Acessibilidade / Reportar erro

The seven constitutive respiratory defense barriers against SARS-CoV-2 infection

Abstract

Before eliciting an adaptive immune response, SARS-CoV-2 must overcome seven constitutive respiratory defense barriers. The first is the mucus covering the respiratory tract’s luminal surface, which entraps inhaled particles, including infectious agents, and eliminates them by mucociliary clearance. The second barrier comprises various components present in the airway lining fluid, the surfactants. Besides providing low surface tension that allows efficient gas exchange at the alveoli, surfactants inhibit the invasion of epithelial cells by respiratory viruses, enhance pathogen uptake by phagocytes, and regulate immune cells’ functions. The respiratory tract microbiota constitutes the third defense barrier against SARS-CoV-2. It activates the innate and adaptive immune cells and elicits anti-infectious molecules such as secretory IgA antibodies, defensins, and interferons. The fourth defense barrier comprises the antimicrobial peptides defensins, and lactoferrin. They show direct antiviral activity, inhibit viral fusion, and modulate the innate and adaptive immune responses. Secretory IgA antibodies, the fifth defense barrier, besides protecting the local microbiota against noxious agents, also inhibit SARS-CoV-2 cell invasion. If the virus overcomes this barrier, it reaches its target, the respiratory epithelial cells. However, these cells also act as a defense barrier, the sixth one, since they hinder the virus’ access to receptors and produce antiviral and immunomodulatory molecules such as interferons, lactoferrin, and defensins. Finally, the sensing of the virus by the cells of innate immunity, the last constitutive defense barrier, elicits a cascade of signals that activate adaptive immune cells and may inhibit the development of productive infection. The subject of the present essay is discussing these mechanisms.

Keywords:
Respiratory tract microbiota; Defensins and lactoferrin; Surfactant proteins; Epithelial cell interferons; Innate immunity

INTRODUCTION

SARS-CoV-2 needs to bind, enter, and replicate into the respiratory tract’s epithelial cells to originate a productive pulmonary infection. However, to reach these cells, the virus must overcome several constitutive defensive barriers, such as the mucus and mucociliary clearance, surfactant proteins, the respiratory tract microbiota, antimicrobial peptides, and secretory IgA antibodies. Furthermore, the respiratory epithelial cells have mechanisms to counter virus infection, which are amplified by two other defense barriers, the innate and the adaptive immune systems. This review pinpoints how the seven constitutive respiratory defense barriers act to hinder SARS-CoV-2 infection.

The 1 st barrier: Mucus and mucociliary clearance

The airway mucus constitutes a thin layer of a dense, gel-like material covering the respiratory tract’s luminal surface. The mucus’s primary function is to protect the lungs through the mucociliary clearance of inhaled foreign particles, including infectious agents and noxious pollutants. Mucins comprise the mucus’ major protein component and are present as secreted and cell-associated glycoproteins11. Lillehoj EP, Kato K, Lu W, Kim KC. Cellular and molecular biology of airway mucins. Int Rev Cell Mol Biol. 2013;303:139-202.. The five main mucins, out of the 15 characterized in the human respiratory tract, are distributed in two distinct yet interacting layers. The outer or mucus layer contains two gel-forming mucins (MUC5AC and MUC5B) tightly associated with various defensive molecules. The inner or periciliary liquid layer contains three membrane-tethered mucins (MUC1, MUC4, and MUC16) shed from the apical cell surface22. Kim KC. Role of epithelial mucins during airway infection. Pulm Pharmacol Ther. 2012;25(6):415-9.. Secreted MUC5AC and MUC5B act as a physical barrier by binding to pathogens, besides performing an immunomodulatory role by capturing, retaining and releasing cytokines and growth factors11. Lillehoj EP, Kato K, Lu W, Kim KC. Cellular and molecular biology of airway mucins. Int Rev Cell Mol Biol. 2013;303:139-202.. The membrane-bound mucins, MUC1, MUC4, and MUC16, form the cell glycocalyx and activate intracellular signal transduction pathways that control epithelial cell shape, differentiation, and proliferation and modulate the inflammatory and immune responses to infectious agents33. Shurer CR, Kuo JCH, Roberts LM, Gandhi JG, Colville MJ, Enoki TA, et al. Physical principles of membrane shape regulation by the glycocalyx. Cell. 2019;177(7):1757-70..

The mucociliary clearance performed by the respiratory epithelium depends on the interactions between mucus and cilia. While the mucus entraps inhaled pathogens and other particulate material, the coordinated beating of cilia sweeps the trapped material away from the lungs toward the pharynx and mouth, where they are swallowed or expectorated. The efficient transport of mucus depends on the ciliary beating rate and the mucus’s hydration, which contributes to its viscoelastic properties. In general, more hydrated mucus is cleared more efficiently from the lungs44. Bustamante-Marin XM, Ostrowski LE. Cilia and mucociliary clearance. Cold Spring Harb Perspect Biol. 2017;9(4):a028241.. During winter, the high transmission of the three major respiratory viruses (influenza viruses, respiratory syncytial virus, and human coronaviruses) may be due to the disruption of mucociliary clearance by the season’s cold and dry air characteristics55. Moriyama M, Hugentobler WJ, Iwasaki A. Seasonality of respiratory viral infections. Annu Rev Virol. 2020;7(1):83-101.. A study of the effect of ambient temperature on the ciliary beat frequency of the nasal and tracheal ciliated cells isolated from human subjects showed that mucociliary beating begins to decline as the ambient temperature dips below 20°C and is no longer observed at 5°C55. Moriyama M, Hugentobler WJ, Iwasaki A. Seasonality of respiratory viral infections. Annu Rev Virol. 2020;7(1):83-101.. Moreover, the relative humidity (RH) of 10% decreases mucociliary clearance compared to 50% RH, resulting in impaired viral clearance following influenza virus infection of mice66. Clary-Meinesz CF, Cosson J, Huitorel P, Blaive B. Temperature effect on the ciliary beat frequency of human nasal and tracheal ciliated cells. Biol. Cell. 1992;76(3):335-8.,77. Kudo E, Song E, Yockey LJ, Rakib T, Wong PW, Homer RJ, et al. Low ambient humidity impairs barrier function and innate resistance against influenza infection. Proc Natl Acad Sci USA. 2019;116(22):10905-10..

The mucus’ proper hydration maintains the protective effect of the first defense barrier by avoiding the deterioration of the ciliary function by low temperatures and reduced humidity. Air pollutants, mainly smoking, are major disruptors of the mucociliary function. It has been extensively demonstrated that smoking causes its dysfunction and damage, including shortening of airway cilia88. Leopold PL, O’Mahony MJ, Lian XJ, Tilley AE, Harvey BG, Crystal RG, et al. Smoking is associated with shortened airway cilia. PLoS One. 2009;4(12):8157., besides increasing the expression of ACE2, the cell receptor of SARS-CoV-299. Smith JC, Sausville EL, Girish V, Yuan ML, Vasudevan A, John KM, et al. Cigarette smoke exposure and inflammatory signaling increase the expression of the SARS-CoV-2 receptor ACE2 in the respiratory tract. Dev Cell. 2020; 53(5):514-29.e3.. Furthermore, drugs as glucocorticoids1010. Chen Y, Watson AM, Williamson CD, Rahimi M, Liang C, Colberg-Poley AM, et al. Glucocorticoid receptor and HDAC2 mediate dexamethasone-induced repression of MUC5AC gene expression. Am J Respir Cell Mol Biol. 2012;47(5):637-44. and macrolide antibiotics1111. Tamaoki J. The effects of macrolides on inflammatory cells. Chest. 2004; 125(2 Suppl):41S-50S. inhibit mucin secretion, while others such as anticholinergics, aspirin, anesthetic agents, and benzodiazepines depress the mucociliary transport system1212. Houtmeyers E, Gosselink R, Gayan-Ramirez G, Decramer M. Effects of drugs on mucus clearance. Eur Respir J. 1999;14(2):452-67. and, hence, disrupt the mucociliary protective barrier.

The 2 nd barrier: Surfactant proteins

The second barrier that respiratory viruses, such as SARS-CoV-2, must overcome is a variety of soluble inhibitors in the airway lining fluid, collectively known as surfactant proteins or surfactants. Surfactants are complexes of phospholipids with four surfactant-associated proteins (SP-A, SP-B, SP-C, and SP-D), produced mainly by type II alveolar cells. They provide low surface tension to the 100 to 150 m22. Kim KC. Role of epithelial mucins during airway infection. Pulm Pharmacol Ther. 2012;25(6):415-9. of alveolar epithelium necessary to allow efficient gas exchange and prevent alveolar collapse and flooding1313. Mason RJ, Dobbs LG. Alveolar epithelium and pulmonary surfactant. In: Murray and Nadel’s Textbook of Respiratory Medicine. 2016; pp 134-49.e5, Elsevier.. SP-A and SP-D are family members of immune proteins known as collectins, or collagen-like lectins, which interact with pathogens through their lectin domains and regulate the functions of T lymphocytes, macrophages, dendritic cells, and neutrophils1414. Wright JR. Immunoregulatory functions of surfactant proteins. Nat Rev. Immunol. 2005;5(1):58-68.,1515. Nayak A, Dodagatta-Marri E, Tsolaki ZAG, Kishore U. An insight into the diverse roles of surfactant proteins, SP-A and SP-D in innate and adaptive immunity. Front Immunol. 2012;3:131.. Surfactant proteins also inhibit the invasion of epithelial cells by several respiratory viruses1616. Hsieh IN, De Luna X, White MR, Hartshorn KL. The role and molecular mechanism of action of surfactant protein D in innate host defense against influenza A virus. Front Immunol. 2018;9:1368.. The sharing of 17 pentapeptides of SARS-CoV-2 spike glycoprotein with surfactant molecules1717. Kanduc D, Shoenfeld Y. On the molecular determinants of the SARS-CoV-2 attack. Clin Immunol. 2020;215:108426. may explain the ability of SP-D to neutralize the virus and enhance its phagocytosis1818. Leth-Larsen R, Zhong F, Chow VTK, Holmkov U. The SARS coronavirus spike glycoprotein is selectively recognized by lung surfactant protein D and activates macrophages. Immunobiology. 2007;212(3):201-11.. However, the capacity of SP-D to enhance phagocytosis may lead to macrophage activation, which is involved in the development of the respiratory distress syndrome associated with severe cases of Covid-191919. Kerget B, Kerget F, Koçak AO, Kisiltunç A, Araz O, Uçar EY, et al. Are serum interleukin 6 and surfactant protein D levels associated with the clinical course of COVID-19? Lung. 2020;198:777-84..

The finding that SARS-CoV-2 can downregulate surfactant proteins and their regulators2020. Islam ABMMK, Khan MAAK. Lung biopsy cells transcriptional landscape from COVID-19 patient stratified lung injury in SARS-CoV-2 infection through impaired pulmonary surfactant metabolism. bioRxiv. 2020; https://doi.org/10.1101/2020.05.07.082297
https://doi.org/10.1101/2020.05.07.08229...
, probably due to the disruption of type II alveolar cells2121. Mirastschijski U, Dembinski R, Maedler K. Lung surfactant for pulmonary barrier restoration in patients with COVID-19 pneumonia. Front Med. 2020;7:254., has posed the possibility of using commercially available surfactants as an adjuvant treatment for Covid-19 pneumonia2121. Mirastschijski U, Dembinski R, Maedler K. Lung surfactant for pulmonary barrier restoration in patients with COVID-19 pneumonia. Front Med. 2020;7:254.,2222. Koumbourlis AC, Motoyama EK. Lung mechanics in COVID-19 resemble respiratory distress syndrome, not acute respiratory distress syndrome: could surfactant be a treatment? Am J Respir Crit Care Med. 2020;202(4):624-6.. In addition, the demonstration that the lung mechanics in Covid-19 pneumonia resembles respiratory distress syndrome caused by surfactant deficiency2222. Koumbourlis AC, Motoyama EK. Lung mechanics in COVID-19 resemble respiratory distress syndrome, not acute respiratory distress syndrome: could surfactant be a treatment? Am J Respir Crit Care Med. 2020;202(4):624-6. strengthens this claim.

The 3 rd barrier: Respiratory tract microbiota

The respiratory viruses that survive the inhibitory effects of surfactant proteins must overcome another barrier: the respiratory tract microbiota. The microbiota is a collection of microorganisms (bacteria, viruses, fungi, and archaea) that inhabit the respiratory mucosa, keep a symbiotic relationship with the organism, and play a significant role in shaping the immune system and maintaining homeostasis2323. Man WH, de Steenhuijsen Piters WAA, Bogaert D. The microbiota of the respiratory tract: gatekeeper to respiratory health. Nat Rev Microbiol. 2017;15(5):259-70.

24. Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature. 2016;535(7610):75-84.

25. Nguyen LDN, Viscogliosi E, Delhaes L. The lung mycobiome: an emerging field of the human respiratory microbiome. Front Microbiol. 2015;6:89.
-2626. Lysholm F, Wetterbom A, Lindau C, Darban H, Bjerkner A, Fahlander K, et al. Characterization of the viral microbiome in patients with severe lower respiratory tract infections, using metagenomic sequencing. PLoS One . 2012;(2):e30875.. As humans are coevolving with microorganisms since our species’ diversion, the microbiota is considered an evolutionary advantage2727. Vissers M, de Groot R, Ferwerda G. Severe viral respiratory infections: are bugs bugging? Mucosal Immunol. 2014;7(2):227-38.. Indeed, it has been shown that the respiratory tract microbiota acts as a barrier against the invasion of pathogens, including respiratory viruses, such as influenza virus2828. Lee KH, Gordon A, Shedden K, Kuan G, Ng S, Balmaseda A, et al. The respiratory microbiome and susceptibility to influenza virus infection. PLoS One . 2019;14(1):e0207898., respiratory syncytial virus2929. Antunes KH. Microbiota-derived acetate protects against respiratory syncytial virus infection through a GPR43-type 1 interferon response. Nat Commun. 2019;10(1):3273., and SARS-CoV-23030. Khatiwada S, Subedi A. Lung microbiome and coronavirus disease 2019 (COVID-19): Possible link and implications. Hum Microb J. 2020;17:100073.. The antivirus environment elicited by the respiratory tract microbiome includes activated innate and adaptive immune cells and anti-infectious molecules such as secretory IgA antibodies, defensins, acetate, and interferons2929. Antunes KH. Microbiota-derived acetate protects against respiratory syncytial virus infection through a GPR43-type 1 interferon response. Nat Commun. 2019;10(1):3273.,3131. Kim HJ, Jo A, Jeon YJ, An S, Lee KM, Yoon SS, et al. Nasal commensal Staphylococcus epidermidis enhances interferon-λ-dependent immunity against influenza virus. Microbiome. 2019;7:80..

The association between microbial dysbiosis - disruption of either the composition or the overall number of microbiota components - and increased morbidity and mortality of several respiratory infections validates the importance of the respiratory microbiota for human health3232. Invernizzi R, Lloyd CM, Molyneaux PL. Respiratory microbiome and epithelial interactions shape immunity in the lungs. Immunology. 2020;160(2):171-82.. Frequent causes of respiratory tract dysbiosis are antibiotics and intranasal corticosteroids3333. Kumpitsch C, Koskinen K, Schöpf V, Moissl-Eichinger C. The microbiome of the upper respiratory tract in health and disease. BMC Biol. 2019;17:87., smoking3434. Li KJ, Chen ZL, Huang Y, Zhang R, Luan XQ, Lei T, et al. Dysbiosis of lower respiratory tract microbiome are associated with inflammation and microbial function variety. Respir Res. 2019;20:272., and respiratory inflammations, such as asthma and chronic obstructive pulmonary disease3535. Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017;17:219-32.. Dysbiosis facilitates SARS-CoV-2 infection, and its causative factors are considered risk factors for severe disease. Moreover, it has been shown that SARS-CoV-2 causes dysbiosis of respiratory microbiota3636. He Y, Wang J, Li F, Shi Y. Main clinical features of COVID-19 and potential prognostic and therapeutic value of the microbiota in SARS-CoV-2 infections. Front Microbiol. 2020;11:1302.,3737. Han Y, Jia Z, Shi J, Wang W, He K. The active lung microbiota landscape of COVID-19 patients. medRxiv. 2020; Available from: https://doi.org/10.1101/2020.08.20.20144014.
https://doi.org/10.1101/2020.08.20.20144...
, which may facilitate the occurrence of complex mixed bacterial and fungal lung infections, a major cause of death of Covid-19 patients3838. Fan J, Li X, Gao Y, Zhou J, Wang S, Huang B, et al. The lung tissue microbiota features of 20 deceased patients with COVID-19. J Infect. 2020;81(3):e64-e67..

The best ways to maintain the respiratory microbiota as a significant defensive barrier are avoiding its disruption by drugs and smoking and strengthening its anti-pathogen capacity by using probiotic microorganisms. Clinical and experimental studies have demonstrated that probiotics exert a protective activity against respiratory viruses. Indeed, a Cochrane meta-analysis of 12 randomized control trials including 3,720 adults and children reported a 2-fold lower risk of developing upper respiratory tract infections (over 90% of them caused by viral pathogens) in subjects taking probiotics and a small but significant reduction in disease severity in those infected3939. Hao Q, Dong BR, Wu T. Probiotics for preventing acute upper respiratory tract infections. Cochrane Database Syst Rev. 2015;2:CD006895.. The possible mechanisms involved in the probiotic protection against viruses are increased levels of type I interferons, the number and activity of antigen-presenting cells, NK cells, T cells, and the levels of systemic and mucosal specific antibodies in the lungs4040. Baud D, Agri VD, Gibson GR, Reid G, Giannoni E. Using probiotics to flatten the curve of coronavirus disease COVID-2019 pandemic. Front Public Health. 2020;8:186.,4141. Villena J, Kitazawa H. The modulation of mucosal antiviral immunity by immunobiotics: could they offer any benefit in the SARS-CoV-2 pandemic? Front Physiol. 2020;11:699.. Probiotics are administered by the oral route, and their primary target is gut microbiota, which is also disrupted in SARS-CoV-2 infection4242. Zuo T, Zhang F, Lui GCY, Yeoh YK, Li AYL, Zhan H, et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology. 2020;159(3):944-955.e8.. However, the intense crosstalk between intestinal and pulmonary microbiotas mediated by microorganisms, immune cells, and their products benefit both microbiotas4343. He LH, Ren LF, Li JF, Wu YN, Li X, Zhang L. Intestinal flora as a potential strategy to fight SARS-CoV-2 infection. Front Microbiol. 2020;11:1388..

The 4 th barrier: Antimicrobial peptides - defensins and lactoferrin

Defensins are antimicrobial peptides produced by neutrophils (α-defensin) and epithelial cells (β-defensins). They are induced by microbial products or pro-inflammatory cytokines and exert multiple effects against viruses, including direct antivirus activity, modification of viral pathogenesis, and modulation of antiviral immune responses4444. Holly MK, Diaz K, Smith JG. Defensins in viral infection and pathogenesis. Annu Rev Virol . 2017;4(1):369-91.,4545. Schutte BC, McCray PB Jr. β-defensins in lung host defense. Annu Rev Physiol. 2002;64:709-48.. Defensins are potent chemotactic agents that induce migration of innate and adaptive immunity cells4646. Grigat J, Soruri A, Forssmann U, Riggert J, Zwirner J. Chemoattraction of macrophages, T lymphocytes, and mast cells is evolutionarily conserved within the human α-defensin family. J Immunol. 2007;179(6):3958-65., promote phagocytosis and present anti-inflammatory activity4747. Shelley JR, Davidson DJ, Dorin JR. The dichotomous responses driven by β-defensins. Front Immunol. 2020;11:1176.. Their immunomodulatory effects include activating immune cells and modulating cytokines’ expression and secretion4444. Holly MK, Diaz K, Smith JG. Defensins in viral infection and pathogenesis. Annu Rev Virol . 2017;4(1):369-91.,4848. Kim J, Yang YL, Jang YS. Human β-defensin 2 is involved in CCR2-mediated Nod2 signal transduction, leading to activation of the innate immune response in macrophages. Immunobiology. 2019;224(4):502-10.. In addition, defensins display direct antiviral activity on respiratory viruses4949. Klotman ME, Chang TL. Defensins in innate antiviral immunity. Nat Rev Immunol . 2006;6(6):447-56. by targeting viral envelopes, glycoproteins, capsids, inhibiting viral fusion, and providing post-entry neutralization5050. Wilson SS, Wiens ME, Smith JG. Antiviral mechanisms of human defensins. J Mol Biol. 2013;425(24):4965-80..

Studies on the effect of defensins on coronaviruses are still limited. Recent in silico5151. Kit O, Kit Y. Features of the interaction of human defensins with the SARS-CoV-2 spike protein: An in silico comparative analysis. Preprint. 2020; DOI: 10.13140/RG.2.2.22222.41281.
https://doi.org/10.13140/RG.2.2.22222.41...
and in vitro data5252. Wang C, Wang S, Li D, Zhao X, Han S, Wang T, et al. Lectin-like intestinal defensin inhibits 2019-nCoV spike binding to ACE2. bioRxiv. 2020; doi.org/10.1101/2020.03.29.013490.
https://doi.org/10.1101/2020.03.29.01349...
showed that human β-defensin-5 could block the binding of SARS-CoV-2 to ACE2+ cells. An in vivo study showed that the prophylactic treatment of BALB/c mice with theta-defensin-1 from rhesus monkey before the infection with a mouse-adapted strain of SARS-CoV-1 caused 100% survival, in contrast to 75% of untreated mice, with a modest reduction in lung pathology and without a reduction in virus titer5353. Wohlford-Lenane CL, Meyerholz DK, Perlman S, Zhou H, Tran D, Selsted ME, et al. 2009. Rhesus theta-defensin prevents death in a mouse model of Severe Acute Respiratory Syndrome Coronavirus pulmonary disease. J Virol. 2009;83(21):11385-90.. Moreover, Zhao and colleagues showed that a short peptide derived from mouse β-defensin-4 exhibited potent antiviral activity to SARS-CoV and MERS-CoV and different serotypes of influenza A virus5454. Zhao H, Zhou J, Zhang K, Chu H, Liu D, Poon VKM, et al. A novel peptide with potent and broad-spectrum antiviral activities against multiple respiratory viruses. Sci Rep. 2016;6:22008.. The authors have demonstrated that the defensin peptide bound to viral particles enters the cells via endocytosis and prevents the endosomal acidification, which blocked the membrane fusion and subsequent viral RNA release5454. Zhao H, Zhou J, Zhang K, Chu H, Liu D, Poon VKM, et al. A novel peptide with potent and broad-spectrum antiviral activities against multiple respiratory viruses. Sci Rep. 2016;6:22008.. That finding opens an avenue for developing new prophylactic and therapeutic agents with broad-spectrum antiviral activities, such as exploring human defensins as vaccine adjuvants5555. Park MS, Kim JI, Lee I, Park S, Bae JY, Park MS. Towards the application of human defensins as antivirals. Biomol Ther. 2018;26(3):242-54.. The demonstration that vitamin D induces the synthesis of β-defensins by the respiratory tract’s epithelial cells5656. Wang TT, Nestel FP, Bourdeau V, Nagai Y, Wang Q, Liao J, et al. Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. J Immunol. 2004;173(5):2909-12. poses the possibility that its deficiency might influence the prognosis of Covid-19 patients. Indeed, an association between vitamin D deficiency and the severity/mortality of Covid-19 has been demonstrated5757. Munshi R, Hussein MH, Toraih EA, Elshazli RM, Jardak C, Sultana N, et al. Vitamin D insufficiency as a potential culprit in critical COVID-19 patients. J Med Virol. 2020; 93(2):733-40.,5858. Radujkovic A, Hippchen T, Tiwari-Heckler S, Dreher S, Boxberger M, Merle U. Vitamin D deficiency and outcome of COVID-19 patients. Nutrients. 2020;2(9):2757., highlighting its possible use for improving defensin status5757. Munshi R, Hussein MH, Toraih EA, Elshazli RM, Jardak C, Sultana N, et al. Vitamin D insufficiency as a potential culprit in critical COVID-19 patients. J Med Virol. 2020; 93(2):733-40.,5959. Panfili FM, Roversi M, D’Argenio P, Rossi P, Cappa M, Fintini D. Possible role of vitamin D in Covid-19 infection in pediatric population. J Endocrinol Invest. 2021;44(1):27-35..

Another antimicrobial peptide acting on the respiratory tract’s antiviral defense is lactoferrin, considered the most versatile molecule of the organism due to the multitude of functions it exerts6060. Legrand D. Overview of lactoferrin as a natural immune modulator. J Pediatr. 2016;173:Suppl:S10-5.. Lactoferrin is a glycoprotein synthesized by mucosal epithelial cells and neutrophils, and its multiple activities rely on its ability to sequestrate metals, especially iron6060. Legrand D. Overview of lactoferrin as a natural immune modulator. J Pediatr. 2016;173:Suppl:S10-5.. The powerful immunomodulatory effects lactoferrin exerts arise from its ability to strongly bind to negatively charged structures, including immune cells receptors (e.g., toll-like, cytokine, and chemokine), DNA and RNA molecules (from host cells, viruses, and other infectious agents), as well as to microbial immunomodulatory molecules, such as bacterial lipopolysaccharide6060. Legrand D. Overview of lactoferrin as a natural immune modulator. J Pediatr. 2016;173:Suppl:S10-5.,6161. Baker EN, Baker HM. Molecular structure, binding properties and dynamics of lactoferrin. Cell Mol Life Sci. 2005;62(22):2531-9.. Lactoferrin acts on both innate and adaptive immune cells. It modulates the production of pro-inflammatory cytokines and type I interferons by macrophages and enhances their phagocytic activity; influences the maturation, migration, antigen uptake, and presentation by dendritic cells; suppresses the release of extracellular traps by neutrophils; stimulates cytotoxicity and production of IL-18 and type I interferon by NK cells; modulates T lymphocyte maturation, differentiation, activation, and the balance between Th1 and Th2 subsets; and promotes B lymphocyte maturation, enhances their capacity of antigen presentation, and secretion of IgA and IgG6060. Legrand D. Overview of lactoferrin as a natural immune modulator. J Pediatr. 2016;173:Suppl:S10-5.,6262. Okubo K, Kamiya M, Urano Y, Nishi H, Herter JM, Mayadas T, et al. Lactoferrin suppresses neutrophil extracellular traps released in inflammation. EBioMedicine. 2016;10:204-15.,6363. Suzuki YA, Lopez V, Lönnerdal B. Mammalian lactoferrin receptors: structure and function. Cell Mol Life Sci . 2005;62(22):2560-75..

Lactoferrin exerts a broad range of antiviral activity on both RNA and DNA viruses due to its ability to strongly bind to their negatively charged components (surface epitopes, nucleic acids) and host cells (membrane receptors, nucleic acids), therefore inhibiting cell invasion and intracellular replication6464. Redwan EM, Uversky VN, El-Fakharany EM, Al-Mehdar H. Potential lactoferrin activity against pathogenic viruses. CR Biologies. 2014;337(10):581-95.,6565. Jenssen H, Hancock REW. Antimicrobial properties of lactoferrin. Biochimie. 2009;91(1):19-29.. The antiviral effect of lactoferrin on respiratory viruses has been well documented6666. Scala MC, Sala M, Pietrantoni A, Spensiero A, Di Micco S, Agamennone M, et al. Lactoferrin-derived peptides active towards influenza: identification of three potent tetrapeptide inhibitors. Sci Rep. 2017;7(1):10593.. It has been found that lactoferrin exerts its function in SARS-CoV-1 infection by enhancing NK cell activity and stimulating neutrophil aggregation and adhesion6767. Reghunathan R, Jayapal M, Hsu L, Chng H, Tai D, Leung BP, et al. Expression profile of immune response genes in patients with Severe Acute Respiratory Syndrome. BMC Immunol. 2005;6:2.. An in vitro study demonstrated that lactoferrin binds to heparan sulfate proteoglycans at the host cell surface, blocks the preliminary interactions of SARS-CoV pseudovirus with the host cell, and inhibits its entry into the cell6868. Lang J, Yang N, Deng J, Liu K, Yang P, Zhang G, et al. Inhibition of SARS pseudovirus cell entry by lactoferrin binding to heparan sulfate proteoglycans. PLoS One . 2011;6:e23710.. These findings have encouraged the proposition of using exogenous lactoferrin to prevent and treat SARS-CoV-2 infection6969. Chang R, Sun WZ, Ng TB. Lactoferrin as potential preventative and treatment for COVID-19. Preprint. 2020; Available from: https://doi.org/10.31232/osf.io/mdpxc
https://doi.org/10.31232/osf.io/mdpxc...
,7070. Campione E, Cosio T, Rosa L, Lanna C, Di Girolamo S, Gaziano R, et al. Lactoferrin as protective natural barrier of respiratory and intestinal mucosa against coronavirus infection and inflammation. Int J Mol Sci. 2020;21(14):4903..

The 5 th barrier: Secretory IgA antibodies

SARS-CoV-2 virions must overcome another barrier to promote productive infection: the secretory IgA antibodies. Besides possessing a protective secretory component attached to their dimeric molecule, secretory IgA displays polyreactivity. It binds to numerous microbial antigens with low affinity, including lipopolysaccharides, DNA, flagellin, capsular polysaccharides, and virus components7171. Bunker JJ, Bendelac A. IgA responses to microbiota. Immunity. 2018;49(2):211-24.,7272. Bunker JJ, Erickson SA, Flynn TM, Henry C, Koval JC, Meisel M, et al. Natural polyreactive IgA antibodies coat the intestinal microbiota. Science. 2017;358(6361):eaan6619.. The polyreactivity of secretory IgA antibodies allows these molecules to play an essential role in the protection and homeostatic regulation of mucosal surfaces by separating the pathogen-laden outside environment from the inside of the body.

The secretory IgA barrier exerts two protective functions: it facilitates the beneficial local microbiota’s permanence and helps to eliminate noxious agents, as pathogens and pollutant particles. The finding that a substantial fraction of the microbiota components is coated with IgA antibodies without any detrimental effect suggests its functional and evolutionary relevance. Indeed, experimental data indicate that polyreactive IgA antibodies bind to microbial structures and facilitate the clustering of pathogens at the mucus layer, securing the niche from invasion by competing species7171. Bunker JJ, Bendelac A. IgA responses to microbiota. Immunity. 2018;49(2):211-24.,7373. Brandtzaeg P. Secretory IgA: designed for antimicrobial defense. Front Immunol. 2013;4:222.. Furthermore, secretory IgA molecules contribute to the survival and diversity of microbiota7474. Fransen F, Zagato E, Mazzini E, Fosso B, Manzari C, El Aidy S, et al. BALB/c and C57BL/6 mice differ in polyreactive IgA abundance, which impacts the generation of antigen-specific IgA and microbiota diversity. Immunity. 2015;43(3):527-40..

The antimicrobial effects of IgA antibodies occur at three different sites of the mucosa: (1) at the lamina propria - IgA can neutralize invading pathogens that have penetrated through breaches in the inflamed epithelium and are subsequently cleared as immune complexes; (2) during transcytosis - the crossing of the epithelial cell by IgA from the lamina propria, where it is produced, to the surface of the mucosa, by intercepting occasional incoming pathogens that are further eliminated; (3) at the surface of the mucosa - by neutralizing, delaying or abolishing the invasion of the epithelium by pathogens7575. Corthési B. Multi-faceted functions of secretory IgA at mucosal surfaces. Front Immunol . 2013;4:185.,7676. Metzger DW. IgA and respiratory immunity. In: Kaetzel CS (ed). Mucosal Immune Defense: Immunoglobulin A. pp.269-90, 2007. Springer.. Secretory IgA antibodies can restrain viruses at each of these three mucosa sites7777. Mazanec MB, Nedrud JG, Kaetzel CS, Lamm ME. A three-tiered view of the role of IgA in mucosal defense. Immunol Today. 1993;14(9):430-5.,7878. Lamm ME. Protection of mucosal epithelia by IgA: intracellular neutralization and excretion of antigens. In: Kaetzel CS (ed). Mucosal Immune Defense: Immunoglobulin A. pp.173-82, 2007. Springer., and their polyreactivity can provide cross-protection against infections with different strains and probably other species of viruses7979. Asahi-Ozaki Y, Yoshikawa T, Iwakura Y, Suzuki Y, Tamura S, Kurata T, et al. Secretory IgA antibodies provide cross-protection against infection with different strains of influenza B virus. J Med Virol . 2004;74(2):328-35.,8080. Tamura S, Funato H, Hirabayashi Y, Suzuki Y, Nagamine T, Aizawa C, et al. Cross-protection against influenza A virus infection by passively transferred respiratory tract IgA antibodies to different hemagglutinin molecules. Eur J Immunol. 1991;21(6):1337-44.. However, although both SARS-CoV-2 infection8181. Fox A, Marino J, Amanat F, Krammer F, Hahn-Holbrook J, Zolla-Pazner S, et al. Robust and specific secretory IgA against SARS-CoV-2 detected in human milk. iScience 2020; 23(11):101735. and vaccination8282. Perl SH, Uzan-Yulzari A, Klainer H, Asiskovich L, Youngster M, Rinott E, et al. SARS-CoV-2-specific antibodies in breast milk after COVID-19 vaccination of breastfeeding women. JAMA. 2021;325(19):2013-4. induce secretory IgA antibodies, their role in protection remains unsettled.

Since the microbiota induces secretory IgA, it is crucial to avoid its disruption by drugs (antibiotics, corticosteroids) or smoking, while probiotics3939. Hao Q, Dong BR, Wu T. Probiotics for preventing acute upper respiratory tract infections. Cochrane Database Syst Rev. 2015;2:CD006895. can strengthen this defense barrier. Moreover, exogenous lactoferrin can enhance IgA synthesis8383. Jang YS, Seo GY, Lee JM, Seo HY, Han HJ, Kim SJ, et al. Lactoferrin causes IgA and IgG2b isotype switching through betaglycan binding and activation of canonical TGF-β signaling. Mucosal Immunol . 2015;8(4):906-17., especially if associated with retinoic acid8484. Lee JM, Jang YS, Jin BR, Kim SJ, Kim HJ, Kwon BE, et al. Retinoic acid enhances lactoferrin-induced IgA responses by increasing betaglycan expression. Cell Mol Immunol. 2016;13(6):862-70.. In addition, the synthesis of secretory IgA could be stimulated by mucosal vaccines targeting SARS-CoV-2 given by oral or nasal routes8585. Chao YX, Rötzschke O, Tan EK. The role of IgA in COVID-19. Brain Behav Immun. 2020;87:182-3..

The 6 th barrier: Respiratory epithelial cells

For coronaviruses and eight other human respiratory tract viruses, the respiratory epithelium is, at the same time, the target of the infection and a barrier against it. In the epithelial cell’s interior and using its resources, viruses replicate and subsequently shed virions to invade other cells. However, the respiratory epithelial cells display various mechanisms to counter virus invasion and help keep the organism’s homeostatic equilibrium. Firstly, it functions as a physical barrier against invaders. Respiratory epithelial cells, which cover the whole mucosal surface in contact with the air, are tightly attached, forming an effective mechanical barrier to the virus entry and dissemination into the submucosa. Furthermore, they hinder viral access to receptors within the basolateral epithelial membrane, which is a significant entry site for several viruses8686. Bergelson JM. Intercellular junctional proteins as receptors and barriers to virus infection and spread. Cell Host Microbe. 2009;5(6):517-21.,8787. Vareille M, Kieninger E, Edwards MR, Regamey N. The airway epithelium: soldier in the fight against respiratory viruses. Clin Microbiol Rev. 2011;24(1):210-29.. As argued previously, the respiratory epithelium’s function as a physical barrier also includes the mucociliary escalator and mucin production that form the mucus layer.

The second mechanism used by respiratory epithelial cells to counter virus infection is producing antiviral molecules (interferons, lactoferrin, and defensins) that activate innate and adaptive antiviral immunity. Interferons exhibit both a direct antiviral effect and an indirect one by acting on immune cells. Type I interferons (IFN-α and IFN-β) bind to ubiquitously expressed cell receptors and induce the expression of hundreds of genes, which serve to limit further virus spread and infection. The direct antiviral activity of type I interferons includes impairment of the viral processes of cell entry, replication, transcription, translation, and the degradation of viral nucleic acids and proteins. Type I interferons also act on the immune cells by causing enhancement of phagocytosis, maturation of dendritic cells, and stimulation of cytokines and chemokines production by respiratory epithelial cells8888. Newton AH, Cardani A, Braciale TJ. The host immune response in respiratory virus infection: balancing virus clearance and immunopathology. Semin Immunopathol. 2016;38(4):471-82.. Type III interferons (IFN-λ1, IFN-λ2, and IFN-λ3) use a distinct receptor complex for signaling. They are expressed on only a few cell types, including respiratory and gastrointestinal epithelial cells, and trigger highly similar gene expression as type I interferons, suggesting that both IFN types might serve similar functions8989. Mordstein M, Neugebauer E, Ditt V, Jessen B, Rieger T, Falcone V, et al. Lambda interferon renders epithelial cells of the respiratory and gastrointestinal tracts resistant to viral infections. J Virol. 2010;84(11):5670-7..

Notwithstanding the variety of sophisticated antiviral mechanisms displayed by the respiratory epithelial cells and the fact that SARS-CoV-2 is sensitive to interferons produced by these cells9090. Lokugamage KG, Hage A, Schindewolf C, Rajsbaum R, Menachery VD. SARS-CoV-2 is sensitive to type I interferon pretreatment. bioRxiv. 2020 Apr 9;2020.03.07.982264.,9191. Mantlo E, Bukreyeva N, Maruyama J, Paessler S, Huang C. Potent antiviral activities of type I interferons to SARS-CoV-2 infection. Antiviral Res. 2020;179:104811., this virus exhibits a worrying ability to overcome this barrier by disrupting the tight junction formation that maintains the integrity of the epithelium9292. Teoh KT, Siu YL, Chan WL, Schlüter MA, Liu CJ, Peiris JSM. The SARS coronavirus E protein interacts with PALS1 and alters tight junction formation and epithelial morphogenesis. Mol Biol Cell. 2010;21(22):3838-52.. Moreover, it has been found that SARS-CoV-2 causes cell fusion, apoptosis, destruction of epithelium integrity, cilium shrinking, and beaded changes in human airway epithelium cultures9393. Zhu N, Wang W, Liu Z, Liang C, Wang W, Ye F, et al. Morphogenesis and cytopathic effect of SARS-CoV-2 infection in human airway epithelial cells. Nat Commun. 2020;11(1):3910.. In addition, it has been suggested that interferons play a role in disrupting the epithelial cell barrier during SARS-CoV-2 infection9494. Broggi A, Ghosh S, Sposito B, Spreafico R, Balzarini F, Lo Cascio A, et al. Type III interferons disrupt the lung epithelial barrier upon viral recognition. Science. 2020;369(6504):706-12. and in its repair during recovery9595. Major J, Crotta S, Llorian M, McCabe TM, Gad HH, Priestnall SL, et al. Type I and III interferons disrupt lung epithelial repair during recovery from viral infection. Science. 2020;369(6504):712-7..

Smoking is a major disruptor of the epithelial cell barrier9696. Ghosh B, Reyes-Caballero H, Akgün-Ölmez S, Nishida K, Chandrala L, Smirnova L, et al. Effect of sub-chronic exposure to cigarette smoke, electronic cigarette and waterpipe on human lung epithelial barrier function. BMC Pulm Med. 2020;20:216., besides increasing the expression of SARS-CoV-2 receptor ACE2 in the respiratory tract epithelium99. Smith JC, Sausville EL, Girish V, Yuan ML, Vasudevan A, John KM, et al. Cigarette smoke exposure and inflammatory signaling increase the expression of the SARS-CoV-2 receptor ACE2 in the respiratory tract. Dev Cell. 2020; 53(5):514-29.e3., and when in association with the virus, reduces interferon β-1 antiviral response and alters the stem cell-derived airway repair response9797. Purkayastha A, Sen C, Garcia G, Langerman J, Vijayaraj P, Durra A, et al. Direct exposure to SARS-CoV-2 and cigarette smoke increases infection severity and alters the stem cell-derived airway repair response. Cell Stem Cell.2020;27(6):869-75.. Hence, avoiding smoking is crucial for maintaining the integrity of the epithelial cell barrier. On the other hand, zinc possibly plays a role in protecting the respiratory epithelium due to its antioxidant, anti-inflammatory, and anti-apoptotic effects. Furthermore, its ability to stabilize organelles acts as a cofactor for DNA synthesis and enhances wound repair9898. Truong-Tran AQ, Carter J, Ruffin R, Zalewski PD. New insights into the role of zinc in the respiratory epithelium. Immunol Cell Biol. 2001;79(2):170-7..

The 7 th barrier: Innate immunity

The last constitutive respiratory defense barrier, the innate immunity, functions in close association with the sixth barrier, the epithelial cells, both maintaining productive crosstalk between them to fine-tuning their responses. The mechanisms of innate immunity are brought into play at the portal of entry of SARS-CoV-2, frequently the nasal goblet cells and ciliated cells of nose mucosa, which express both the ACE2 receptor and the protease TMPRSS2 necessary for host cell invasion. Interestingly, many of the top genes associated with the ACE2 gene code for innate immunity functions with antiviral activity9999. Sungnak W, Huang N, Bécavin C, Berg M, HCA Lung Biological Network. SARS-CoV-2 entry genes are most highly expressed in nasal goblet and ciliated cells within human airways. arXiv 2020; arXiv:2003.06122.. Different cell types (monocytes, macrophages, dendritic cells, innate lymphoid cells, granulocytes, and epithelial cells) and molecules (complement, surfactant, mannose-binding lectin, cytokines, and chemokines) participate in the mechanisms of innate immunity100100. Yoshikawa T, Hill TE, Yoshikawa N, Popov VL, Galindo CL. Dynamic innate immune responses of human bronchial epithelial cells to severe acute respiratory syndrome-associated coronavirus infection. PLoS One . 2010;5(1):e8729.

101. Chen J, Subbarao K. The immunobiology of SARS. Annu Rev Immunol. 2007;25:443-72.
-102102. Li G, Fan Y, Lai Y, Han T, Li Z, Zhou P, et al. Coronavirus infections, and immune responses. J Med Virol . 2020;92(4):424-32.. The sensing of virus molecules by the cells of innate immunity elicits a cascade of intra- and intercellular signals with the potential to inhibiting the development of productive infection, thereby preventing or at least mitigating illness before adaptive immunity is activated. As a countermeasure against the elaborate human defense mechanisms, SARS-CoVs develop ways to circumvent or suppress the innate immune responses to ensure a window of opportunity for efficient replication, eventually followed by disease103103. Kikkert M. Innate immune evasion by human respiratory RNA viruses. J Innate Immun. 2020;12(1):4-20..

Macrophages are the most important cells of the innate immune system at the portals of entry of SARS-CoV-2. They play significant roles in detecting viruses and virus-infected cells, clearing apoptotic/damaged cells, and inducing and regulating adaptive immune responses. Alveolar macrophages abundantly secrete cytokines, chemokines, and growth factors that ensure rapid and effective communication with epithelial, stromal, dendritic cells, T regulatory lymphocytes, and innate lymphoid cells in the pulmonary environment104104. Soroosh P, Doherty TA, Duan W, Mehta AK, Choi H, Adams YF, et al. Lung-resident tissue macrophages generate Foxp3+ regulatory T cells and promote airway tolerance. J Exp Med. 2013;210(4):775-88.

105. Westphalen K, Gusarova GA, Islam MN, Subramanian M, Cohen TS, Prince AS, et al. Sessile alveolar macrophages communicate with alveolar epithelium to modulate immunity. Nature. 2014;506(7489):503-6.
-106106. Hussell T, Bell TJ. Alveolar macrophages: plasticity in a tissue-specific context. Nat Rev Immunol . 2014;14:81-93.. They, therefore, act as a coordinator of local antivirus response. The demonstration that the viral load of SARS-CoV-2 peaked during the first week of illness then gradually declined over the second week107107. He X, Lau EHY, Wu P, Deng X, Wang J, Hao X, et al. Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat Med. 2020; 26(5):672-5.

108. Chen Y, Li L. SARS-CoV-2: virus dynamics and host response. Lancet Infect Dis. 2020;20(5):515-6.

109. Thevarajan I, Nguyen THO, Koutsakos M, Druce J, Caly L, van de Sandt C, et al. Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat Med. 2020;26(4):453-5.
-110110. Pan Y, Zhang D, Yang P, Poon LLM, Wang Q. Viral load of SARS-CoV-2 in clinical samples. Lancet Infect Dis . 2020;20(4):411-2., which means before the full development of the adaptive immune response, points to the importance of the innate immune system for controlling SARS-CoV-2 infection. The existence of individuals showing RT-PCR positive tests for the virus who, a few days later, became negative without showing any symptom attributable to Covid-19111111. Zhou X, Li Y, Li T, Zhang W. Follow-up of asymptomatic patients with SARS-CoV-2 infection. Clin Microbiol Infect. 2020;26(7):957-9.,112112. Ng OT, Marimuthu K, Chia P-Y, Koh V, Chiew CJ. SARS-CoV-2 infection among travelers returning from Wuhan, China. N Engl J Med. 2020;382(15):1476-8. https://doi.org/10.1056/NEJMc2003100.
https://doi.org/10.1056/NEJMc2003100...
is also a possible indication of the efficiency of the innate immunity to overcome the infection. Moreover, innate immunity mechanisms can control SARS-CoV-1 infection of mice in the absence of CD4+ and CD8+ T lymphocytes and antibodies113113. Chen J, Lau YF, Lamirande EW, Paddock CD, Bartlett JH, Zaki SR, et al. Cellular immune responses to Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) infection in senescent BALB/c mice: CD4+ T cells are important in control of SARS-CoV infection. J Virol. 2010;84:1289-301..

Since the excessive activation of macrophages may lead to hyperinflammation, a significant cause of disease severity and death in SARS-CoV-2 infection114114. Merad M, Martin JC. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat Rev Immunol . 2020;20:355-62., regulating their function may strengthen the innate immunity barrier of defense. Different compounds seem to display the ability to modulate macrophage functions, including zinc115115. Pyle CJ, Akhter S, Bao SY, Dodd CE, Schlesinger LS, Knoell DL. Zinc modulates endotoxin-induced human macrophage inflammation through ZIP8 induction and C/EBPβ inhibition. PLoS One . 2017;12(1):e0169531., vitamin D116116. Carlsberg C. Vitamin D signaling in the context of innate immunity: focus on human monocytes. Front Immunol . 2019;10:2211., thalidomide117117. Ye Q, Chen B, Tong Z, Nakamura S, Sarria R, Costabel U, et al. Thalidomide reduces IL-18, IL-8 and TNF-α release from alveolar macrophages in interstitial lung disease. Eur Respir J . 2006;28(4):824-31., probiotics118118. Hu R, Lin H, Li J, Zhao Y, Wang M, Sun X, et al. Probiotic Escherichia coli Nissle 1917-derived outer membrane vesicles enhance immunomodulation and antimicrobial activity in RAW264.7 macrophages. BMC Microbiol. 2020;20(1):68., omega-3 fatty acids119119. Xue B, Yang Z, Wang X, Shi H. Omega-3 polyunsaturated fatty acids antagonize macrophage inflammation via activation of AMPK/SIRT1 pathway. PLoS One . 2012;7(10):e45990., metformin120120. Zhang L, Lu L, Zhong X, Yue Y, Hong Y, Li Y, et al. Metformin reduced NLRP3 inflammasome activity in Ox-LDL stimulated macrophages through adenosine monophosphate activated protein kinase and protein phosphatase 2A. Eur J Pharmacol. 2019;852:99-106., curcumin121121. Zhou Y, Zhang T, Wang X, Wei X, Chen Y, Guo L, et al. Curcumin modulates macrophage polarization through the inhibition of the toll-like receptor 4 expression and its signaling pathways. Cell Physiol Biochem. 2015;36(2):631-41., and coenzyme Q10122122. Zhai J, Bo Y, Lu Y, Liu C, Zhang L. Effects of coenzyme Q10 on markers of inflammation: a systematic review and meta-analysis. PLoS One . 2017;2(1):e0170172.. Therefore, the investigation of their possible use to mitigate Covid-19 severity is warranted.

CONCLUSION

Before causing a productive infection, SARS-CoV-2 must overcome seven constitutive respiratory defense barriers and an elicited one, the adaptive immunity (discussed elsewhere123123. Tosta E. The protective immunity induced by SARS-CoV-2 infection and vaccination: a critical appraisal. Explor Immunol. 2021;1:199-225.). The fact that months after its emergence, SARS-CoV-2 had infected over 100 million people indicates that the virus possesses a piece of machinery that allows it to evade all those defense barriers. Some of these mechanisms include the impairment of interferon production by host cells, the ability to hide immunogenic motifs from cell receptors, the concealment of viral RNA to avoid detection by cell sensors, the triggering of human defense cell death, and the impairment of lymphocyte functionality. This remarkable ability to evade human defense mechanisms implies that precursors SARS-CoV-2 of have probably been circulating among humans for a reasonable yet unknown time before the pandemics started. During this adaptation phase, virus variants were gradually ‘learning’ how to overcome the different human defense barriers until a full-brown variant emerged in December 2019, leading to the Covid-19 pandemic. It is anticipated that as far as adaptation progresses under the selective pressure of the immune system, the infection’s destructive burden will reduce, and eventually, SARS-CoV-2 may become part of human respiratory virome, as had occurred with other coronaviruses.

ACKNOWLEDGMENTS

I dedicate this work to my scientist colleagues worldwide who are giving their best to mitigating the tremendous suffering caused by SARS-CoV-2 pandemic in an unprecedentedly short time. I am grateful to Luzia Guimarães, Lena Dias-Tosta, and João Luiz Pacini for their helpful support.

REFERENCES

  • 1
    Lillehoj EP, Kato K, Lu W, Kim KC. Cellular and molecular biology of airway mucins. Int Rev Cell Mol Biol. 2013;303:139-202.
  • 2
    Kim KC. Role of epithelial mucins during airway infection. Pulm Pharmacol Ther. 2012;25(6):415-9.
  • 3
    Shurer CR, Kuo JCH, Roberts LM, Gandhi JG, Colville MJ, Enoki TA, et al. Physical principles of membrane shape regulation by the glycocalyx. Cell. 2019;177(7):1757-70.
  • 4
    Bustamante-Marin XM, Ostrowski LE. Cilia and mucociliary clearance. Cold Spring Harb Perspect Biol. 2017;9(4):a028241.
  • 5
    Moriyama M, Hugentobler WJ, Iwasaki A. Seasonality of respiratory viral infections. Annu Rev Virol. 2020;7(1):83-101.
  • 6
    Clary-Meinesz CF, Cosson J, Huitorel P, Blaive B. Temperature effect on the ciliary beat frequency of human nasal and tracheal ciliated cells. Biol. Cell. 1992;76(3):335-8.
  • 7
    Kudo E, Song E, Yockey LJ, Rakib T, Wong PW, Homer RJ, et al. Low ambient humidity impairs barrier function and innate resistance against influenza infection. Proc Natl Acad Sci USA. 2019;116(22):10905-10.
  • 8
    Leopold PL, O’Mahony MJ, Lian XJ, Tilley AE, Harvey BG, Crystal RG, et al. Smoking is associated with shortened airway cilia. PLoS One. 2009;4(12):8157.
  • 9
    Smith JC, Sausville EL, Girish V, Yuan ML, Vasudevan A, John KM, et al. Cigarette smoke exposure and inflammatory signaling increase the expression of the SARS-CoV-2 receptor ACE2 in the respiratory tract. Dev Cell. 2020; 53(5):514-29.e3.
  • 10
    Chen Y, Watson AM, Williamson CD, Rahimi M, Liang C, Colberg-Poley AM, et al. Glucocorticoid receptor and HDAC2 mediate dexamethasone-induced repression of MUC5AC gene expression. Am J Respir Cell Mol Biol. 2012;47(5):637-44.
  • 11
    Tamaoki J. The effects of macrolides on inflammatory cells. Chest. 2004; 125(2 Suppl):41S-50S.
  • 12
    Houtmeyers E, Gosselink R, Gayan-Ramirez G, Decramer M. Effects of drugs on mucus clearance. Eur Respir J. 1999;14(2):452-67.
  • 13
    Mason RJ, Dobbs LG. Alveolar epithelium and pulmonary surfactant. In: Murray and Nadel’s Textbook of Respiratory Medicine. 2016; pp 134-49.e5, Elsevier.
  • 14
    Wright JR. Immunoregulatory functions of surfactant proteins. Nat Rev. Immunol. 2005;5(1):58-68.
  • 15
    Nayak A, Dodagatta-Marri E, Tsolaki ZAG, Kishore U. An insight into the diverse roles of surfactant proteins, SP-A and SP-D in innate and adaptive immunity. Front Immunol. 2012;3:131.
  • 16
    Hsieh IN, De Luna X, White MR, Hartshorn KL. The role and molecular mechanism of action of surfactant protein D in innate host defense against influenza A virus. Front Immunol. 2018;9:1368.
  • 17
    Kanduc D, Shoenfeld Y. On the molecular determinants of the SARS-CoV-2 attack. Clin Immunol. 2020;215:108426.
  • 18
    Leth-Larsen R, Zhong F, Chow VTK, Holmkov U. The SARS coronavirus spike glycoprotein is selectively recognized by lung surfactant protein D and activates macrophages. Immunobiology. 2007;212(3):201-11.
  • 19
    Kerget B, Kerget F, Koçak AO, Kisiltunç A, Araz O, Uçar EY, et al. Are serum interleukin 6 and surfactant protein D levels associated with the clinical course of COVID-19? Lung. 2020;198:777-84.
  • 20
    Islam ABMMK, Khan MAAK. Lung biopsy cells transcriptional landscape from COVID-19 patient stratified lung injury in SARS-CoV-2 infection through impaired pulmonary surfactant metabolism. bioRxiv. 2020; https://doi.org/10.1101/2020.05.07.082297
    » https://doi.org/10.1101/2020.05.07.082297
  • 21
    Mirastschijski U, Dembinski R, Maedler K. Lung surfactant for pulmonary barrier restoration in patients with COVID-19 pneumonia. Front Med. 2020;7:254.
  • 22
    Koumbourlis AC, Motoyama EK. Lung mechanics in COVID-19 resemble respiratory distress syndrome, not acute respiratory distress syndrome: could surfactant be a treatment? Am J Respir Crit Care Med. 2020;202(4):624-6.
  • 23
    Man WH, de Steenhuijsen Piters WAA, Bogaert D. The microbiota of the respiratory tract: gatekeeper to respiratory health. Nat Rev Microbiol. 2017;15(5):259-70.
  • 24
    Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature. 2016;535(7610):75-84.
  • 25
    Nguyen LDN, Viscogliosi E, Delhaes L. The lung mycobiome: an emerging field of the human respiratory microbiome. Front Microbiol. 2015;6:89.
  • 26
    Lysholm F, Wetterbom A, Lindau C, Darban H, Bjerkner A, Fahlander K, et al. Characterization of the viral microbiome in patients with severe lower respiratory tract infections, using metagenomic sequencing. PLoS One . 2012;(2):e30875.
  • 27
    Vissers M, de Groot R, Ferwerda G. Severe viral respiratory infections: are bugs bugging? Mucosal Immunol. 2014;7(2):227-38.
  • 28
    Lee KH, Gordon A, Shedden K, Kuan G, Ng S, Balmaseda A, et al. The respiratory microbiome and susceptibility to influenza virus infection. PLoS One . 2019;14(1):e0207898.
  • 29
    Antunes KH. Microbiota-derived acetate protects against respiratory syncytial virus infection through a GPR43-type 1 interferon response. Nat Commun. 2019;10(1):3273.
  • 30
    Khatiwada S, Subedi A. Lung microbiome and coronavirus disease 2019 (COVID-19): Possible link and implications. Hum Microb J. 2020;17:100073.
  • 31
    Kim HJ, Jo A, Jeon YJ, An S, Lee KM, Yoon SS, et al. Nasal commensal Staphylococcus epidermidis enhances interferon-λ-dependent immunity against influenza virus. Microbiome. 2019;7:80.
  • 32
    Invernizzi R, Lloyd CM, Molyneaux PL. Respiratory microbiome and epithelial interactions shape immunity in the lungs. Immunology. 2020;160(2):171-82.
  • 33
    Kumpitsch C, Koskinen K, Schöpf V, Moissl-Eichinger C. The microbiome of the upper respiratory tract in health and disease. BMC Biol. 2019;17:87.
  • 34
    Li KJ, Chen ZL, Huang Y, Zhang R, Luan XQ, Lei T, et al. Dysbiosis of lower respiratory tract microbiome are associated with inflammation and microbial function variety. Respir Res. 2019;20:272.
  • 35
    Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017;17:219-32.
  • 36
    He Y, Wang J, Li F, Shi Y. Main clinical features of COVID-19 and potential prognostic and therapeutic value of the microbiota in SARS-CoV-2 infections. Front Microbiol. 2020;11:1302.
  • 37
    Han Y, Jia Z, Shi J, Wang W, He K. The active lung microbiota landscape of COVID-19 patients. medRxiv. 2020; Available from: https://doi.org/10.1101/2020.08.20.20144014.
    » https://doi.org/10.1101/2020.08.20.20144014
  • 38
    Fan J, Li X, Gao Y, Zhou J, Wang S, Huang B, et al. The lung tissue microbiota features of 20 deceased patients with COVID-19. J Infect. 2020;81(3):e64-e67.
  • 39
    Hao Q, Dong BR, Wu T. Probiotics for preventing acute upper respiratory tract infections. Cochrane Database Syst Rev. 2015;2:CD006895.
  • 40
    Baud D, Agri VD, Gibson GR, Reid G, Giannoni E. Using probiotics to flatten the curve of coronavirus disease COVID-2019 pandemic. Front Public Health. 2020;8:186.
  • 41
    Villena J, Kitazawa H. The modulation of mucosal antiviral immunity by immunobiotics: could they offer any benefit in the SARS-CoV-2 pandemic? Front Physiol. 2020;11:699.
  • 42
    Zuo T, Zhang F, Lui GCY, Yeoh YK, Li AYL, Zhan H, et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology. 2020;159(3):944-955.e8.
  • 43
    He LH, Ren LF, Li JF, Wu YN, Li X, Zhang L. Intestinal flora as a potential strategy to fight SARS-CoV-2 infection. Front Microbiol. 2020;11:1388.
  • 44
    Holly MK, Diaz K, Smith JG. Defensins in viral infection and pathogenesis. Annu Rev Virol . 2017;4(1):369-91.
  • 45
    Schutte BC, McCray PB Jr. β-defensins in lung host defense. Annu Rev Physiol. 2002;64:709-48.
  • 46
    Grigat J, Soruri A, Forssmann U, Riggert J, Zwirner J. Chemoattraction of macrophages, T lymphocytes, and mast cells is evolutionarily conserved within the human α-defensin family. J Immunol. 2007;179(6):3958-65.
  • 47
    Shelley JR, Davidson DJ, Dorin JR. The dichotomous responses driven by β-defensins. Front Immunol. 2020;11:1176.
  • 48
    Kim J, Yang YL, Jang YS. Human β-defensin 2 is involved in CCR2-mediated Nod2 signal transduction, leading to activation of the innate immune response in macrophages. Immunobiology. 2019;224(4):502-10.
  • 49
    Klotman ME, Chang TL. Defensins in innate antiviral immunity. Nat Rev Immunol . 2006;6(6):447-56.
  • 50
    Wilson SS, Wiens ME, Smith JG. Antiviral mechanisms of human defensins. J Mol Biol. 2013;425(24):4965-80.
  • 51
    Kit O, Kit Y. Features of the interaction of human defensins with the SARS-CoV-2 spike protein: An in silico comparative analysis. Preprint. 2020; DOI: 10.13140/RG.2.2.22222.41281.
    » https://doi.org/10.13140/RG.2.2.22222.41281
  • 52
    Wang C, Wang S, Li D, Zhao X, Han S, Wang T, et al. Lectin-like intestinal defensin inhibits 2019-nCoV spike binding to ACE2. bioRxiv. 2020; doi.org/10.1101/2020.03.29.013490.
    » https://doi.org/10.1101/2020.03.29.013490
  • 53
    Wohlford-Lenane CL, Meyerholz DK, Perlman S, Zhou H, Tran D, Selsted ME, et al. 2009. Rhesus theta-defensin prevents death in a mouse model of Severe Acute Respiratory Syndrome Coronavirus pulmonary disease. J Virol. 2009;83(21):11385-90.
  • 54
    Zhao H, Zhou J, Zhang K, Chu H, Liu D, Poon VKM, et al. A novel peptide with potent and broad-spectrum antiviral activities against multiple respiratory viruses. Sci Rep. 2016;6:22008.
  • 55
    Park MS, Kim JI, Lee I, Park S, Bae JY, Park MS. Towards the application of human defensins as antivirals. Biomol Ther. 2018;26(3):242-54.
  • 56
    Wang TT, Nestel FP, Bourdeau V, Nagai Y, Wang Q, Liao J, et al. Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. J Immunol. 2004;173(5):2909-12.
  • 57
    Munshi R, Hussein MH, Toraih EA, Elshazli RM, Jardak C, Sultana N, et al. Vitamin D insufficiency as a potential culprit in critical COVID-19 patients. J Med Virol. 2020; 93(2):733-40.
  • 58
    Radujkovic A, Hippchen T, Tiwari-Heckler S, Dreher S, Boxberger M, Merle U. Vitamin D deficiency and outcome of COVID-19 patients. Nutrients. 2020;2(9):2757.
  • 59
    Panfili FM, Roversi M, D’Argenio P, Rossi P, Cappa M, Fintini D. Possible role of vitamin D in Covid-19 infection in pediatric population. J Endocrinol Invest. 2021;44(1):27-35.
  • 60
    Legrand D. Overview of lactoferrin as a natural immune modulator. J Pediatr. 2016;173:Suppl:S10-5.
  • 61
    Baker EN, Baker HM. Molecular structure, binding properties and dynamics of lactoferrin. Cell Mol Life Sci. 2005;62(22):2531-9.
  • 62
    Okubo K, Kamiya M, Urano Y, Nishi H, Herter JM, Mayadas T, et al. Lactoferrin suppresses neutrophil extracellular traps released in inflammation. EBioMedicine. 2016;10:204-15.
  • 63
    Suzuki YA, Lopez V, Lönnerdal B. Mammalian lactoferrin receptors: structure and function. Cell Mol Life Sci . 2005;62(22):2560-75.
  • 64
    Redwan EM, Uversky VN, El-Fakharany EM, Al-Mehdar H. Potential lactoferrin activity against pathogenic viruses. CR Biologies. 2014;337(10):581-95.
  • 65
    Jenssen H, Hancock REW. Antimicrobial properties of lactoferrin. Biochimie. 2009;91(1):19-29.
  • 66
    Scala MC, Sala M, Pietrantoni A, Spensiero A, Di Micco S, Agamennone M, et al. Lactoferrin-derived peptides active towards influenza: identification of three potent tetrapeptide inhibitors. Sci Rep. 2017;7(1):10593.
  • 67
    Reghunathan R, Jayapal M, Hsu L, Chng H, Tai D, Leung BP, et al. Expression profile of immune response genes in patients with Severe Acute Respiratory Syndrome. BMC Immunol. 2005;6:2.
  • 68
    Lang J, Yang N, Deng J, Liu K, Yang P, Zhang G, et al. Inhibition of SARS pseudovirus cell entry by lactoferrin binding to heparan sulfate proteoglycans. PLoS One . 2011;6:e23710.
  • 69
    Chang R, Sun WZ, Ng TB. Lactoferrin as potential preventative and treatment for COVID-19. Preprint. 2020; Available from: https://doi.org/10.31232/osf.io/mdpxc
    » https://doi.org/10.31232/osf.io/mdpxc
  • 70
    Campione E, Cosio T, Rosa L, Lanna C, Di Girolamo S, Gaziano R, et al. Lactoferrin as protective natural barrier of respiratory and intestinal mucosa against coronavirus infection and inflammation. Int J Mol Sci. 2020;21(14):4903.
  • 71
    Bunker JJ, Bendelac A. IgA responses to microbiota. Immunity. 2018;49(2):211-24.
  • 72
    Bunker JJ, Erickson SA, Flynn TM, Henry C, Koval JC, Meisel M, et al. Natural polyreactive IgA antibodies coat the intestinal microbiota. Science. 2017;358(6361):eaan6619.
  • 73
    Brandtzaeg P. Secretory IgA: designed for antimicrobial defense. Front Immunol. 2013;4:222.
  • 74
    Fransen F, Zagato E, Mazzini E, Fosso B, Manzari C, El Aidy S, et al. BALB/c and C57BL/6 mice differ in polyreactive IgA abundance, which impacts the generation of antigen-specific IgA and microbiota diversity. Immunity. 2015;43(3):527-40.
  • 75
    Corthési B. Multi-faceted functions of secretory IgA at mucosal surfaces. Front Immunol . 2013;4:185.
  • 76
    Metzger DW. IgA and respiratory immunity. In: Kaetzel CS (ed). Mucosal Immune Defense: Immunoglobulin A. pp.269-90, 2007. Springer.
  • 77
    Mazanec MB, Nedrud JG, Kaetzel CS, Lamm ME. A three-tiered view of the role of IgA in mucosal defense. Immunol Today. 1993;14(9):430-5.
  • 78
    Lamm ME. Protection of mucosal epithelia by IgA: intracellular neutralization and excretion of antigens. In: Kaetzel CS (ed). Mucosal Immune Defense: Immunoglobulin A. pp.173-82, 2007. Springer.
  • 79
    Asahi-Ozaki Y, Yoshikawa T, Iwakura Y, Suzuki Y, Tamura S, Kurata T, et al. Secretory IgA antibodies provide cross-protection against infection with different strains of influenza B virus. J Med Virol . 2004;74(2):328-35.
  • 80
    Tamura S, Funato H, Hirabayashi Y, Suzuki Y, Nagamine T, Aizawa C, et al. Cross-protection against influenza A virus infection by passively transferred respiratory tract IgA antibodies to different hemagglutinin molecules. Eur J Immunol. 1991;21(6):1337-44.
  • 81
    Fox A, Marino J, Amanat F, Krammer F, Hahn-Holbrook J, Zolla-Pazner S, et al. Robust and specific secretory IgA against SARS-CoV-2 detected in human milk. iScience 2020; 23(11):101735.
  • 82
    Perl SH, Uzan-Yulzari A, Klainer H, Asiskovich L, Youngster M, Rinott E, et al. SARS-CoV-2-specific antibodies in breast milk after COVID-19 vaccination of breastfeeding women. JAMA. 2021;325(19):2013-4.
  • 83
    Jang YS, Seo GY, Lee JM, Seo HY, Han HJ, Kim SJ, et al. Lactoferrin causes IgA and IgG2b isotype switching through betaglycan binding and activation of canonical TGF-β signaling. Mucosal Immunol . 2015;8(4):906-17.
  • 84
    Lee JM, Jang YS, Jin BR, Kim SJ, Kim HJ, Kwon BE, et al. Retinoic acid enhances lactoferrin-induced IgA responses by increasing betaglycan expression. Cell Mol Immunol. 2016;13(6):862-70.
  • 85
    Chao YX, Rötzschke O, Tan EK. The role of IgA in COVID-19. Brain Behav Immun. 2020;87:182-3.
  • 86
    Bergelson JM. Intercellular junctional proteins as receptors and barriers to virus infection and spread. Cell Host Microbe. 2009;5(6):517-21.
  • 87
    Vareille M, Kieninger E, Edwards MR, Regamey N. The airway epithelium: soldier in the fight against respiratory viruses. Clin Microbiol Rev. 2011;24(1):210-29.
  • 88
    Newton AH, Cardani A, Braciale TJ. The host immune response in respiratory virus infection: balancing virus clearance and immunopathology. Semin Immunopathol. 2016;38(4):471-82.
  • 89
    Mordstein M, Neugebauer E, Ditt V, Jessen B, Rieger T, Falcone V, et al. Lambda interferon renders epithelial cells of the respiratory and gastrointestinal tracts resistant to viral infections. J Virol. 2010;84(11):5670-7.
  • 90
    Lokugamage KG, Hage A, Schindewolf C, Rajsbaum R, Menachery VD. SARS-CoV-2 is sensitive to type I interferon pretreatment. bioRxiv. 2020 Apr 9;2020.03.07.982264.
  • 91
    Mantlo E, Bukreyeva N, Maruyama J, Paessler S, Huang C. Potent antiviral activities of type I interferons to SARS-CoV-2 infection. Antiviral Res. 2020;179:104811.
  • 92
    Teoh KT, Siu YL, Chan WL, Schlüter MA, Liu CJ, Peiris JSM. The SARS coronavirus E protein interacts with PALS1 and alters tight junction formation and epithelial morphogenesis. Mol Biol Cell. 2010;21(22):3838-52.
  • 93
    Zhu N, Wang W, Liu Z, Liang C, Wang W, Ye F, et al. Morphogenesis and cytopathic effect of SARS-CoV-2 infection in human airway epithelial cells. Nat Commun. 2020;11(1):3910.
  • 94
    Broggi A, Ghosh S, Sposito B, Spreafico R, Balzarini F, Lo Cascio A, et al. Type III interferons disrupt the lung epithelial barrier upon viral recognition. Science. 2020;369(6504):706-12.
  • 95
    Major J, Crotta S, Llorian M, McCabe TM, Gad HH, Priestnall SL, et al. Type I and III interferons disrupt lung epithelial repair during recovery from viral infection. Science. 2020;369(6504):712-7.
  • 96
    Ghosh B, Reyes-Caballero H, Akgün-Ölmez S, Nishida K, Chandrala L, Smirnova L, et al. Effect of sub-chronic exposure to cigarette smoke, electronic cigarette and waterpipe on human lung epithelial barrier function. BMC Pulm Med. 2020;20:216.
  • 97
    Purkayastha A, Sen C, Garcia G, Langerman J, Vijayaraj P, Durra A, et al. Direct exposure to SARS-CoV-2 and cigarette smoke increases infection severity and alters the stem cell-derived airway repair response. Cell Stem Cell.2020;27(6):869-75.
  • 98
    Truong-Tran AQ, Carter J, Ruffin R, Zalewski PD. New insights into the role of zinc in the respiratory epithelium. Immunol Cell Biol. 2001;79(2):170-7.
  • 99
    Sungnak W, Huang N, Bécavin C, Berg M, HCA Lung Biological Network. SARS-CoV-2 entry genes are most highly expressed in nasal goblet and ciliated cells within human airways. arXiv 2020; arXiv:2003.06122.
  • 100
    Yoshikawa T, Hill TE, Yoshikawa N, Popov VL, Galindo CL. Dynamic innate immune responses of human bronchial epithelial cells to severe acute respiratory syndrome-associated coronavirus infection. PLoS One . 2010;5(1):e8729.
  • 101
    Chen J, Subbarao K. The immunobiology of SARS. Annu Rev Immunol. 2007;25:443-72.
  • 102
    Li G, Fan Y, Lai Y, Han T, Li Z, Zhou P, et al. Coronavirus infections, and immune responses. J Med Virol . 2020;92(4):424-32.
  • 103
    Kikkert M. Innate immune evasion by human respiratory RNA viruses. J Innate Immun. 2020;12(1):4-20.
  • 104
    Soroosh P, Doherty TA, Duan W, Mehta AK, Choi H, Adams YF, et al. Lung-resident tissue macrophages generate Foxp3+ regulatory T cells and promote airway tolerance. J Exp Med. 2013;210(4):775-88.
  • 105
    Westphalen K, Gusarova GA, Islam MN, Subramanian M, Cohen TS, Prince AS, et al. Sessile alveolar macrophages communicate with alveolar epithelium to modulate immunity. Nature. 2014;506(7489):503-6.
  • 106
    Hussell T, Bell TJ. Alveolar macrophages: plasticity in a tissue-specific context. Nat Rev Immunol . 2014;14:81-93.
  • 107
    He X, Lau EHY, Wu P, Deng X, Wang J, Hao X, et al. Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat Med. 2020; 26(5):672-5.
  • 108
    Chen Y, Li L. SARS-CoV-2: virus dynamics and host response. Lancet Infect Dis. 2020;20(5):515-6.
  • 109
    Thevarajan I, Nguyen THO, Koutsakos M, Druce J, Caly L, van de Sandt C, et al. Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat Med. 2020;26(4):453-5.
  • 110
    Pan Y, Zhang D, Yang P, Poon LLM, Wang Q. Viral load of SARS-CoV-2 in clinical samples. Lancet Infect Dis . 2020;20(4):411-2.
  • 111
    Zhou X, Li Y, Li T, Zhang W. Follow-up of asymptomatic patients with SARS-CoV-2 infection. Clin Microbiol Infect. 2020;26(7):957-9.
  • 112
    Ng OT, Marimuthu K, Chia P-Y, Koh V, Chiew CJ. SARS-CoV-2 infection among travelers returning from Wuhan, China. N Engl J Med. 2020;382(15):1476-8. https://doi.org/10.1056/NEJMc2003100.
    » https://doi.org/10.1056/NEJMc2003100
  • 113
    Chen J, Lau YF, Lamirande EW, Paddock CD, Bartlett JH, Zaki SR, et al. Cellular immune responses to Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) infection in senescent BALB/c mice: CD4+ T cells are important in control of SARS-CoV infection. J Virol. 2010;84:1289-301.
  • 114
    Merad M, Martin JC. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat Rev Immunol . 2020;20:355-62.
  • 115
    Pyle CJ, Akhter S, Bao SY, Dodd CE, Schlesinger LS, Knoell DL. Zinc modulates endotoxin-induced human macrophage inflammation through ZIP8 induction and C/EBPβ inhibition. PLoS One . 2017;12(1):e0169531.
  • 116
    Carlsberg C. Vitamin D signaling in the context of innate immunity: focus on human monocytes. Front Immunol . 2019;10:2211.
  • 117
    Ye Q, Chen B, Tong Z, Nakamura S, Sarria R, Costabel U, et al. Thalidomide reduces IL-18, IL-8 and TNF-α release from alveolar macrophages in interstitial lung disease. Eur Respir J . 2006;28(4):824-31.
  • 118
    Hu R, Lin H, Li J, Zhao Y, Wang M, Sun X, et al. Probiotic Escherichia coli Nissle 1917-derived outer membrane vesicles enhance immunomodulation and antimicrobial activity in RAW264.7 macrophages. BMC Microbiol. 2020;20(1):68.
  • 119
    Xue B, Yang Z, Wang X, Shi H. Omega-3 polyunsaturated fatty acids antagonize macrophage inflammation via activation of AMPK/SIRT1 pathway. PLoS One . 2012;7(10):e45990.
  • 120
    Zhang L, Lu L, Zhong X, Yue Y, Hong Y, Li Y, et al. Metformin reduced NLRP3 inflammasome activity in Ox-LDL stimulated macrophages through adenosine monophosphate activated protein kinase and protein phosphatase 2A. Eur J Pharmacol. 2019;852:99-106.
  • 121
    Zhou Y, Zhang T, Wang X, Wei X, Chen Y, Guo L, et al. Curcumin modulates macrophage polarization through the inhibition of the toll-like receptor 4 expression and its signaling pathways. Cell Physiol Biochem. 2015;36(2):631-41.
  • 122
    Zhai J, Bo Y, Lu Y, Liu C, Zhang L. Effects of coenzyme Q10 on markers of inflammation: a systematic review and meta-analysis. PLoS One . 2017;2(1):e0170172.
  • 123
    Tosta E. The protective immunity induced by SARS-CoV-2 infection and vaccination: a critical appraisal. Explor Immunol. 2021;1:199-225.
  • Financial Support: The author received no financial support and declares no conflict of interests.

Publication Dates

  • Publication in this collection
    17 Dec 2021
  • Date of issue
    2021

History

  • Received
    27 July 2021
  • Accepted
    17 Sept 2021
Sociedade Brasileira de Medicina Tropical - SBMT Caixa Postal 118, 38001-970 Uberaba MG Brazil, Tel.: +55 34 3318-5255 / +55 34 3318-5636/ +55 34 3318-5287, http://rsbmt.org.br/ - Uberaba - MG - Brazil
E-mail: rsbmt@uftm.edu.br