quality and Efficiency in Health Care (IQWiG): Cologne, Germany, 2006. Available online: https://www.ncbi.nlm.nih.gov/books/NBK279396/ (accessed on 9 March 2022).
- Marshall, J.S.; Warrington, R.; Watson, W.; Kim, H.L. An introduction to immunology and immunopathology. Allergy Asthma Clin. Immunol. 2018, 14 (Suppl. S2), 49. [Google Scholar] [CrossRef] [Green Version]
- Alberts, B.; Johnson, A.; Lewis, J. Molecular Biology of the Cell, 4th ed.; The Adaptive Immune System; Garland Science: New York, NY, USA, 2002; Chapter 24. Available online: https://www.ncbi.nlm.nih.gov/books/NBK21070 (accessed on 9 March 2022).
- Diamond, M.S.; Kanneganti, T.D. Innate immunity: The first line of defense against SARS-CoV-2. Nat. Immunol. 2022, 23, 165–176. [Google Scholar] [CrossRef]
- Kasuga, Y.; Zhu, B.; Jang, K.J.; Yoo, J.S. Innate immune sensing of coronavirus and viral evasion strategies. Exp. Mol. Med. 2021, 53, 723–736. [Google Scholar] [CrossRef]
- Kanneganti, T.D. Intracellular innate immune receptors: Life inside the cell. Immunol. Rev. 2020, 297, 5–12. [Google Scholar] [CrossRef]
- Iwasaki, A. A virological view of innate immune recognition. Annu. Rev. Microbiol. 2012, 66, 177–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kikkert, M. Innate Immune Evasion by Human Respiratory RNA Viruses. J. Innate Immun. 2020, 12, 4–20. [Google Scholar] [CrossRef]
- O’Connell, P.; Aldhamen, Y.A. Systemic innate and adaptive immune responses to SARS-CoV-2 as it relates to other coronaviruses. Hum. Vaccin. Immunother. 2020, 16, 2980–2991. [Google Scholar] [CrossRef] [PubMed]
- Boechat, J.L.; Chora, I.; Morais, A.; Delgado, L. The immune response to SARS-CoV-2 and COVID-19 immunopathology—Current perspectives. Pulmonology 2021, 27, 423–437. [Google Scholar] [CrossRef] [PubMed]
- Amarante-Mendes, G.P.; Adjemian, S.; Branco, L.M.; Zanetti, L.C.; Weinlich, R.; Bortoluci, K.R. Pattern Recognition Receptors and the Host Cell Death Molecular Machinery. Front. Immunol. 2018, 9, 2379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mogensen, T.H. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin. Microbiol. Rev. 2009, 22, 240–273. [Google Scholar] [CrossRef] [Green Version]
- Combadière, B. Immunité adaptative contre le virus SARS-CoV-2 (Adaptive immunity against SARS-CoV-2). Med. Sci. 2020, 36, 908–913. [Google Scholar] [CrossRef]
- Bonilla, F.A.; Oettgen, H.C. Adaptive immunity. J. Allergy Clin. Immunol. 2010, 125 (Suppl. S2), S33–S40. [Google Scholar] [CrossRef]
- Paust, S.; Senman, B.; von Andrian, U.H. Adaptive immune responses mediated by natural killer cells. Immunol. Rev. 2010, 235, 286–296. [Google Scholar] [CrossRef]
- Sette, A.; Crotty, S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 2021, 184, 861–880. [Google Scholar] [CrossRef]
- Patel, M.; Shahjin, F.; Cohen, J.D.; Hasan, M.; Machhi, J.; Chugh, H. The Immunopathobiology of SARS-CoV-2 Infection. FEMS Microbiol. Rev. 2021, 45, fuab035. [Google Scholar] [CrossRef] [PubMed]
- Shah, V.K.; Firmal, P.; Alam, A.; Ganguly, D.; Chattopadhyay, S. Overview of Immune Response During SARS-CoV-2 Infection: Lessons from the Past. Front. Immunol. 2020, 11, 1949. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Trincado, J.L.; Gomez-Perosanz, M.; Reche, P.A. Fundamentals and Methods for T- and B-Cell Epitope Prediction. J. Immunol. Res. 2017, 2017, 2680160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Costela-Ruiz, V.J.; Illescas-Montes, R.; Puerta-Puerta, J.M.; Ruiz, C.; Melguizo-Rodríguez, L. SARS-CoV-2 infection: The role of cytokines in COVID-19 disease. Cytokine Growth Factor Rev. 2020, 54, 62–75. [Google Scholar] [CrossRef] [PubMed]
- García, L.F. Immune Response, Inflammation, and the Clinical Spectrum of COVID-19. Front. Immunol. 2020, 11, 1441. [Google Scholar] [CrossRef] [PubMed]
- Liou, T.G.; Adler, F.R.; Cahill, B.C.; Cox, D.R.; Cox, J.E.; Grant, G.J. SARS-CoV-2 innate effector associations and viral load in early nasopharyngeal infection. Physiol. Rep. 2021, 9, e14761. [Google Scholar] [CrossRef] [PubMed]
- Min, Y.Q.; Huang, M.; Sun, X.; Deng, F.; Wang, H.; Ning, Y.J. Immune evasion of SARS-CoV-2 from interferon antiviral system. Comput. Struct. Biotechnol. J. 2021, 19, 4217–4225. [Google Scholar] [CrossRef]
- Bouayad, A. Innate immune evasion by SARS-CoV-2: Comparison with SARS-CoV. Rev. Med. Virol. 2020, 30, 1–9. [Google Scholar] [CrossRef]
- Merad, M.; Martin, J.C. Pathological inflammation in patients with COVID-19: A key role for monocytes and macrophages. Nat. Rev. Immunol. 2020, 20, 355–362. [Google Scholar] [CrossRef]
- Amor, S.; Fernández Blanco, L.; Baker, D. Innate immunity during SARS-CoV-2: Evasion strategies and activation trigger hypoxia and vascular damage. Clin. Exp. Immunol. 2020, 202, 193–209. [Google Scholar] [CrossRef]
- Zhou, R.; To, K.K.; Wong, Y.C.; Liu, L.; Zhou, B.; Li, X. Acute SARS-CoV-2 Infection Impairs Dendritic Cell and T Cell Responses. Immunity 2020, 53, 864–877.e5. [Google Scholar] [CrossRef] [PubMed]
- Martin-Gayo, E.; Yu, X.G. Role of Dendritic Cells in Natural Immune Control of HIV-1 Infection. Front. Immunol. 2019, 10, 1306. [Google Scholar] [CrossRef] [PubMed]
- Cai, G.; Du, M.; Bossé, Y.; Albrecht, H.; Qin, F.; Luo, X. Xiao, F. SARS-CoV-2 Impairs Dendritic Cells and Regulates DC-SIGN Gene Expression in Tissues. Int. J. Mol. Sci. 2021, 22, 9228. [Google Scholar] [CrossRef] [PubMed]
- Merad, M.; Blish, C.A.; Sallusto, F.; Iwasaki, A. The immunology and immunopathology of COVID-19. Science 2022, 375, 1122–1127. [Google Scholar] [CrossRef] [PubMed]
- Alipoor, S.D.; Mortaz, E.; Jamaati, H.; Tabarsi, P.; Bayram, H.; Varahram, M. COVID-19: Molecular and Cellular Response. Front. Cell. Infect. Microbiol. 2021, 11, 563085. [Google Scholar] [CrossRef]
- Wen, W.; Su, W.; Tang, H.; Le, W.; Zhang, X.; Zheng, Y. Immune cell profiling of COVID-19 patients in the recovery stage by single-cell sequencing. Cell Discov. 2020, 6, 31. [Google Scholar] [CrossRef]
- Rosales, C. Neutrophil: A Cell with Many Roles in Inflammation or Several Cell Types? Front. Physiol. 2018, 9, 113. [Google Scholar] [CrossRef]
- Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef]
- Chen, Y.M.; Zheng, Y.; Yu, Y.; Wang, Y.; Huang, Q.; Qian, F. Blood molecular markers associated with COVID-19 immunopathology and multi-organ damage. EMBO J. 2020, 39, e105896. [Google Scholar] [CrossRef]
- Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J. Clinical Characteristics of 138 Hospitalized Patients with 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA 2020, 323, 1061–1069. [Google Scholar] [CrossRef]
- Wilk, A.J.; Rustagi, A.; Zhao, N.Q.; Roque, J.; Martínez-Colón, G.J.; McKechnie, J.L. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 2020, 26, 1070–1076. [Google Scholar] [CrossRef] [PubMed]
- Ronit, A.; Berg, R.M.G.; Bay, J.T.; Haugaard, A.K.; Ahlström, M.G.; Burgdorf, K.S. Compartmental immunophenotyping in COVID-19 ARDS: A case series. J. Allergy Clin. Immunol. 2021, 147, 81–91. [Google Scholar] [CrossRef] [PubMed]
- Schulte-Schrepping, J.; Reusch, N.; Paclik, D.; Baßler, K.; Schlickeiser, S.; Zhang, B. Severe COVID-19 Is Marked by a Dysregulated Myeloid Cell Compartment. Cell 2020, 182, 1419–1440.e23. [Google Scholar] [CrossRef] [PubMed]
- Palsson-McDermott, E.M.; Curtis, A.M.; Goel, G.; Lauterbach, M.A.R.; Sheedy, F.J.; Gleeson, L.E. Pyruvate Kinase M2 Regulates Hif-1α Activity and IL-1β Induction. It Is a Critical Determinant of the Warburg Effect in LPS-Activated Macrophages. Cell Metab. 2015, 21, 347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McElvaney, O.J.; McEvoy, N.L.; McElvaney, O.F.; Carroll, T.P.; Murphy, M.P.; Dunlea, D.M.; Ní Choileáin, O. Characterization of the Inflammatory Response to Severe COVID-19 Illness. Am. J. Respir. Crit. Care Med. 2020, 202, 812–821. [Google Scholar] [CrossRef]
- Jenne, C.N.; Wong, C.H.; Zemp, F.J.; McDonald, B.; Rahman, M.M.; Forsyth, P.A. Neutrophils recruited to sites of infection protect from virus challenge by releasing neutrophil extracellular traps. Cell Host Microbe 2013, 13, 169–180. [Google Scholar] [CrossRef] [Green Version]
- Grabcanovic-Musija, F.; Obermayer, A.; Stoiber, W.; Krautgartner, W.D.; Steinbacher, P.; Winterberg, N. Neutrophil extracellular trap (NET) formation characterizes stable and exacerbated COPD and correlates with airflow limitation. Respir. Res. 2015, 16, 59. [Google Scholar] [CrossRef] [Green Version]
- Mikacenic, C.; Moore, R.; Dmyterko, V.; West, T.E.; Altemeier, W.A.; Liles, W.C. Neutrophil extracellular traps (NETs) are increased in the alveolar spaces of patients with ventilator-associated pneumonia. Crit. Care 2018, 22, 358. [Google Scholar] [CrossRef] [Green Version]
- Bendib, I.; de Chaisemartin, L.; Granger, V.; Schlemmer, F.; Maitre, B.; Hüe, S. Neutrophil Extracellular Traps Are Elevated in Patients with Pneumonia-related Acute Respiratory Distress Syndrome. Anesthesiology 2019, 130, 581–591. [Google Scholar] [CrossRef]
- Guan, W.J.; Ni, Z.Y.; Hu, Y.; Liang, W.H.; Ou, C.Q.; He, J.X. Clinical Characteristics of Coronavirus Disease 2019 in China. N. Engl. J. Med. 2020, 382, 1708–1720. [Google Scholar] [CrossRef]
- Veras, F.P.; Pontelli, M.C.; Silva, C.M.; Toller-Kawahisa, J.E.; de Lima, M.; Nascimento, D.C. SARS-CoV-2-triggered neutrophil extracellular traps mediate COVID-19 pathology. J. Exp. Med. 2020, 217, e20201129. [Google Scholar] [CrossRef] [PubMed]
- Zuo, Y.; Yalavarthi, S.; Shi, H.; Gockman, K.; Zuo, M.; Madison, J.A. Neutrophil extracellular traps in COVID-19. JCI Insight 2020, 5, e138999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muraro, S.P.; De Souza, G.F.; Gallo, S.W.; Da Silva, B.K.; De Oliveira, S.D.; Vinolo, M.A.R. Respiratory Syncytial Virus induces the classical ROS-dependent NETosis through PAD-4 and necroptosis pathways activation. Sci. Rep. 2018, 8, 14166. [Google Scholar] [CrossRef] [PubMed]
- Cheng, O.Z.; Palaniyar, N. NET balancing: A problem in inflammatory lung diseases. Front. Immunol. 2013, 4, 1. [Google Scholar] [CrossRef] [Green Version]
- Borges, L.; Pithon-Curi, T.C.; Curi, R.; Hatanaka, E. COVID-19, and Neutrophils: The Relationship between Hyperinflammation and Neutrophil Extracellular Traps. Mediat. Inflamm. 2020, 2020, 8829674. [Google Scholar] [CrossRef]
- Qin, C.; Zhou, L.; Hu, Z.; Zhang, S.; Yang, S.; Tao, Y. Dysregulation of Immune Response in Patients with Coronavirus 2019 (COVID-19) in Wuhan, China. Clin. Infect. Dis. 2020, 71, 762–768. [Google Scholar] [CrossRef]
- Wang, F.; Nie, J.; Wang, H.; Zhao, Q.; Xiong, Y.; Deng, L. Characteristics of Peripheral Lymphocyte Subset Alteration in COVID-19 Pneumonia. J. Infect. Dis. 2020, 221, 1762–1769. [Google Scholar] [CrossRef] [Green Version]
- Wang, K.; Chen, W.; Zhang, Z.; Deng, Y.; Lian, J.Q.; Du, P. CD147-spike protein is a novel route for SARS-CoV-2 infection to host cells. Signal Transduct. Target. Ther. 2020, 5, 283. [Google Scholar] [CrossRef]
- Hadjadj, J.; Yatim, N.; Barnabei, L.; Corneau, A.; Boussier, J.; Smith, N. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 2020, 369, 718–724. [Google Scholar] [CrossRef]
- Fang, L.; Karakiulakis, G.; Roth, M. Are patients with hypertension and diabetes mellitus at increased risk for COVID-19 infection? Lancet Respir. Med. 2020, 8, e21. [Google Scholar] [CrossRef]
- Diao, B.; Wang, C.; Tan, Y.; Chen, X.; Liu, Y.; Ning, L. Reduction and Functional Exhaustion of T Cells in Patients with Coronavirus Disease 2019 (COVID-19). Front. Immunol. 2020, 11, 827. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.J.; Dong, X.; Cao, Y.Y.; Yuan, Y.D.; Yang, Y.B.; Yan, Y.Q. Clinical characteristics of 140 patients infected with SARS-CoV-2 in Wuhan, China. Allergy 2020, 75, 1730–1741. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Liu, S.; Liu, J.; Zhang, Z.; Wan, X.; Huang, B. COVID-19: Immunopathogenesis and Immunotherapeutics. Signal Transduct. Target. Ther. 2020, 5, 128. [Google Scholar] [CrossRef] [PubMed]
- Zheng, H.; Li, H.; Guo, L.; Liang, Y.; Li, J.; Wang, X. Virulence and pathogenesis of SARS-CoV-2 infection in rhesus macaques: A nonhuman primate model of COVID-19 progression. PLoS Pathog. 2020, 16, e1008949. [Google Scholar] [CrossRef]
- Wang, X.; Xu, W.; Hu, G.; Xia, S.; Sun, Z.; Liu, Z. Retraction Note to: SARS-CoV-2 infects T lymphocytes through its spike protein-mediated membrane fusion. Cell. Mol. Immunol. 2020, 17, 894. [Google Scholar] [CrossRef]
- Yue, Y.; Nabar, N.R.; Shi, C.S.; Kamenyeva, O.; Xiao, X.; Hwang, I.Y. SARS-Coronavirus Open Reading Frame-3a drives multimodal necrotic cell death. Cell Death Dis. 2018, 9, 904. [Google Scholar] [CrossRef]
- Tan, Y.X.; Tan, T.H.; Lee, M.J.; Tham, P.Y.; Gunalan, V.; Druce, J. Induction of apoptosis by the severe acute respiratory syndrome coronavirus 7a protein is dependent on its interaction with the Bcl-XL protein. J. Virol. 2007, 81, 6346–6355. [Google Scholar] [CrossRef] [Green Version]
- Tan, L.; Wang, Q.; Zhang, D.; Ding, J.; Huang, Q.; Tang, Y.Q. Lymphopenia predicts disease severity of COVID-19: A descriptive and predictive study. Signal Transduct. Target. Ther. 2020, 5, 33. [Google Scholar] [CrossRef]
- Cao, X. COVID-19: Immunopathology and its implications for therapy. Nat. Rev. Immunol. 2020, 20, 269–270. [Google Scholar] [CrossRef] [Green Version]
- Li, D.; Chen, Y.; Liu, H.; Jia, Y.; Li, F.; Wang, W. Immune dysfunction leads to mortality and organ injury in patients with COVID-19 in China: Insights from ERS-COVID-19 study. Signal Transduct. Target. Ther. 2020, 5, 62. [Google Scholar] [CrossRef]
- Sadarangani, M.; Marchant, A.; Kollmann, T.R. Immunological mechanisms of vaccine-induced protection against COVID-19 in humans. Nat. Rev. Immunol. 2021, 21, 475–484. [Google Scholar] [CrossRef] [PubMed]
- Quast, I.; Tarlinton, D. B cell memory: Understanding COVID-19. Immunity 2021, 54, 205–210. [Google Scholar] [CrossRef] [PubMed]
- Nicholson, L.B. The immune system. Essays Biochem. 2016, 60, 275–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palm, A.E.; Henry, C. Remembrance of Things Past: Long-Term B Cell Memory After Infection and Vaccination. Front. Immunol. 2019, 10, 1787. [Google Scholar] [CrossRef] [Green Version]
- Shiakolas, A.R.; Kramer, K.J.; Johnson, N.V.; Wall, S.C.; Suryadevara, N.; Wrapp, D. Efficient discovery of SARS-CoV-2-neutralizing antibodies via B cell receptor sequencing and ligand blocking. Nat. Biotechnol. 2022. [CrossRef]
- Gaebler, C.; Wang, Z.; Lorenzi, J.C.C.; Muecksch, F.; Finkin, S.; Tokuyama, M. Evolution of antibody immunity to SARS-CoV-2. Nature 2021, 591, 639–644. [Google Scholar] [CrossRef]
- Turner, J.S.; Kim, W.; Kalaidina, E.; Goss, C.W.; Rauseo, A.M.; Schmitz, A.J. SARS-CoV-2 infection induces long-lived bone marrow plasma cells in humans. Nature 2021, 595, 421–425. [Google Scholar] [CrossRef]
- Kojima, N.; Klausner, J.D. Protective immunity after recovery from SARS-CoV-2 infection. Lancet Infect. Dis. 2022, 22, 12–14. [Google Scholar] [CrossRef]
- Çölkesen, F.; Kepenek Kurt, E.; Vatansev, H.; Korkmaz, C.; Çölkesen, F.; Yücel, F. Memory B cells and serum immunoglobulins are associated with disease severity and mortality in patients with COVID-19. Postgrad. Med. J. 2022. [Google Scholar] [CrossRef]
- Vo, H.T.M.; Maestri, A.; Auerswald, H.; Sorn, S.; Lay, S.; Seng, H. Robust and Functional Immune Memory Up to 9 Months After SARS-CoV-2 Infection: A Southeast Asian Longitudinal Cohort. Front. Immunol. 2022, 13, 817905. [Google Scholar] [CrossRef]
- Zhao, J.; Yuan, Q.; Wang, H.; Liu, W.; Liao, X.; Su, Y. Antibody Responses to SARS-CoV-2 in Patients with Novel Coronavirus Disease 2019. Clin. Infect. Dis. 2020, 71, 2027–2034. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Zhou, X.; Zhu, C.; Song, Y.; Feng, F.; Qiu, Y. Immune Phenotyping Based on the Neutrophil-to-Lymphocyte Ratio and IgG Level Predicts Disease Severity and Outcome for Patients with COVID-19. Front. Mol. Biosci. 2020, 7, 157. [Google Scholar] [CrossRef] [PubMed]
- Blundell, P.A.; Le, N.P.L.; Allen, J.; Watanabe, Y.; Pleass, R.J. Engineering the fragment crystallizable (Fc) region of human IgG1 multimers and monomers to fine-tune interactions with sialic acid-dependent receptors. J. Biol. Chem. 2017, 292, 12994–13007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Keeler, S.P.; Fox, J.M. Requirement of Fc-Fc Gamma Receptor Interaction for Antibody-Based Protection against Emerging Virus Infections. Viruses 2021, 13, 1037. [Google Scholar] [CrossRef] [PubMed]
- Ben Mkaddem, S.; Benhamou, M.; Monteiro, R.C. Understanding Fc Receptor Involvement in Inflammatory Diseases: From Mechanisms to New Therapeutic Tools. Front. Immunol. 2019, 10, 811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koenderman, L. Inside-Out Control of Fc-Receptors. Front. Immunol. 2019, 10, 544. [Google Scholar] [CrossRef]
- Smyth, M.J.; Cretney, E.; Kelly, J.M.; Westwood, J.A.; Street, S.E.; Yagita, H. Activation of NK cell cytotoxicity. Mol. Immunol. 2005, 42, 501–510. [Google Scholar] [CrossRef]
- Van Erp, E.A.; Luytjes, W.; Ferwerda, G.; van Kasteren, P.B. Fc-Mediated Antibody Effector Functions During Respiratory Syncytial Virus Infection and Disease. Front. Immunol. 2019, 10, 548. [Google Scholar] [CrossRef] [Green Version]
- Tay, M.Z.; Wiehe, K.; Pollara, J. Antibody-Dependent Cellular Phagocytosis in Antiviral Immune Responses. Front. Immunol. 2019, 10, 332. [Google Scholar] [CrossRef]
- Adeniji, O.S.; Giron, L.B.; Purwar, M.; Zilberstein, N.F.; Kulkarni, A.J.; Shaikh, M.W. COVID-19 Severity Is Associated with Differential Antibody Fc-Mediated Innate Immune Functions. MBio 2021, 12, e00281-21. [Google Scholar] [CrossRef]
- Chan, C.E.Z.; Seah, S.G.K.; Chye, H.; Massey, S.; Torres, M.; Lim, A.P.C. The Fc-mediated effector functions of a potent SARS-CoV-2 neutralizing antibody, SC31, isolated from an early convalescent COVID-19 patient, are essential for the optimal therapeutic efficacy of the antibody. PLoS ONE 2021, 16, e0253487. [Google Scholar] [CrossRef] [PubMed]
- Bournazos, S.; Gupta, A.; Ravetch, J.V. The role of IgG Fc receptors in antibody-dependent enhancement. Nat. Rev. Immunol. 2020, 20, 633–643. [Google Scholar] [CrossRef] [PubMed]
- Wan, Y.; Shang, J.; Sun, S.; Tai, W.; Chen, J.; Geng, Q. Molecular Mechanism for Antibody-Dependent Enhancement of Coronavirus Entry. J. Virol. 2020, 94, e02015-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nimmerjahn, F.; Ravetch, J.V. Fc-receptors as regulators of immunity. Adv. Immunol. 2007, 96, 179–204. [Google Scholar] [CrossRef]
- Sánchez-Zuno, G.A.; Matuz-Flores, M.G.; González-Estevez, G.; Nicoletti, F.; Turrubiates-Hernández, F.J.; Mangano, K. A review: Antibody-dependent enhancement in COVID-19: The not so friendly side of antibodies. Int. J. Immunopathol. Pharmacol. 2021, 35, 20587384211050199. [Google Scholar] [CrossRef]
- Narasimhan, P.B.; Marcovecchio, P.; Hamers, A.A.J.; Hedrick, C.C. Nonclassical Monocytes in Health and Disease. Annu. Rev. Immunol. 2019, 37, 439–456. [Google Scholar] [CrossRef]
- Xu, G.; Qi, F.; Li, H.; Yang, Q.; Wang, H.; Wang, X. The differential immune responses to COVID-19 in peripheral and lung revealed by single-cell RNA sequencing. Cell Discov. 2020, 6, 73. [Google Scholar] [CrossRef]
- Honda, K.; Takaoka, A.; Taniguchi, T. Type I interferon [corrected] gene induction by the interferon regulatory factor family of transcription factors. Immunity 2006, 25, 349–360. [Google Scholar] [CrossRef] [Green Version]
- Jensen, S.; Thomsen, A.R. Sensing of RNA viruses: A review of innate immune receptors involved in recognizing RNA virus invasion. J. Virol. 2012, 86, 2900–2910. [Google Scholar] [CrossRef] [Green Version]
- Park, A.; Iwasaki, A. Type I and Type III Interferons—Induction, Signaling, Evasion, and Application to Combat COVID-19. Cell Host Microbe 2020, 27, 870–878. [Google Scholar] [CrossRef]
- Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Møller, R. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef] [PubMed]
- Galani, I.E.; Rovina, N.; Lampropoulou, V.; Triantafyllia, V.; Manioudaki, M.; Pavlos, E. Untuned antiviral immunity in COVID-19 revealed by temporal type I/III interferon patterns and flu comparison. Nat. Immunol. 2021, 22, 32–40. [Google Scholar] [CrossRef] [PubMed]
- Kopecky-Bromberg, S.A.; Martínez-Sobrido, L.; Frieman, M.; Baric, R.A.; Palese, P. Severe acute respiratory syndrome coronavirus open reading frame (ORF) 3b, ORF 6, and nucleocapsid proteins function as interferon antagonists. J. Virol. 2007, 81, 548–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Siu, K.L.; Kok, K.H.; Ng, M.J.; Poon, V.K.M.; Yuen, K.Y.; Zheng, B.J. Severe acute respiratory syndrome coronavirus M protein inhibits type I interferon production by impeding the formation of TRAF3.TANK. TBK1/IKKepsilon complex. J. Biol. Chem. 2009, 284, 16202–16209. [Google Scholar] [CrossRef] [Green Version]
- Minakshi, R.; Padhan, K.; Rani, M.; Khan, N.; Ahmad, F.; Jameel, S. The SARS Coronavirus 3a protein causes endoplasmic reticulum stress and induces ligand-independent downregulation of the type 1 interferon receptor. PLoS ONE 2009, 4, e8342. [Google Scholar] [CrossRef]
- Hu, Y.; Li, W.; Gao, T.; Cui, Y.; Jin, Y.; Li, P. The Severe Acute Respiratory Syndrome Coronavirus Nucleocapsid Inhibits Type I Interferon Production by Interfering with TRIM25-Mediated RIG-I Ubiquitination. J. Virol. 2017, 91, e02143-16. [Google Scholar] [CrossRef] [Green Version]
- Webster, B.; Assil, S.; Dreux, M. Cell-Cell Sensing of Viral Infection by Plasmacytoid Dendritic Cells. J. Virol. 2016, 90, 10050–10053. [Google Scholar] [CrossRef] [Green Version]
- Cervantes-Barragan, L.; Lewis, K.L.; Firner, S.; Thiel, V.; Hugues, S.; Reith, W. Plasmacytoid dendritic cells control T-cell response to chronic viral infection. Proc. Natl. Acad. Sci. USA 2012, 109, 3012–3017. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Z.; Ren, L.; Zhang, L.; Zhong, J.; Xiao, Y.; Jia, Z. Heightened Innate Immune Responses in the Respiratory Tract of COVID-19 Patients. Cell Host Microbe 2020, 27, 883–890.e2. [Google Scholar] [CrossRef]
- Channappanavar, R.; Fehr, A.R.; Vijay, R.; Mack, M.; Zhao, J.; Meyerholz, D.K. Dysregulated Type I Interferon and Inflammatory Monocyte-Macrophage Responses Cause Lethal Pneumonia in SARS-CoV-Infected Mice. Cell Host Microbe 2016, 19, 181–193. [Google Scholar] [CrossRef] [Green Version]
- Channappanavar, R.; Fehr, A.R.; Zheng, J.; Wohlford-Lenane, C.; Abrahante, J.E.; Mack, M. IFN-I response timing relative to virus replication determines MERS coronavirus infection outcomes. J. Clin. Investig. 2019, 129, 3625–3639. [Google Scholar] [CrossRef] [PubMed]
- Ruan, Q.; Yang, K.; Wang, W.; Jiang, L.; Song, J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020, 46, 846–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Herold, T.; Jurinovic, V.; Arnreich, C.; Lipworth, B.J.; Hellmuth, J.C.; von Bergwelt-Baildon, M. Elevated levels of IL-6 and CRP predict the need for mechanical ventilation in COVID-19. J. Allergy Clin. Immunol. 2020, 146, 128–136.e4. [Google Scholar] [CrossRef]
- Laing, A.G.; Lorenc, A.; Del Molino Del Barrio, I.; Das, A.; Fish, M.; Monin, L. A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat. Med. 2020, 26, 1623–1635. [Google Scholar] [CrossRef] [PubMed]
- Price, C.C.; Altice, F.L.; Shyr, Y.; Koff, A.; Pischel, L.; Goshua, G. Tocilizumab Treatment for Cytokine Release Syndrome in Hospitalized Patients with Coronavirus Disease 2019: Survival and Clinical Outcomes. Chest 2020, 158, 1397–1408. [Google Scholar] [CrossRef] [PubMed]
- Giamarellos-Bourboulis, E.J.; Netea, M.G.; Rovina, N.; Akinosoglou, K.; Antoniadou, A.; Antonakos, N. Complex Immune Dysregulation in COVID-19 Patients with Severe Respiratory Failure. Cell Host Microbe 2020, 27, 992–1000.e3. [Google Scholar] [CrossRef]
- Hirano, T.; Murakami, M. COVID-19: A New Virus, but a Familiar Receptor and Cytokine Release Syndrome. Immunity 2020, 52, 731–733. [Google Scholar] [CrossRef]
- Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef]
- Tanaka, T.; Narazaki, M.; Kishimoto, T. Immunotherapeutic implications of IL-6 blockade for cytokine storm. Immunotherapy 2016, 8, 959–970. [Google Scholar] [CrossRef] [PubMed]
- Gulhar, R.; Ashraf, M.A.; Jialal, I. Physiology, Acute Phase Reactants. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
- Alonzi, T.; Maritano, D.; Gorgoni, B.; Rizzuto, G.; Libert, C.; Poli, V. Essential role of STAT3 in the control of the acute-phase response as revealed by inducible gene inactivation [correction of activation] in the liver. Mol. Cell. Biol. 2001, 21, 1621–1632. [Google Scholar] [CrossRef] [Green Version]
- McLoughlin, R.M.; Hurst, S.M.; Nowell, M.A.; Harris, D.A.; Horiuchi, S.; Morgan, L.W.; Wilkinson, T.S.; Yamamoto, N.; Topley, N.; Jones, S.A. Differential regulation of neutrophil-activating chemokines by IL-6 and its soluble receptor isoforms. J. Immunol. 2004, 172, 5676–5683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van der Meer, I.M.; de Maat, M.P.; Bots, M.L.; Breteler, M.M.; Meijer, J.; Kiliaan, A.J.; Hofman, A.; Witteman, J.C. Inflammatory mediators and cell adhesion molecules as indicators of severity of atherosclerosis: The Rotterdam Study. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 838–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiang, S.; Dong, N.G.; Liu, J.P.; Wang, Y.; Shi, J.W.; Wei, Z.J. Inhibitory effects of suppressor of cytokine signaling 3 on inflammatory cytokine expression and migration and proliferation of IL-6/IFN-γ-induced vascular smooth muscle cells. J. Huazhong Univ. Sci. Technol. Med. Sci. 2013, 33, 615–622. [Google Scholar] [CrossRef] [PubMed]
- Deshmane, S.L.; Kremlev, S.; Amini, S.; Sawaya, B.E. Monocyte chemoattractant protein-1 (MCP-1): An overview. J. Interferon Cytokine Res. 2009, 29, 313–326. [Google Scholar] [CrossRef]
- Moore, J.B.; June, C.H. Cytokine release syndrome in severe COVID-19. Science 2020, 368, 473–474. [Google Scholar] [CrossRef] [Green Version]
- Kuba, K.; Imai, Y.; Rao, S.; Gao, H.; Guo, F.; Guan, B. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat. Med. 2005, 11, 875–879. [Google Scholar] [CrossRef]
- Glowacka, I.; Bertram, S.; Herzog, P.; Pfefferle, S.; Steffen, I.; Muench, M.O. Differential downregulation of ACE2 by the spike proteins of severe acute respiratory syndrome coronavirus and human coronavirus NL63. J. Virol. 2010, 84, 1198–1205. [Google Scholar] [CrossRef] [Green Version]
- Schieffer, B.; Luchtefeld, M.; Braun, S.; Hilfiker, A.; Hilfiker-Kleiner, D.; Drexler, H. Role of NAD(P)H oxidase in angiotensin II-induced JAK/STAT signaling and cytokine induction. Circ. Res. 2000, 87, 1195–1201. [Google Scholar] [CrossRef]
- Recinos A 3rd LeJeune, W.S.; Sun, H.; Lee, C.Y.; Tieu, B.C. Angiotensin II induces IL-6 expression and the Jak-STAT3 pathway in aortic adventitia of LDL receptor-deficient mice. Atherosclerosis 2007, 194, 125–133. [Google Scholar] [CrossRef] [Green Version]
- Catanzaro, M.; Fagiani, F.; Racchi, M.; Corsini, E.; Govoni, S.; Lanni, C. Immune response in COVID-19: Addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2. Signal Transduct. Target. Ther. 2020, 5, 84. [Google Scholar] [CrossRef]
- Schluns, K.S.; Kieper, W.C.; Jameson, S.C.; Lefrançois, L. Interleukin-7 mediates the homeostasis of naïve and memory CD8 T cells in vivo. Nat. Immunol. 2000, 1, 426–432. [Google Scholar] [CrossRef] [PubMed]
- Seddon, B.; Tomlinson, P.; Zamoyska, R. Interleukin 7 and T cell receptor signals regulate homeostasis of CD4 memory cells. Nat. Immunol. 2003, 4, 680–686. [Google Scholar] [CrossRef] [PubMed]
- Fry, T.J.; Mackall, C.L. The many faces of IL-7: From lymphopoiesis to peripheral T cell maintenance. J. Immunol. 2005, 174, 6571–6576. [Google Scholar] [CrossRef] [PubMed]
- Tan, J.T.; Dudl, E.; LeRoy, E.; Murray, R.; Sprent, J.; Weinberg, K.I. IL-7 is critical for homeostatic proliferation and survival of naive T cells. Proc. Natl. Acad. Sci. USA 2001, 98, 8732–8737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
- Saraiva, M.; O’Garra, A. The regulation of IL-10 production by immune cells. Nat. Rev. Immunol. 2010, 10, 170–181. [Google Scholar] [CrossRef] [Green Version]
- Lu, L.; Zhang, H.; Dauphars, D.J.; He, Y.W. A Potential Role of Interleukin 10 in COVID-19 Pathogenesis. Trends Immunol. 2021, 42, 3–5. [Google Scholar] [CrossRef]
- Han, H.; Ma, Q.; Li, C.; Liu, R.; Zhao, L.; Wang, W. Profiling serum cytokines in COVID-19 patients reveals IL-6 and IL-10 are disease severity predictors. Emerg. Microbes Infect. 2020, 9, 1123–1130. [Google Scholar] [CrossRef]
- Zhao, Y.; Qin, L.; Zhang, P.; Li, K.; Liang, L.; Sun, J. Longitudinal COVID-19 profiling associates IL-1RA and IL-10 with disease severity and RANTES with mild disease. JCI Insight 2020, 5, e139834. [Google Scholar] [CrossRef]
- Vignali, D.A.; Kuchroo, V.K. IL-12 family cytokines: Immunological playmakers. Nat. Immunol. 2012, 13, 722–728. [Google Scholar] [CrossRef] [Green Version]
- O’Shea, J.J.; Paul, W.E. Regulation of T(H)1 differentiation--controlling the controllers. Nat. Immunol. 2002, 3, 506–508. [Google Scholar] [CrossRef] [PubMed]
- Rosenberg, S.A. IL-2: The first effective immunotherapy for human cancer. J. Immunol. 2014, 192, 5451–5458. [Google Scholar] [CrossRef] [PubMed]
- Fu, X.; Xu, M.; Song, Y.; Li, Y.; Zhang, H.; Zhang, J. Enhanced interaction between SEC2 mutant and TCR Vβ induces MHC II-independent activation of T cells via PKCθ/NF-κB and IL-2R/STAT5 signaling pathways. J. Biol. Chem. 2018, 293, 19771–19784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yuan, X.; Cheng, G.; Malek, T.R. The importance of regulatory T-cell heterogeneity in maintaining self-tolerance. Immunol. Rev. 2014, 259, 103–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, H.; Wang, W.; Yin, J.; Ouyang, Y.; Pang, L.; Feng, Y. The inhibition of IL-2/IL-2R gives rise to CD8+ T cell and lymphocyte decrease through JAK1-STAT5 in critical patients with COVID-19 pneumonia. Cell Death Dis. 2020, 11, 429. [Google Scholar] [CrossRef]
- Cua, D.J.; Tato, C.M. Innate IL-17-producing cells: The sentinels of the immune system. Nat. Rev. Immunol. 2010, 10, 479–489. [Google Scholar] [CrossRef]
- McInnes, I.B.; Mease, P.J.; Ritchlin, C.T.; Rahman, P.; Gottlieb, A.B.; Kirkham, B. Secukinumab sustains improvement in signs and symptoms of psoriatic arthritis: 2-year results from the phase 3 FUTURE 2 study. Rheumatology 2017, 56, 1993–2003. [Google Scholar] [CrossRef] [Green Version]
- Pacha, O.; Sallman, M.A.; Evans, S.E. COVID-19: A case for inhibiting IL-17? Nat. Rev. Immunol. 2020, 20, 345–346. [Google Scholar] [CrossRef]
- Asrani, P.; Hassan, M.I. SARS-CoV-2 mediated lung inflammatory responses in host: Targeting the cytokine storm for therapeutic interventions. Mol. Cell. Biochem. 2021, 476, 675–687. [Google Scholar] [CrossRef]
- Pasquereau, S.; Kumar, A.; Herbein, G. Targeting TNF, and TNF Receptor Pathway in HIV-1 Infection: From Immune Activation to Viral Reservoirs. Viruses 2017, 9, 64. [Google Scholar] [CrossRef]
- Filik, H.; Avan, A.A. Electrochemical immunosensors for the detection of cytokine tumor necrosis factor-alpha: A review. Talanta 2020, 211, 120758. [Google Scholar] [CrossRef] [PubMed]
- Aggarwal, B.B. Signaling pathways of the TNF superfamily: A double-edged sword. Nat. Rev. Immunol. 2003, 3, 745–756. [Google Scholar] [CrossRef] [PubMed]
- Vanden Berghe, T.; Linkermann, A.; Jouan-Lanhouet, S.; Walczak, H.; Vandenabeele, P. Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 2014, 15, 135–147. [Google Scholar] [CrossRef] [PubMed]
- Dutta, J.; Fan, Y.; Gupta, N.; Fan, G.; Gélinas, C. Current insights into the regulation of programmed cell death by NF-kappaB. Oncogene 2006, 25, 6800–6816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lucas, C.; Wong, P.; Klein, J.; Castro, T.B.R.; Silva, J.; Sundaram, M. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature 2020, 584, 463–469. [Google Scholar] [CrossRef] [PubMed]
- Caricchio, R.; Gallucci, M.; Dass, C.; Zhang, X.; Gallucci, S.; Fleece, D. Preliminary predictive criteria for COVID-19 cytokine storm. Ann. Rheum. Dis. 2021, 80, 88–95. [Google Scholar] [CrossRef]
- Masso-Silva, J.A.; Moshensky, A.; Lam, M.T.Y.; Odish, M.F.; Patel, A.; Xu, L. Increased Peripheral Blood Neutrophil Activation Phenotypes and Neutrophil Extracellular Trap Formation in Critically Ill Coronavirus Disease 2019 (COVID-19) Patients: A Case Series and Review of the Literature. Clin. Infect. Dis. 2022, 74, 479–489. [Google Scholar] [CrossRef]
- Luo, W.; Li, Y.X.; Jiang, L.J.; Chen, Q.; Wang, T.; Ye, D.W. Targeting JAK-STAT Signaling to Control Cytokine Release Syndrome in COVID-19. Trends Pharmacol. Sci. 2020, 41, 531–543. [Google Scholar] [CrossRef]
- Farrar Ragab, D.; Salah Eldin, H.; Taeimah, M.; Khattab, R.; Salem, R. The COVID-19 Cytokine Storm; What We Know So Far. Front. Immunol. 2020, 11, 1446. [Google Scholar] [CrossRef]
- Tisoncik, J.R.; Korth, M.J.; Simmons, C.P.; Farrar, J.; Martin, T.R.; Katze, M.G. Into the eye of the cytokine storm. Microbiol. Mol. Biol. Rev. 2012, 76, 16–32. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Liu, X.; Wu, S.; Chen, S.; Li, Y.; Nong, L. Definition and Risks of Cytokine Release Syndrome in 11 Critically Ill COVID-19 Patients with Pneumonia: Analysis of Disease Characteristics. J. Infect. Dis. 2020, 222, 1444–1451. [Google Scholar] [CrossRef] [PubMed]
- Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H. Clinical and immunological features of severe and moderate coronavirus disease 2019. J. Clin. Investig. 2020, 130, 2620–2629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ather, J.L.; Ckless, K.; Martin, R.; Foley, K.L.; Suratt, B.T.; Boyson, J.E. Serum amyloid A activates the NLRP3 inflammasome and promotes Th17 allergic asthma in mice. J. Immunol. 2011, 187, 64–73. [Google Scholar] [CrossRef] [PubMed]
- Mridha, A.R.; Wree, A.; Robertson, A.A.B.; Yeh, M.M.; Johnson, C.D.; Van Rooyen, D.M. NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. J. Hepatol. 2017, 66, 1037–1046. [Google Scholar] [CrossRef]
- He, L.; Ding, Y.; Zhang, Q.; Che, X.; He, Y.; Shen, H. Expression of elevated levels of pro-inflammatory cytokines in SARS-CoV-infected ACE2+ cells in SARS patients: Relation to the acute lung injury and pathogenesis of SARS. J. Pathol. 2006, 210, 288–297. [Google Scholar] [CrossRef]
- Alosaimi, B.; Hamed, M.E.; Naeem, A.; Alsharef, A.A.; AlQahtani, S.Y.; AlDosari, K.M. MERS-CoV infection is associated with downregulation of genes encoding Th1 and Th2 cytokines/chemokines and elevated inflammatory innate immune response in the lower respiratory tract. Cytokine 2020, 126, 154895. [Google Scholar] [CrossRef]
- Lau, S.K.P.; Lau, C.C.Y.; Chan, K.H.; Li, C.P.Y.; Chen, H.; Jin, D.Y. Delayed induction of pro-inflammatory cytokines and suppression of innate antiviral response by the novel Middle East respiratory syndrome coronavirus: Implications for pathogenesis and treatment. J. Gen. Virol. 2013, 94 Pt 12, 2679–2690. [Google Scholar] [CrossRef]
- Zhang, W.; Zhao, Y.; Zhang, F.; Wang, Q.; Li, T.; Liu, Z. The use of anti-inflammatory drugs in the treatment of people with severe coronavirus disease 2019 (COVID-19): The Perspectives of clinical immunologists from China. Clin. Immunol. 2020, 214, 108393. [Google Scholar] [CrossRef]
- Dostert, C.; Pétrilli, V.; Van Bruggen, R.; Steele, C.; Mossman, B.T.; Tschopp, J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 2008, 320, 674–677. [Google Scholar] [CrossRef] [Green Version]
- Cassel, S.L.; Eisenbarth, S.C.; Iyer, S.S.; Sadler, J.J.; Colegio, O.R.; Tephly, L.A. The Nalp3 inflammasome is essential for the development of silicosis. Proc. Natl. Acad. Sci. USA 2008, 105, 9035–9040. [Google Scholar] [CrossRef] [Green Version]
- Cruz, C.M.; Rinna, A.; Forman, H.J.; Ventura, A.L.; Persechini, P.M. ATP activates a reactive oxygen species-dependent oxidative stress response and secretion of pro-inflammatory cytokines in macrophages. J. Biol. Chem. 2007, 282, 2871–2879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Becher, B.; Tugues, S.; Greter, M. GM-CSF: From Growth Factor to Central Mediator of Tissue Inflammation. Immunity 2016, 45, 963–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guilliams, M.; De Kleer, I.; Henri, S.; Post, S.; Vanhoutte, L.; De Prijck, S. Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J. Exp. Med. 2013, 210, 1977–1992. [Google Scholar] [CrossRef] [Green Version]
- Hamilton, J.A. GM-CSF in inflammation. J. Exp. Med. 2020, 217, e20190945. [Google Scholar] [CrossRef] [Green Version]
- Jain, S.; Gautam, V.; Naseem, S. Acute-phase proteins: As diagnostic tool. J. Pharm. Bioallied Sci. 2011, 3, 118–127. [Google Scholar] [CrossRef] [PubMed]
- Yusof, Y.A.M.; Haque, M. Biomarkers for The Detection of SARS-CoV2-Virus and in the Prediction of COVID-19 Infection Severity. J. App. Pharm. Sci. 2022, 12, i–v. [Google Scholar] [CrossRef]
- Yang, M.; Chen, X.; Xu, Y. A Retrospective Study of the C-Reactive Protein to Lymphocyte Ratio and Disease Severity in 108 Patients with Early COVID-19 Pneumonia from January to March 2020 Wuhan, China. Med. Sci. Monit. 2020, 26, e926393. [Google Scholar] [CrossRef]
- Ali, A.; Noman, M.; Guo, Y.; Liu, X.; Zhang, R.; Zhou, J. Myoglobin and C-reactive protein are efficient and reliable early predictors of COVID-19 associated mortality. Sci. Rep. 2021, 11, 5975. [Google Scholar] [CrossRef]
- Noris, M.; Benigni, A.; Remuzzi, G. The case of complement activation in COVID-19 multiorgan impact. Kidney Int. 2020, 98, 314–322. [Google Scholar] [CrossRef]
- Luan, Y.Y.; Yin, C.H.; Yao, Y.M. Update Advances on C-Reactive Protein in COVID-19 and Other Viral Infections. Front. Immunol. 2021, 12, 720363. [Google Scholar] [CrossRef]
- Stringer, D.; Braude, P.; Myint, P.K.; Evans, L.; Collins, J.T.; Verduri, A. The role of C-reactive protein as a prognostic marker in COVID-19. Int. J. Epidemiol. 2021, 50, 420–429. [Google Scholar] [CrossRef] [PubMed]
- Xie, J.; Covassin, N.; Fan, Z.; Singh, P.; Gao, W.; Li, G. Association Between Hypoxemia and Mortality in Patients with COVID-19. Mayo Clin. Proc. 2020, 95, 1138–1147. [Google Scholar] [CrossRef]
- Wang, L. C-reactive protein levels in the early stage of COVID-19. Med. Mal. Infect. 2020, 50, 332–334. [Google Scholar] [CrossRef] [PubMed]
- Favresse, J.; Lippi, G.; Roy, P.M.; Chatelain, B.; Jacqmin, H.; Ten Cate, H. D-dimer: Preanalytical, analytical, post-analytical variables, and clinical applications. Crit. Rev. Clin. Lab. Sci. 2018, 55, 548–577. [Google Scholar] [CrossRef] [PubMed]
- Riley, R.S.; Gilbert, A.R.; Dalton, J.B.; Pai, S.; McPherson, R.A. Widely Used Types and Clinical Applications of D-Dimer Assay. Lab. Med. 2016, 47, 90–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poudel, A.; Poudel, Y.; Adhikari, A.; Aryal, B.B.; Dangol, D.; Bajracharya, T. D-dimer as a biomarker for assessment of COVID-19 prognosis: D-dimer levels on admission and its role in predicting disease outcome in hospitalized patients with COVID-19. PLoS ONE 2021, 16, e0256744. [Google Scholar] [CrossRef] [PubMed]
- Yao, Y.; Cao, J.; Wang, Q.; Shi, Q.; Liu, K.; Luo, Z. D-dimer as a biomarker for disease severity and mortality in COVID-19 patients: A case-control study. J. Intensive Care 2020, 8, 49. [Google Scholar] [CrossRef]
- Cui, S.; Chen, S.; Li, X.; Liu, S.; Wang, F. Prevalence of venous thromboembolism in patients with severe novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 1421–1424. [Google Scholar] [CrossRef]
- Azkur, A.K.; Akdis, M.; Azkur, D.; Sokolowska, M.; van de Veen, W.; Brüggen, M.C. Immune response to SARS-CoV-2 and mechanisms of immunopathological changes in COVID-19. Allergy 2020, 75, 1564–1581. [Google Scholar] [CrossRef]
- Debuc, B.; Smadja, D.M. Is COVID-19 a New Hematologic Disease? Stem Cell Rev. Rep. 2021, 17, 4–8. [Google Scholar] [CrossRef]
- Li, X.; Geng, M.; Peng, Y.; Meng, L.; Lu, S. Molecular immune pathogenesis and diagnosis of COVID-19. J. Pharm. Anal. 2020, 10, 102–108. [Google Scholar] [CrossRef] [PubMed]
- Wilson, M.P.; Jack, A.S. Coronavirus disease 2019 (COVID-19) in neurology and neurosurgery: A scoping review of the early literature. Clin. Neurol. Neurosurg. 2020, 193, 105866. [Google Scholar] [CrossRef] [PubMed]
- Nile, S.H.; Nile, A.; Qiu, J.; Li, L.; Jia, X.; Kai, G. COVID-19: Pathogenesis, cytokine storm, and therapeutic potential of interferons. Cytokine Growth Factor Rev. 2020, 53, 66–70. [Google Scholar] [CrossRef] [PubMed]
- Fajgenbaum, D.C.; June, C.H. Cytokine Storm. N. Engl. J. Med. 2020, 383, 2255–2273. [Google Scholar] [CrossRef]
- Khanmohammadi, S.; Rezaei, N. Role of Toll-like receptors in the pathogenesis of COVID-19. J. Med. Virol. 2021, 93, 2735–2739. [Google Scholar] [CrossRef]
- Trasino, S.E. A role for retinoids in the treatment of COVID-19? Clin. Exp. Pharmacol. Physiol. 2020, 47, 1765–1767. [Google Scholar] [CrossRef]
- Onomoto, K.; Onoguchi, K.; Yoneyama, M. Regulation of RIG-I-like receptor-mediated signaling: Interaction between host and viral factors. Cell. Mol. Immunol. 2021, 18, 539–555. [Google Scholar] [CrossRef]
- Liu, T.; Feng, M.; Wen, Z.; He, Y.; Lin, W.; Zhang, M. Comparison of the Characteristics of Cytokine Storm and Immune Response Induced by SARS-CoV, MERS-CoV, and SARS-CoV-2 Infections. J. Inflamm. Res. 2021, 14, 5475–5487. [Google Scholar] [CrossRef]
- Al-Samkari, H.; Berliner, N. Hemophagocytic Lymphohistiocytosis. Annu. Rev. Pathol. 2018, 13, 27–49. [Google Scholar] [CrossRef]
- Suntharalingam, G.; Perry, M.R.; Ward, S.; Brett, S.J.; Castello-Cortes, A.; Brunner, M.D. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N. Engl. J. Med. 2006, 355, 1018–1028. [Google Scholar] [CrossRef]
- Roberts, K.A.; Colley, L.; Agbaedeng, T.A.; Ellison-Hughes, G.M.; Ross, M.D. Vascular Manifestations of COVID-19—Thromboembolism and Microvascular Dysfunction. Front. Cardiovasc. Med. 2020, 7, 598400. [Google Scholar] [CrossRef] [PubMed]
- Joly, B.S.; Siguret, V.; Veyradier, A. Understanding pathophysiology of hemostasis disorders in critically ill patients with COVID-19. Intensive Care Med. 2020, 46, 1603–1606. [Google Scholar] [CrossRef] [PubMed]
- D’Elia, R.V.; Harrison, K.; Oyston, P.C.; Lukaszewski, R.A.; Clark, G.C. Targeting the “cytokine storm” for therapeutic benefit. Clin. Vaccine Immunol. 2013, 20, 319–327. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.Y.; Alvarez, C.A.; Cobb, B.A. Integration of IL-2 and IL-4 signals coordinates divergent regulatory T cell responses and drives therapeutic efficacy. Elife 2021, 10, e57417. [Google Scholar] [CrossRef]
- Chen, L.Y.C.; Hoiland, R.L.; Stukas, S.; Wellington, C.L.; Sekhon, M.S. Confronting the controversy: Interleukin-6 and the COVID-19 cytokine storm syndrome. Eur. Respir. J. 2020, 56, 2003006. [Google Scholar] [CrossRef]
- Cavalli, G.; Dinarello, C.A. Suppression of inflammation and acquired immunity by IL-37. Immunol. Rev. 2018, 281, 179–190. [Google Scholar] [CrossRef]
- Liu, Q.; Wang, R.S.; Qu, G.Q.; Wang, Y.Y.; Liu, P.; Zhu, Y.Z. Gross examination report of a COVID-19 death autopsy. Fa Yi Xue Za Zhi 2020, 36, 21–23. [Google Scholar] [CrossRef]
- Su, H.; Yang, M.; Wan, C.; Yi, L.X.; Tang, F.; Zhu, H.Y.; Nie, X.; Zhang, C. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int. 2020, 98, 219–227. [Google Scholar] [CrossRef]
- Wichmann, D.; Sperhake, J.P.; Lütgehetmann, M.; Steurer, S.; Edler, C.; Heinemann, A. Autopsy Findings and Venous Thromboembolism in Patients With COVID-19: A Prospective Cohort Study. Ann. Intern. Med. 2020, 173, 268–277. [Google Scholar] [CrossRef]
- Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020, 8, 420–422. [Google Scholar] [CrossRef]
- Da, B.L.; Mitchell, R.A.; Lee, B.T.; Perumalswami, P.; Im, G.Y.; Agarwal, R.; Schiano, T.D. Kinetic patterns of liver enzyme elevation with COVID-19 in the USA. Eur. J. Gastroenterol. Hepatol. 2020, 32, 1466–1469. [Google Scholar] [CrossRef] [PubMed]
- Kermali, M.; Khalsa, R.K.; Pillai, K.; Ismail, Z.; Harky, A. The role of biomarkers in diagnosis of COVID-19—A systematic review. Life Sci. 2020, 254, 117788. [Google Scholar] [CrossRef] [PubMed]
- Tobiczyk, E.; Winiarska, H.M.; Springer, D.; Wysocka, E.; Cofta, S. Clinical significance of basic laboratory parameters in predicting the use of various methods of oxygen supplementation in COVID-19. Adv. Respir. Med. 2022, 90, 77–85. [Google Scholar] [CrossRef]
- Yu, D.; Du, Q.; Yan, S.; Guo, X.G.; He, Y.; Zhu, G.; Zhao, K.; Ouyang, S. Liver injury in COVID-19: Clinical features and treatment management. Virol. J. 2021, 18, 121. [Google Scholar] [CrossRef]
- Lehmann, A.; Prosch, H.; Zehetmayer, S.; Gysan, M.R.; Bernitzky, D.; Vonbank, K.; Idzko, M.; Gompelmann, D. Impact of persistent D-dimer elevation following recovery from COVID-19. PLoS ONE 2021, 16, e0258351. [Google Scholar] [CrossRef]
- Thachil, J.; Cushman, M.; Srivastava, A. A proposal for staging COVID-19 coagulopathy. Res. Pract. Thromb. Haemost. 2020, 4, 731–736. [Google Scholar] [CrossRef]
- Bartziokas, K.; Kostikas, K. Lactate dehydrogenase, COVID-19 and mortality. Med. Clin. 2021, 156, 37. [Google Scholar] [CrossRef]
- Henry, B.M.; Aggarwal, G.; Wong, J.; Benoit, S.; Vikse, J.; Plebani, M.; Lippi, G. Lactate dehydrogenase levels predict coronavirus disease 2019 (COVID-19) severity and mortality: A pooled analysis. Am. J. Emerg. Med. 2020, 38, 1722–1726. [Google Scholar] [CrossRef]
- Szarpak, L.; Ruetzler, K.; Safiejko, K.; Hampel, M.; Pruc, M.; Kanczuga-Koda, L.; Filipiak, K.J.; Jaguszewski, M.J. Lactate dehydrogenase level as a COVID-19 severity marker. Am. J. Emerg. Med. 2021, 45, 638–639. [Google Scholar] [CrossRef]
- Abbasi, J. Researchers Investigate What COVID-19 Does to the Heart. JAMA 2021, 325, 808–811. [Google Scholar] [CrossRef]
- Majure, D.T.; Gruberg, L.; Saba, S.G.; Kvasnovsky, C.; Hirsch, J.S.; Jauhar, R. Northwell Health COVID-19 Research Consortium. Usefulness of Elevated Troponin to Predict Death in Patients with COVID-19 and Myocardial Injury. Am. J. Cardiol. 2021, 138, 100–106. [Google Scholar] [CrossRef] [PubMed]
- Tersalvi, G.; Vicenzi, M.; Calabretta, D.; Biasco, L.; Pedrazzini, G.; Winterton, D. Elevated Troponin in Patients with Coronavirus Disease 2019: Possible Mechanisms. J. Card. Fail. 2020, 26, 470–475. [Google Scholar] [CrossRef] [PubMed]
- Melo, A.K.G.; Milby, K.M.; Caparroz, A.L.M.A.; Pinto, A.C.P.N.; Santos, R.R.P.; Rocha, A.P.; Ferreira, G.A.; Souza, V.A.; Valadares, L.D.A.; Vieira, R.M.R.A. Biomarkers of cytokine storm as red flags for severe and fatal COVID-19 cases: A living systematic review and meta-analysis. PLoS ONE 2021, 16, e0253894. [Google Scholar] [CrossRef] [PubMed]
- Shcherbak, S.G.; Anisenkova, A.Y.; Mosenko, S.V.; Glotov, O.S.; Chernov, A.N.; Apalko, S.V. Basic Predictive Risk Factors for Cytokine Storms in COVID-19 Patients. Front. Immunol. 2021, 12, 745515. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Yin, L.; Patel, J.; Tang, L.; Huang, Y. The inflammatory markers of multisystem inflammatory syndrome in children (MIS-C) and adolescents associated with COVID-19: A meta-analysis. J. Med. Virol. 2021, 93, 4358–4369. [Google Scholar] [CrossRef] [PubMed]
- Avila-Nava, A.; Cortes-Telles, A.; Torres-Erazo, D.; López-Romero, S.; Chim Aké, R.; Gutiérrez Solis, A.L. Serum IL-6: A potential biomarker of mortality among SARS-CoV-2 infected patients in Mexico. Cytokine 2021, 143, 155543. [Google Scholar] [CrossRef]
- Pan, F.; Yang, L.; Li, Y.; Liang, B.; Li, L.; Ye, T. Factors associated with death outcome in patients with severe coronavirus disease-19 (COVID-19): A case-control study. Int. J. Med. Sci. 2020, 17, 1281–1292. [Google Scholar] [CrossRef]
- Zhang, J.; Hao, Y.; Ou, W.; Ming, F.; Liang, G.; Qian, Y. Serum interleukin-6 is an indicator for severity in 901 patients with SARS-CoV-2 infection: A cohort study. J. Transl. Med. 2020, 18, 406. [Google Scholar] [CrossRef]
- Gupta, A.; Marzook, H.; Ahmad, F. Comorbidities and clinical complications associated with SARS-CoV-2 infection: An overview. Clin. Exp. Med. 2022, 1–19. [Google Scholar] [CrossRef]
- Ejaz, H.; Alsrhani, A.; Zafar, A.; Javed, H.; Junaid, K.; Abdalla, A.E. COVID-19, and comorbidities: Deleterious impact on infected patients. J. Infect. Public Health 2020, 13, 1833–1839. [Google Scholar] [CrossRef]
- Siddiqi, Z.; Fatima, J.; Bhatt, D.; Shukla, V.; Malik, M.; Ashfaq, A. Prevalence of Comorbidities in Survivors and Non-Survivors of Severe COVID-19 at a Dedicated COVID Care Centre. J. Assoc. Physicians India 2022, 70, 11–12. [Google Scholar] [PubMed]
- Gasmi, A.; Peana, M.; Pivina, L.; Srinath, S.; Gasmi Benahmed, A.; Semenova, Y. Interrelations between COVID-19 and other disorders. Clin. Immunol. 2021, 224, 108651. [Google Scholar] [CrossRef] [PubMed]
- Ramatillah, D.L.; Gan, S.H.; Pratiwy, I.; Syed Sulaiman, S.A.; Jaber, A.A.S.; Jusnita, N. Impact of cytokine storm on severity of COVID-19 disease in a private hospital in West Jakarta prior to vaccination. PLoS ONE 2022, 17, e0262438. [Google Scholar] [CrossRef] [PubMed]
- Buford, T.W. Hypertension and aging. Ageing Res. Rev. 2016, 26, 96–111. [Google Scholar] [CrossRef] [Green Version]
- Kay, A.M.; Simpson, C.L.; Stewart, J.A., Jr. The Role of AGE/RAGE Signaling in Diabetes-Mediated Vascular Calcification. J. Diabetes Res. 2016, 2016, 6809703. [Google Scholar] [CrossRef] [Green Version]
- Yazdanpanah, F.; Hamblin, M.R.; Rezaei, N. The immune system and COVID-19: Friend or foe? Life Sci. 2020, 256, 117900. [Google Scholar] [CrossRef]
- Kunnumakkara, A.B.; Rana, V.; Parama, D.; Banik, K.; Girisa, S.; Henamayee, S.; Thakur, K.K.; Dutta, U.; Garodia, P.; Gupta, S.C. COVID-19, cytokines, inflammation, and spices: How are they related? Life Sci. 2021, 284, 119201. [Google Scholar] [CrossRef]
- Klok, F.A.; Kruip, M.J.H.A.; van der Meer, N.J.M.; Arbous, M.S.; Gommers, D.A.M.P.J.; Kant, K.M.; Kaptein, F.H.J.; van Paassen, J.; Stals, M.A.M.; Huisman, M.V. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb. Res. 2020, 191, 145–147. [Google Scholar] [CrossRef]
- Tang, N.; Bai, H.; Chen, X.; Gong, J.; Li, D.; Sun, Z. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J. Thromb. Haemost. 2020, 18, 1094–1099. [Google Scholar] [CrossRef]
- Llitjos, J.F.; Leclerc, M.; Chochois, C.; Monsallier, J.M.; Ramakers, M.; Auvray, M.; Merouani, K. High incidence of venous thromboembolic events in anticoagulated severe COVID-19 patients. J. Thromb. Haemost. 2020, 18, 1743–1746. [Google Scholar] [CrossRef]
- Abou-Ismail, M.Y.; Diamond, A.; Kapoor, S.; Arafah, Y.; Nayak, L. The hypercoagulable state in COVID-19: Incidence, pathophysiology, and management. Thromb. Res. 2020, 194, 101–115. [Google Scholar] [CrossRef] [PubMed]
- Merrill, J.T.; Erkan, D.; Winakur, J.; James, J.A. Emerging evidence of a COVID-19 thrombotic syndrome has treatment implications. Nat. Rev. Rheumatol. 2020, 16, 581–589. [Google Scholar] [CrossRef] [PubMed]
- Tang, N.; Li, D.; Wang, X.; Sun, Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 844–847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.; Liu, Z.; Wu, G.; Yi, M.; Chen, Y.; Li, K.; Xu, X.; Xiao, L.; Wu, Q.; Chen, J. D-Dimer as a Prognostic Indicator in Critically Ill Patients Hospitalized with COVID-19 in Leishenshan Hospital, Wuhan, China. Front. Pharmacol. 2020, 11, 600592. [Google Scholar] [CrossRef]
- Aires, R.B.; Soares, A.A.S.M.; Gomides, A.P.M.; Nicola, A.M.; Teixeira-Carvalho, A.; da Silva, D.L.M. Thromboelastometry demonstrates endogenous coagulation activation in nonsevere and severe COVID-19 patients and has applicability as a decision algorithm for intervention. PLoS One 2022, 17, e0262600. [Google Scholar] [CrossRef]
- Tay, M.Z.; Poh, C.M.; Rénia, L.; MacAry, P.A.; Ng, L.F.P. The trinity of COVID-19: Immunity, inflammation, and intervention. Nat. Rev. Immunol. 2020, 20, 363–374. [Google Scholar] [CrossRef]
- Pedersen, S.F.; Ho, Y.C. SARS-CoV-2: A storm is raging. J. Clin. Investig. 2020, 130, 2202–2205. [Google Scholar] [CrossRef]
- Magadum, A.; Kishore, R. Cardiovascular Manifestations of COVID-19 Infection. Cells 2020, 9, 2508. [Google Scholar] [CrossRef]
- Tavazzi, G.; Pellegrini, C.; Maurelli, M.; Belliato, M.; Sciutti, F.; Bottazzi, A.; Sepe, P.A.; Resasco, T.; Camporotondo, R.; Bruno, R. Myocardial localization of coronavirus in COVID-19 cardiogenic shock. Eur. J. Heart Fail. 2020, 22, 911–915. [Google Scholar] [CrossRef] [Green Version]
- Chen, T.; Wu, D.; Chen, H.; Yan, W.; Yang, D.; Chen, G.; Ma, K.; Xu, D.; Yu, H.; Wang, H. Clinical characteristics of 113 deceased patients with coronavirus disease 2019: Retrospective study. BMJ 2020, 368, m1091. [Google Scholar] [CrossRef] [Green Version]
- Yu, W.L.; Toh, H.S.; Liao, C.T.; Chang, W.T. Cardiovascular Complications of COVID-19 and Associated Concerns: A Review. Acta Cardiol. Sin. 2021, 37, 9–17. [Google Scholar] [CrossRef] [PubMed]
- Ahmadian, E.; Hosseiniyan Khatibi, S.M.; Razi Soofiyani, S.; Abediazar, S.; Shoja, M.M.; Ardalan, M.; Zununi Vahed, S. COVID-19, and kidney injury: Pathophysiology and molecular mechanisms. Rev. Med. Virol. 2021, 31, e2176. [Google Scholar] [CrossRef] [PubMed]
- Buonaguro, F.M.; Ascierto, P.A.; Morse, G.D.; Buonaguro, L.; Puzanov, I.; Tornesello, M.L.; Bréchot, C.; Gallo, R.C. COVID-19: Time for a paradigm change. Rev. Med. Virol. 2020, 30, e2134. [Google Scholar] [CrossRef] [PubMed]
- Li, X.Q.; Liu, H.; Meng, Y.; Yin, H.Y.; Gao, W.Y.; Yang, X.; Xu, D.S.; Cai, X.D.; Guan, Y.; Lerman, L.O. Critical roles of cytokine storm and secondary bacterial infection in acute kidney injury development in COVID-19: A multi-center retrospective cohort study. J. Med. Virol. 2021, 93, 6641–6652. [Google Scholar] [CrossRef]
- Rabb, H. Kidney diseases in the time of COVID-19: Major challenges to patient care. J. Clin. Investig. 2020, 130, 2749–2751. [Google Scholar] [CrossRef] [Green Version]
- Poyiadji, N.; Shahin, G.; Noujaim, D.; Stone, M.; Patel, S.; Griffith, B. COVID-19-associated Acute Hemorrhagic Necrotizing Encephalopathy: Imaging Features. Radiology 2020, 296, E119–E120. [Google Scholar] [CrossRef] [Green Version]
- Shehata, G.A.; Lord, K.C.; Grudzinski, M.C.; Elsayed, M.; Abdelnaby, R.; Elshabrawy, H.A. Neurological Complications of COVID-19: Underlying Mechanisms and Management. Int. J. Mol. Sci. 2021, 22, 4081. [Google Scholar] [CrossRef]
- Bertolini, A.; van de Peppel, I.P.; Bodewes, F.A.J.A.; Moshage, H.; Fantin, A.; Farinati, F. Abnormal Liver Function Tests in Patients with COVID-19: Relevance and Potential Pathogenesis. Hepatology 2020, 72, 1864–1872. [Google Scholar] [CrossRef]
- Feng, G.; Zheng, K.I.; Yan, Q.Q.; Rios, R.S.; Targher, G.; Byrne, C.D. COVID-19 and Liver Dysfunction: Current Insights and Emergent Therapeutic Strategies. J. Clin. Transl. Hepatol. 2020, 8, 18–24. [Google Scholar] [CrossRef] [Green Version]
- Kumar, M.P.; Mishra, S.; Jha, D.K.; Shukla, J.; Choudhury, A.; Mohindra, R. Coronavirus disease (COVID-19) and the liver: A comprehensive systematic review and meta-analysis. Hepatol. Int. 2020, 14, 711–722. [Google Scholar] [CrossRef]
- Mao, R.; Qiu, Y.; He, J.S.; Tan, J.Y.; Li, X.H.; Liang, J. Manifestations and prognosis of gastrointestinal and liver involvement in patients with COVID-19: A systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2020, 5, 667–678. [Google Scholar] [CrossRef]
- Anirvan, P.; Narain, S.; Hajizadeh, N.; Aloor, F.Z.; Singh, S.P.; Satapathy, S.K. Cytokine-induced liver injury in coronavirus disease-2019 (COVID-19): Untangling the knots. Eur. J. Gastroenterol. Hepatol. 2021, 33 (Suppl. S1), e42–e49. [Google Scholar] [CrossRef] [PubMed]
- Polak, S.B.; Van Gool, I.C.; Cohen, D.; von der Thüsen, J.H.; van Paassen, J. A systematic review of pathological findings in COVID-19: A pathophysiological timeline and possible mechanisms of disease progression. Mod. Pathol. 2020, 33, 2128–2138. [Google Scholar] [CrossRef] [PubMed]
- Boeckmans, J.; Rodrigues, R.M.; Demuyser, T.; Piérard, D.; Vanhaecke, T.; Rogiers, V. COVID-19, and drug-induced liver injury: A problem of plenty or a petty point? Arch. Toxicol. 2020, 94, 1367–1369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Licata, A.; Minissale, M.G.; Distefano, M.; Montalto, G. Liver injury, SARS-CoV-2 infection, and COVID-19: What physicians should really know? GastroHep 2021, 3, 121–130. [Google Scholar] [CrossRef]
- Vitiello, A.; La Porta, R.; D’Aiuto, V.; Ferrara, F. The risks of liver injury in COVID-19 patients and pharmacological management to reduce or prevent the damage induced. Egypt. Liver J. 2021, 11, 11. [Google Scholar] [CrossRef]
- Hu, K.; Patel, J.; Swiston, C. Ophthalmic Manifestations of Coronavirus (COVID-19). In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK556093/ (accessed on 9 March 2022).
- Nasiri, N.; Sharifi, H.; Bazrafshan, A.; Noori, A.; Karamouzian, M.; Sharifi, A. Ocular Manifestations of COVID-19: A Systematic Review and Meta-analysis. J. Ophthalmic Vis. Res. 2021, 16, 103–112. [Google Scholar] [CrossRef]
- Zhong, Y.; Wang, K.; Zhu, Y.; Lyu, D.; Yu, Y.; Li, S. Ocular manifestations in COVID-19 patients: A systematic review and meta-analysis. Travel Med. Infect. Dis. 2021, 44, 102191. [Google Scholar] [CrossRef]
- Bostanci Ceran, B.; Ozates, S. Ocular manifestations of coronavirus disease 2019. Graefes Arch. Clin. Exp. Ophthalmol. 2020, 258, 1959–1963. [Google Scholar] [CrossRef]
- Tang, Y.; Liu, J.; Zhang, D.; Xu, Z.; Ji, J.; Wen, C. Cytokine Storm in COVID-19: The Current Evidence and Treatment Strategies. Front. Immunol. 2020, 11, 1708. [Google Scholar] [CrossRef]
- Kaye, A.G.; Siegel, R. The efficacy of IL-6 inhibitor Tocilizumab in reducing severe COVID-19 mortality: A systematic review. PeerJ 2020, 8, e10322. [Google Scholar] [CrossRef] [PubMed]
- Boregowda, U.; Perisetti, A.; Nanjappa, A.; Gajendran, M.; Kutti Sridharan, G.; Goyal, H. Addition of Tocilizumab to the Standard of Care Reduces Mortality in Severe COVID-19: A Systematic Review and Meta-Analysis. Front. Med. 2020, 7, 586221. [Google Scholar] [CrossRef] [PubMed]
- Rubio-Rivas, M.; Forero, C.G.; Mora-Luján, J.M.; Montero, A.; Formiga, F.; Homs, N.A. Beneficial and harmful outcomes of tocilizumab in severe COVID-19: A systematic review and meta-analysis. Pharmacotherapy 2021, 41, 884–906. [Google Scholar] [CrossRef] [PubMed]
- Rubio-Rivas, M.; Ronda, M.; Padulles, A.; Mitjavila, F.; Riera-Mestre, A.; García-Forero, C. Beneficial effect of corticosteroids in preventing mortality in patients receiving tocilizumab to treat severe COVID-19 illness. Int. J. Infect. Dis. 2020, 101, 290–297. [Google Scholar] [CrossRef]
- Hashimoto, S.; Yoshizaki, K.; Uno, K.; Kitajima, H.; Arai, T.; Tamura, Y. Prompt Reduction in CRP, IL-6, IFN-γ, IP-10, and MCP-1 and a Relatively Low Basal Ratio of Ferritin/CRP Is Possibly Associated with the Efficacy of Tocilizumab Monotherapy in Severely to Critically Ill Patients with COVID-19. Front. Med. 2021, 8, 734838. [Google Scholar] [CrossRef]
- Kolilekas, L.; Loverdos, K.; Giannakaki, S.; Vlassi, L.; Levounets, A.; Zervas, E. Can steroids reverse the severe COVID-19 induced “cytokine storm”? J. Med. Virol. 2020, 92, 2866–2869. [Google Scholar] [CrossRef]
- Olbei, M.; Hautefort, I.; Modos, D.; Treveil, A.; Poletti, M.; Gul, L. SARS-CoV-2 Causes a Different Cytokine Response Compared to Other Cytokine Storm-Causing Respiratory Viruses in Severely Ill Patients. Front. Immunol. 2021, 12, 629193. [Google Scholar] [CrossRef]
- Chifu, I.; Detomas, M.; Dischinger, U.; Kimpel, O.; Megerle, F.; Hahner, S. Management of Patients with Glucocorticoid-Related Diseases and COVID-19. Front. Endocrinol. 2021, 12, 705214. [Google Scholar] [CrossRef]
- Tang, X.; Feng, Y.M.; Ni, J.X.; Zhang, J.Y.; Liu, L.M.; Hu, K. Early Use of Corticosteroid May Prolong SARS-CoV-2 Shedding in Non-Intensive Care Unit Patients with COVID-19 Pneumonia: A Multicenter, Single-Blind, Randomized Control Trial. Respiration 2021, 100, 116–126. [Google Scholar] [CrossRef]
- Ranjbar, K.; Moghadami, M.; Mirahmadizadeh, A.; Fallahi, M.J.; Khaloo, V.; Shahriarirad, R. Methylprednisolone or dexamethasone, which one is superior corticosteroid in the treatment of hospitalized COVID-19 patients: A triple-blinded randomized controlled trial. BMC Infect. Dis. 2021, 21, 337. [Google Scholar] [CrossRef]
- Isidori, A.M.; Arnaldi, G.; Boscaro, M.; Falorni, A.; Giordano, C.; Giordano, R. COVID-19 infection and glucocorticoids: Update from the Italian Society of Endocrinology Expert Opinion on steroid replacement in adrenal insufficiency. J. Endocrinol. Investig. 2020, 43, 1141–1147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aghbash, P.S.; Eslami, N.; Shamekh, A.; Entezari-Maleki, T.; Baghi, H.B. SARS-CoV-2 infection: The role of PD-1/PD-L1 and CTLA-4 axis. Life Sci. 2021, 270, 119124. [Google Scholar] [CrossRef] [PubMed]
- Nardelli, P.; Pieri, M.; Fominskiy, E.; Scandroglio, A.M. Cytokine adsorption and ECMO in patients with COVID-19. Lancet Respir. Med. 2021, 9, e71. [Google Scholar] [CrossRef]
- Rybalko, A.; Voronin, A.V.; Karpun, N.A. Cytokine adsorption and ECMO in patients with COVID-19. Lancet Respir. Med. 2021, 9, e69–e70. [Google Scholar] [CrossRef]
- Kohler, H.; Kaveri, S. How IvIg Can Mitigate COVID-19 Disease: A Symmetrical Immune Network Model. Monoclon. Antibodies Immunodiagn. Immunother. 2021, 40, 17–20. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Cao, W.; Li, T. High-Dose Intravenous Immunoglobulins in the Treatment of Severe Acute Viral Pneumonia: The Known Mechanisms and Clinical Effects. Front. Immunol. 2020, 11, 1660. [Google Scholar] [CrossRef] [PubMed]
- Zhao, M. Cytokine storm and immunomodulatory therapy in COVID-19: Role of chloroquine and anti-IL-6 monoclonal antibodies. Int. J. Antimicrob. Agents 2020, 55, 105982. [Google Scholar] [CrossRef]
- Gatti, E.; Piotto, M.; Lelii, M.; Pensabene, M.; Madini, B.; Cerrato, L. Therapeutic Strategies for COVID-19 Lung Disease in Children. Front. Pediatr. 2022, 10, 829521. [Google Scholar] [CrossRef]
- Li, Y.; Zhao, W.; Liu, J.; Chen, Z.; Lv, Q.; Zhang, Z. Immunotherapy Summary for Cytokine Storm in COVID-19. Front. Pharmacol. 2021, 12, 731847. [Google Scholar] [CrossRef]
- Conti, P.; Pregliasco, F.E.; Calvisi, V.; Caraffa Al Gallenga, C.E.; Kritas, S.K. Monoclonal antibody therapy in COVID-19. J. Biol. Regul. Homeost. Agents 2021, 35, 423–427. [Google Scholar] [CrossRef]
- Shi, C.; Wang, C.; Wang, H.; Yang, C.; Cai, F.; Zeng, F. The Potential of Low Molecular Weight Heparin to Mitigate Cytokine Storm in Severe COVID-19 Patients: A Retrospective Cohort Study. Clin. Transl. Sci. 2020, 13, 1087–1095. [Google Scholar] [CrossRef] [PubMed]
- Gozzo, L.; Viale, P.; Longo, L.; Vitale, D.C.; Drago, F. The Potential Role of Heparin in Patients with COVID-19: Beyond the Anticoagulant Effect. A Review. Front. Pharmacol. 2020, 11, 1307. [Google Scholar] [CrossRef] [PubMed]
- Drago, F.; Gozzo, L.; Li, L.; Stella, A.; Cosmi, B. Use of Enoxaparin to Counteract COVID-19 Infection and Reduce Thromboembolic Venous Complications: A Review of the Current Evidence. Front. Pharmacol. 2020, 11, 579886. [Google Scholar] [CrossRef] [PubMed]
- Sinha, P.; Matthay, M.A.; Calfee, C.S. Is a “Cytokine Storm” Relevant to COVID-19? JAMA Intern. Med. 2020, 180, 1152–1154. [Google Scholar] [CrossRef] [PubMed]
- Calfee, C.S.; Delucchi, K.; Parsons, P.E.; Thompson, B.T.; Ware, L.B.; Matthay, M.A. Subphenotypes in acute respiratory distress syndrome: Latent class analysis of data from two randomized controlled trials. Lancet Respir. Med. 2014, 2, 611–620. [Google Scholar] [CrossRef] [Green Version]
- Wong, C.K.H.; Wan, E.Y.F.; Luo, S.; Ding, Y.; Lau, E.H.Y.; Ling, P. Clinical outcomes of different therapeutic options for COVID-19 in two Chinese case cohorts: A propensity-score analysis. EClinicalMedicine 2021, 32, 100743. [Google Scholar] [CrossRef]
- Nold, M.F.; Nold-Petry, C.A.; Zepp, J.A.; Palmer, B.E.; Bufler, P.; Dinarello, C.A. IL-37 is a fundamental inhibitor of innate immunity. Nat. Immunol. 2010, 11, 1014–1022. [Google Scholar] [CrossRef] [Green Version]
- Banchereau, J.; Pascual, V.; O’Garra, A. From IL-2 to IL-37: The expanding spectrum of anti-inflammatory cytokines. Nat. Immunol. 2012, 13, 925–931. [Google Scholar] [CrossRef]
Reference | Study Population | Study Design | Study Period | Subgroup | Results |
---|---|---|---|---|---|
Masso-Silva et al. 2022 [183] | N = 31 | Case series | 11 days |
Case:16 Critically ill COVID-19 patients with APACHE(Acute Physiology and Chronic Health Evaluation) II scores of 7–27 on intensive care unit (ICU) admission. Control:15 healthy subjects | Plasma cytokine profiles and complete blood counts of COVID-19 patient demonstrated elevations in IL-8, IL-6, neutrophil:lymphocyte ratio (mean, 9.3). Profiling of specific cytokines relevant to neutrophil activity showed broad elevations across IP-10, IL-6, IL-8, granulocyte macrophage colony-stimulating factor (GM-CSF), interleukin 1β, interleukin 10, and tumor necrosis factor alpha (TNF-α) in the circulation of critically ill COVID-19 patients both early in their hospitalization and were remained raised throughout their hospitalization, measured at multiple time points |
Wang et al. 2020 [65] | N = 138 Age= 56 years(median age) | Case series | 1 month (1 January–3 February 2020) | 102 (73.9%) were admitted to isolation wards, and 36 (26.1%) were admitted and transferred to the ICU because of development of dysfunction of organ | Common symptoms included fever (136 [98.6%]), fatigue (96 [69.6%]), and dry cough (82 [59.4%]). Lymphopenia (lymphocyte count, 0.8 × 109/L [interquartile range 0.6–1.1]) in 97 patients (70.3%), Raised Neutrophil count in 36 ICU patients 4.6 (2.6–7.9) p = <0.001 and elevated lactate dehydrogenase (261 U/L [IQR, 182–403]) in 55 patients (39.9%). Chest computed tomographic scans revealed bilateral patchy shadows or ground glass opacity in the lungs of all patients |
Wilk et al. 2020 [66] | N = 13 age ≥18 years | Cross sectional study | 2–3 weeks | single-cell RNA sequencing (scRNA-seq) to profile peripheral blood mononuclear cells (PBMCs) was done. Case: 7 patients hospitalized for COVID-19, 4 of whom had acute respiratory distress syndrome Control: 6 healthy controls. | HLA class II downregulation was noted and a developing neutrophil population were observed that appears closely related to plasmablasts appearing in patients with acute respiratory failure requiring mechanical ventilation. |
Ronit et al. 2021 [67] | N = 4 Age = 40–75 years | Cross sectional study | 2months 21 days | SARS-CoV-2 patients confirmed by PCR, with presence of ARDS determined according to the Berlin criteria and less than 72 h of mechanical ventilation after admittance to the intensive care unit (ICU) | Immature neutrophils were raised in both blood and lungs, whereas CD4 and CD8 T-cell lymphopenia was observed in the 2 compartments. However, regulatory T cells and TH17 cells were found in higher fractions in the lung. Lung CD4 and CD8 T cells and macrophages expressed an even higher upregulation of activation markers than in blood. Cytokines were expressed at high levels both in the blood and in the lungs, most markedly, IL-1RA, IL-6, IL-8, IP-10, and monocyte chemoattactant protein-1, pointing to hyperinflammation. |
Wang et al. (2020) [82] | N = 60 Age = 60 years(medan) | Cross sectional study | 5 weeks | Levels of peripheral lymphocyte subsets were measured by flow cytometry in 60 hospitalized COVID-19 patients before and after treatment | Total lymphocytes, CD4+ T cells, CD8+ T cells, NK cells and B cells reduced in COVID-19 patients, and severe cases had a lower level than mild cases. Lymphocyte subsets showed a significant relation with inflammatory state in COVID-19, especially CD4+/CD8+ ratio and CD8+ T cells. Following treatment, clinical response was observed in 37 patients (67%), with an rise in CD8+ T cells and B cells |
Hadjadj et al. (2020) [84] | N = 68 | Cross sectional study | 10 days | COVID 19 patient = 50 Mild-moderate = 15 Severe = 17 Critical = 18 Healthy subjects = 18 | in severe and critical patients, there was highly impaired interferon (IFN) type I response (characterized by no IFN-β and low IFN-α production and activity), which was related with a persistent viral load in blood and hyperinflammatory response. Inflammation was characterized by increased tumor necrosis factor–α and interleukin-6 production and signaling. |
Herold et al. (2020) [138] | N= 89 | Cohort study | 5 weeks | initial evaluation cohort (n = 40) which was followed by a validation cohort that was separated temporally (n = 49) | CRP and IL 6 levels in the evaluation cohort were0.86 and 0.97, and they were similar in the validation cohort (0.83 and 0.90, respectively) |
Laing et al. (2020) [139] | N = 73; Age = 61 years(median) | Cross sectional study | 3 weeks | Patients with COVID-19 = 63 Control group not suffering from COVID-19 = 10 | patients exhibited considerable person to person in number of variation in B cell, ranging from overt cytopenia (<104 B cells ml−1) to atypically high counts (2–3 × 105 mL−1). IL-6 and IL-10 levels were also highly raised in COVID-19 and the rise were related to severity. |
Caricchio et al. (2021) [182] | N = 513; Age = 58.3 years | Cohort study | 5 weeks | 513 patients were enrolled in the cohort and considered eligible must have met the following criteria on hospital admission: (1) signs and symptoms of COVID-19 infection (fever, generalised malaise, cough and shortness of breath) up to 1 week prior to admission to hospital and (2) findings of ground-glass opacity (GGO) by high-resolution CT (HRCT) of the chest as per radiology reading | Elevated levels of IL6 was observed in most COVID 19 patients which was higher significantly in COVID-CS (35 vs. 96 pg/mL) confirming strong inflammation. The white blood cells, and particularly neutrophils and monocytes, were significantly increased in the COVID-CS group, suggesting innate immunity has a active role in Cytokine storm. The lymphocytes were decreased, on average half of the lower limit of normal value, indicating a functional depletion of the adaptive immunity |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ahmad, R.; Haque, M. Surviving the Storm: Cytokine Biosignature in SARS-CoV-2 Severity Prediction. Vaccines 2022, 10, 614. https://doi.org/10.3390/vaccines10040614
Ahmad R, Haque M. Surviving the Storm: Cytokine Biosignature in SARS-CoV-2 Severity Prediction. Vaccines. 2022; 10(4):614. https://doi.org/10.3390/vaccines10040614
Chicago/Turabian StyleAhmad, Rahnuma, and Mainul Haque. 2022. "Surviving the Storm: Cytokine Biosignature in SARS-CoV-2 Severity Prediction" Vaccines 10, no. 4: 614. https://doi.org/10.3390/vaccines10040614