key: cord-0761178-0zwakfah authors: Rebuli, Meghan E.; Brocke, Stephanie A.; Jaspers, Ilona title: Impact of inhaled pollutants on response to viral infection in controlled exposures date: 2021-07-10 journal: J Allergy Clin Immunol DOI: 10.1016/j.jaci.2021.07.002 sha: fd1c907c75110b593d1150ba1f42e0d91a640863 doc_id: 761178 cord_uid: 0zwakfah Air pollutants are a major source of increased risk of disease, hospitalization, morbidity, and mortality worldwide. The respiratory tract is a primary target of potential concurrent exposure to both inhaled pollutants and pathogens, including viruses. Although there are various associative studies linking adverse outcomes to co- or subsequent exposures to inhaled pollutants and viruses, knowledge about causal linkages and mechanisms by which pollutant exposure may alter human respiratory responses to viral infection is more limited. In this article, we review what is known about the impact of pollutant exposure on antiviral host defense responses and describe potential mechanisms by which pollutants can alter the viral infection cycle. This review focuses on evidence from human observational and controlled exposure, ex vivo, and in vitro studies. Overall, there are a myriad of points throughout the viral infection cycle that inhaled pollutants can alter to modulate appropriate host defense responses. These alterations may contribute to observed increases in rates of viral infection and associated morbidity and mortality in areas of the world with high ambient pollution levels or in people using tobacco products. Although the understanding of mechanisms of interaction is advancing through controlled in vivo and in vitro exposure models, more studies are needed because emerging infectious pathogens, such as severe acute respiratory syndrome coronavirus 2, present a significant threat to public health. Air pollutants are a major source of increased risk for disease, hospitalization, morbidity, and mortality worldwide. [1] [2] [3] Because the lungs filter more than 12,000 liters of air per day, they are a primary target of potential concurrent exposure to both inhaled pollutants and pathogens, including viruses. Although there are various associative studies linking adverse outcomes to co-or subsequent exposures to inhaled pollutants and viruses, causal linkages and mechanisms by which pollutant exposure may alter human respiratory response to viral infection are more limited. [4] [5] [6] [7] The limited knowledge on how pollutants may impact response to viral infection has been acutely highlighted with the onset of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic and the resulting flurry of review articles that nicely summarize identified associations but do little to capture causal linkages. 8, 9 In this article, we will focus on the impact of pollutant exposure on antiviral host defense responses and describe potential mechanisms by which pollutants can alter the viral infection cycle that have been identified through human observational and controlled exposure studies, as well as mechanistic in vitro studies. In addition to experimental inoculation with rhinovirus, 10 the Food and Drug Administration-approved live attenuated influenza virus (LAIV) vaccine presents a unique and underused model to mechanistically explore the interaction between pollutants and respiratory viruses in humans in vivo. The live, but attenuated virus replicates only at lower temperatures in the nasal passages, but causes similar innate and adaptive host defense responses, similar to community-acquired infections. 11 Hence, inoculation with LAIV allows the study of active viral infection and immune responses in the nasal passages of human volunteers, without risking the overall safety of the participants. The LAIV model is also ideal for studying the effects of viruses for whom the primary point of entry and activation is likely the nasal passage, such as influenza and SARS-CoV-2. 12 It should be noted that the use of the LAIV model is limited by contraindications in individuals with severe allergic reaction to its ingredients and children and adolescents on asprin therapy. However, the easyto-use LAIV model of viral infection is well suited to determine the effects of ambient air pollutants, such as diesel exhaust and wood smoke, as well as use of tobacco products, including cigarettes and e-cigarettes on host defense responses. Diesel exhaust contributes a significant percentage of trafficrelated air pollution in many cities, due to increased needs for transportation in urban areas. 13 Acute exposure effects include nose and eye irritation, fatigue, headache, and nausea, whereas chronic exposures increase respiratory symptoms, reduce lung function, and change inflammatory profiles. 13 Diesel exhaust is also known to enhance allergic inflammation and due to its contribution to ambient particulate matter (PM) is likely associated with increased susceptibility to viral infection. To explore this hypothesis, a study was conducted exposing adults who were healthy or had allergic rhinitis to diesel exhaust before inoculation with LAIV. 14 Diesel exhaust exposure was associated with increased LAIV-induced eotaxin-1, eosinophil cationic protein, and influenza RNA in nasal cells, supporting previous literature and animal studies, 15 suggesting that diesel exhaust can act as an allergic adjuvant, promoting inflammation and potentially reducing viral clearance, especially in individuals with underlying allergy. Wood smoke is a major and continually growing source of PM in the United States and globally as wildfire events occur with increasing frequency. 16 Furthermore, a substantial percentage of the world population uses wood and biomass for heating or cooking, resulting in up to 30% of ambient fine PM in some areas of the United States during the winter months, and greater percentages in developing countries. [17] [18] [19] Exposure to wood smoke and biomass is associated with increased morbidity, including reduced lung function, upper respiratory tract symptoms, and inflammation, and mortality due to respiratory infection in many populations. [20] [21] [22] [23] [24] [25] The effects of wood smoke on LAIV were examined in healthy study participants exposed to either 500 mg/m 3 of wood smoke particulate or filtered air for 2 hours. 26 LAIV-induced IFN-g-induced protein 10 levels, as measured in nasal lavage, were suppressed in the nasal mucosa of all participants. Furthermore, an Exposure by Sex interaction was observed, with males showing greater inflammation-related gene expression, whereas in females, host defense-related gene expression was mildly decreased in nasal lavage fluid cells. These results support sex-specific responses to viral infection, which can be augmented with the additional interaction of pollutant exposure, in this case wood smoke. Epidemiological evidence repetitively links tobacco smoke exposure to increased risk of viral infection. 7, 27 To better understand the mechanisms of this association between tobacco product use and respiratory infection, various studies in users and nonusers have been conducted using the LAIV model. An observational cohort study was first conducted to understand differential responses in active cigarette smokers, nonsmokers, and those exposed to secondhand smoke after LAIV inoculation. 28 In this initial study, nasal lavage fluid IL-6 response was significantly suppressed in active smokers, along with decreased median IFN-g-induced protein 10, and IFN-g, whereas viral RNA levels were significantly increased in smokers as compared with nonsmokers. Individuals exposed to secondhand smoke generated responses that were intermediate between active smokers and nonsmokers, suggesting mechanisms for increased susceptibility to infection in tobacco product users and those exposed secondhand. Most recently, the LAIV model has been used to investigate potential mechanisms by which electronic cigarettes (e-cigarettes) may affect susceptibility to viral infection. 29 Because e-cigarettes were deemed a tobacco product 30 and, similar to cigarettes, contain nicotine and other additives, it was hypothesized that e-cigarettes may also increase susceptibility to respiratory viral infection. Thus, a cohort of cigarette smokers, e-cigarette users, and nonsmokers was inoculated with LAIV and monitored for effects on immune gene expression and antibody response. 29 Overall, there was substantial downregulation of critical immune genes in nasal biopsy samples, especially in e-cigarette users compared with nonsmokers. In particular, altered host defense mediators, IFN-g, IL-6, and IL12p40, were found in cigarette smokers and e-cigarette users when compared with nonsmokers. It was also found that nasal mucosal anti-LAIV IgA levels were significantly lower in cigarette smokers and e-cigarette users than in nonsmokers, demonstrating that e-cigarette use can alter response to viral infection, affecting host defense mediators and antibody production. Although the LAIV model has provided significant insight into the effects of inhaled pollutants on viral infection in humans in vivo, there are still many aspects of the effects of inhaled pollutant exposures on response to viral infection that are poorly understood. For example, many of these studies focus on acute exposures, whereas most human population is exposed chronically to inhaled pollutants, which may impact response to viral infection. Furthermore, only a limited number of model pollutants have been investigated and more research is needed on pollutants such as PM, gaseous pollutants such as NO 2 and ozone, and pollutant mixtures. The field would also benefit from longer follow-up periods in LAIV-based studies to understand the effect of pollutant exposure on adaptive immunity and studies that include the exploration of potential targeted interventions to prevent pollution-induced effects (eg, therapeutics, dietary, and use of personal and household filtration devices). 31,32 Many viral pathogens, including influenza, parainfluenza, and coronaviruses, including SARS-CoV-2, depend in part on proteolytic activation of the virus, regulating the ability of the virus to enter the host cell 33, 34 (Fig 1, step 1 ). For example, attachment and subsequent entry of SARS-CoV-2 into the host cell occurs via binding of the virus spike (S)-protein to angiotensinconverting enzyme 2 (ACE2) receptors expressed on many different cell types. The S protein has 2 functional domains: the S1 domain, which binds to the ACE2 receptor, and the S2 domain, which mediates the fusion between the virus and the host cell membrane. Proteolytic cleavage of the S protein is required to enable the S2 domain to become active. 35 This can be accomplished by a number of proteases, such as transmembrane protease, serine 2 (TMPRSS2), furin, cathepsins, neutrophil elastase (reviewed in El-Shimy et al 35 and Meyer and Jaspers 36 ), and potentially other proteases that prime and regulate viral entry of SARS-CoV-2 into the host cell. Human nasal and bronchial mucosa abundantly express ACE2 and are rich sources of proteases, such as TMPRSS2, furin, cathepsins, and other proteases. 33, [37] [38] [39] [40] Inhibition of protease activity in the respiratory mucosa has been explored as a therapeutic target, but can also be regulated endogenously by antiproteases, such as alpha 1 antitrypsin and secretory leukocyte protease inhibitor. Pollutant exposure has been shown to dysregulate the protease/antiprotease balance in the respiratory mucosa. For example, exposure to ozone increases secreted levels of TMPRSS2 and decreases levels of secretory leukocyte protease inhibitor, which was linked to increased viral entry of influenza virus. 41 Similarly, analysis of lung tissue from mice chronically exposed to ozone showed elevated expression of Tmprss2. 42 Many pollutants also enhance neutrophil elastase levels in the respiratory tract. The effects of inhaling cigarette smoke on protease/antiprotease balance in the respiratory mucosa are well established and causally linked to smoking-related lung diseases. [43] [44] [45] [46] Some groups have shown that expression of ACE2 and TMPRSS2 is similar in bronchial epithelial cells from current and never smokers, 47 whereas others have observed significant increases in either ACE2 and/or TMPRSS2 in cells from smokers. [48] [49] [50] Expression and activity of antiproteases, including secretory leukocyte protease inhibitor and alpha 1 antitrypsin, are also modified by smoking. 51, 52 Controlled exposures to model particulate air pollutants, such as diesel exhaust, 53 demonstrate increased expression of ACE2 and TMPRSS2 in human pluripotent stem cell-derived alveolar epithelial cells and alveolar organoids. Expression of ACE2 and TMPRSS2 might be regulated by Randomized, placebocontrolled study 500 mg/m 3 wood smoke particles or filtered air and LAIV inoculation Healthy adults IP-10 suppression in nasal mucosa in all exposed participants. Increased inflammatory-related gene expression in male exposed subjects and decreased host defense-related gene expression in female exposed subjects, compared with controls several consensus motifs for binding of the aryl hydrocarbon receptor, a common pathway activated by ambient air pollutants. 54, 55 The pulmonary surfactant, which lines the alveoli, serves as another line of defense against pathogens. Surfactant proteins (SPs) SP-A and SP-D assist in the clearance of bacteria and virus from the lung by directly opsonizing pathogens and enhancing their uptake by phagocytic immune cells. 56 Air pollutants such as PM and ozone as well as cigarette smoke have all been shown to interfere with the pulmonary surfactant. Specifically, ozone A1AT, Alpha 1 antitrypsin; AhR, aryl hydrocarbon receptor; BAL, bronchoalveolar lavage; HA, hemaggluttinin; HAT, human airway trypsin-like protease; IL-12p40, IL-12 subunit p40; IP-10, IFN-g-induced protein 10; NF-kB, nuclear factor kappa-light-chain-enhancer; NK, natural killer; ROR, receptor tyrosine kinase-like orphan receptor; RSV, respiratory syncytial virus; SLPI, secretory leukocyte protease inhibitor; STAT1, signal transducer and activator of transcription 1; TLR, Toll-like receptor. exposure decreases the phagocytic index of SP-A. 57 Diesel exhaust exposures dampened production of SP-D in allergensensitized individuals 58 and decreased SP-A and SP-D levels in mice, which was associated with increased influenza infections. 59 Finally, smokers, who are generally more susceptible to viral infections, have been reported to have less SP-A and SP-D present in bronchoalveolar lavage fluid compared with nonsmokers. 60 Once the virus enters the host cell, it is met with an organized antiviral host defense response, aimed at limiting the replication and release of new virions. 61 Infection triggers a rapid antiviral signaling pathway, beginning by activation of Toll-like receptors and resulting in the secretion of type I and type III interferons (IFNs) by the infected cell (Fig 1, steps 2-4) . Type I IFNs act in either an autocrine fashion or a paracrine fashion to stimulate the second wave of antiviral responses by activating the Janus kinase/signal transducer and activator of transcription signaling pathway and resulting in the expression of numerous IFNstimulated genes, whose role is to shut off or limit the replication of the virus in the host cell. 61 Similarly, type III IFNs, which are massively induced in respiratory epithelium on viral infection, activate Janus kinase/signal transducer and activator of transcription pathways to upregulate expression of immunomodulatory genes. 62 The effects of cigarette smoke exposure on antiviral host defense responses have been well studied, including recent studies demonstrating that exposure to cigarette smoke increases SARS-CoV-2 infection. 63 Effects of smoking include inhibition of Toll-like receptor activation, inactivation of nuclear factor kappa-light-chain-enhancer response, 64 epigenetic modulation of IFN response genes, 65 decreased expression of antiviral host defense genes, 66, 67 and decreased activation of IFN signaling pathways. 68, 69 Much less is known about ambient air pollutant effects on antiviral host defense responses. In the context of model air pollutant exposures, such as diesel exhaust and ozone, type I IFN responses were not affected by the exposures and increase with the level of infection. 7, 41, 70 In contrast, exposure to ambient PM suppresses virus-induced IFN-b expression in macrophages and epithelial cells. 41, 71, 72 Hence, whether and to what extent inhalation of ambient air pollutants modulates the ability to mount an effective antiviral host defense response needs to be further explored. Control of viral replication and pathogenesis depends on the ability of infected epithelial cells to limit spread of virus to neighboring cells while also increasing the production of cytokines and chemokines that recruit virus-specific immune effector cells to the infection site (Fig 1, step 5) . However, increases in pollutant-induced oxidative stress, which has been observed after diesel exhaust, PM, and ozone exposures, 73 can result in epithelial cell damage, limiting their capacity to function. 7, 9, 74, 75 Release of cytokines and chemokines, which recruit and activate immune cells, is also critical for coordinated innate and adaptive immune responses. 76 These include IFN-inducible cytokines, such as C-X-C motif chemokine ligand (CXCL)9/ monokine induced by IFN-g, CXCL10/IFN-g-induced protein 10, and CXCL12/stromal cell-derived factor 1 alpha, as well as neutrophil chemokines (CXCL8/IL-8 and CXCL1/GROa), lymphocyte chemoattractants (C-C motif chemokine ligand 5/ RANTES and CXCL16), and monocyte chemoattractants (C-C motif chemokine ligand 2/monocyte chemoattractant protein 1). The production of many of these inducible cytokines has been shown to be altered by pollutant exposure including tobacco products and wood smoke, indicating potentially impaired coordinated immune responses to infection. 26, 29 Along with reduced production of important immune signaling molecules, the functions of innate immune cells recruited by these signals are also impaired by inhaled pollutant exposure (Fig 1, step 6 ). For example, neutrophil phagocytosis and neutrophil extracellular trap formation, macrophage phagocytosis, and cytotoxic natural killer cell responses were all altered ex vivo with e-cigarette exposure. 77 Furthermore, gaseous pollutants such as ozone have been shown to alter macrophage function 7,74 while diesel exhaust particle exposure induces apoptosis of human macrophages at levels that cause minimal cytotoxicity to bronchial epithelial cells. 78 In addition, critical to a coordinated response to future viral infection is the production of virus-specific antibodies, which have recently been shown to be impaired with exposure to e-cigarettes in an LAIV inoculation model. 29 Overall, there are a myriad of ways throughout the viral infection cycle that inhaled pollutants can alter appropriate host defense responses, which are summarized in Table I . These alterations may contribute to observed increases in rates of viral infection and associated morbidity and mortality in areas of the world with high ambient pollution levels or in people using tobacco products. There are limitations to using human controlled exposure models, such as the limitation of LAIV only permitting viral replication in the nose and limiting our ability to study effects that may differ in the lower airway. [79] [80] [81] Despite these limitations, this experimental design does allow for the examination of the effects of pollutants and viral infection in a model with immediate public health relevance. Controlled in vivo and in vitro exposure models have allowed us to begin to explore these mechanisms, yet as new viruses and pollutants continue to emerge, further study is needed. The current coronavirus disease 2019 pandemic has illustrated that better understanding of potential interactions between pollutants or toxicant exposures and effects on antiviral host defense responses is needed. Air pollution, cardiovascular endpoints and susceptibility by stress and material resources: a systematic review of the evidence A joint ERS/ATS policy statement: what constitutes an adverse health effect of air pollution? An analytical framework World Health Organization. Global database of household air pollution measurements Sex and gender differences in the susceptibility to environmental exposures Respiratory sex differences in response to smoke exposure Estimates of the global, regional, and national morbidity, mortality, and aetiologies of lower respiratory infections in 195 countries, 1990-2016: a systematic analysis for the Global Burden of Disease Study Air pollution and respiratory viral infection The effects of air pollution on COVID-19 infection and mortality -a review on recent evidence Effects of air pollutants on the transmission and severity of respiratory viral infections The effect of an experimental rhinovirus 16 infection on bronchial lavage neutrophils Live attenuated influenza vaccine (FluMistÒ; Fluenzä): a review of its use in the prevention of seasonal influenza in children and adults SARS-CoV-2 reverse genetics reveals a variable infection gradient in the respiratory tract Health effects of diesel exhaust emissions Diesel exhaust exposure and nasal response to attenuated influenza in normal and allergic volunteers Exacerbation of allergic inflammation in mice exposed to diesel exhaust particles prior to viral infection Particulate air pollution from wildfires in the western US under climate change Associations between ambient wood smoke and other particulate pollutants and biomarkers of systemic inflammation, coagulation and thrombosis in cardiac patients Indoor air pollution in developing countries and acute lower respiratory infections in children Domestic wood burning and PM2.5 trace elements: personal exposures, indoor and outdoor levels Wood smoke exposure, poverty and impaired lung function in Malawian adults Indoor pollution as an occupational risk factor for tuberculosis among women: a population-based, gender oriented, case-control study in Southern Mexico Indoor air pollution from biomass combustion and acute respiratory illness in preschool age children in Zimbabwe Lung function indices of children exposed to wood smoke in a fishing port in South-South Nigeria A case report of cor pulmonale in a woman without exposure to tobacco smoke: an example of the risks of indoor wood burning Cooking with biomass stoves and tuberculosis: a case control study Wood smoke exposure alters human inflammatory responses to viral infection in a sex-specific manner: a randomized, placebo-controlled study Cigarette smoking and infection Tobacco smoke exposure and altered nasal responses to live attenuated influenza virus Electronic-cigarette use alters nasal mucosal immune response to liveattenuated influenza virus. A clinical trial The FDA ''Deeming Rule'' and tobacco regulatory research Understanding the asthmatic response to an experimental rhinovirus infection: exploring the effects of blocking IgE Notch4 signaling limits regulatory T-cell-mediated tissue repair and promotes severe lung inflammation in viral infections Activation of influenza viruses by proteases from host cells and bacteria in the human airway epithelium Proteolytic activation of SARS-CoV-2 spike at the S1/S2 boundary: potential role of proteases beyond furin Targeting host cell proteases as a potential treatment strategy to limit the spread of SARS-CoV-2 in the respiratory tract Respiratory protease/antiprotease balance determines susceptibility to viral infection and can be modified by nutritional antioxidants SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis TMPRSS2 and furin are both essential for proteolytic activation of SARS-CoV-2 in human airway cells Exposure to ozone modulates human airway protease/antiprotease balance contributing to increased influenza A infection Ozone exposure upregulates the expression of host susceptibility protein TMPRSS2 to SARS-CoV-2 Chronic E-cigarette use increases neutrophil elastase and matrix metalloprotease levels in the lung Comparison of proteolytic enzyme activity in pulmonary alveolar macrophages and blood leukocytes in smokers and nonsmokers Cathepsin L activity is increased in alveolar macrophages and bronchoalveolar lavage fluid of smokers Increased neutrophil elastase activity in cigarette smokers Smoking and COVID-19: similar bronchial ACE2 and TMPRSS2 expression and higher TMPRSS4 expression in current versus never smokers Tobacco smoking increases the lung gene expression of ACE2, the receptor of SARS-CoV-2 Airways expression of SARS-CoV-2 receptor, ACE2, and TMPRSS2 is lower in children than adults and increases with smoking and COPD Smoking upregulates angiotensin-converting enzyme-2 receptor: a potential adhesion site for novel coronavirus SARS-CoV-2 (Covid-19) Regulation and activity of secretory leukoprotease inhibitor (SLPI) is altered in smokers Effect of cigarette smoke exposure and structural modifications on the a-1 antitrypsin interaction with caspases Diesel particulate matter 2.5 induces epithelial-to-mesenchymal transition and upregulation of SARS-CoV-2 receptor during human pluripotent stem cell-derived alveolar organoid development Assessing the consequences of environmental exposures on the expression of the human receptor and proteases involved in SARS-CoV-2 cell-entry Air particulate matter induced oxidative stress and inflammation in cardiovascular disease and atherosclerosis: the role of Nrf2 and AhR-mediated pathways The role of surfactant in lung disease and host defense against pulmonary infections Impact of ozone exposure on the phagocytic activity of human surfactant protein A (SP-A) and SP-A variants Particle depletion of diesel exhaust restores allergen-induced lung-protective surfactant protein D in human lungs Diesel exhaust enhanced susceptibility to influenza infection is associated with decreased surfactant protein expression Decreased contents of surfactant proteins A and D in BAL fluids of healthy smokers The airway epithelium: soldier in the fight against respiratory viruses The key roles of interferon lambda in human molecular defense against respiratory viral infections Direct exposure to SARS-CoV-2 and cigarette smoke increases infection severity and alters the stem cell-derived airway repair response Cigarette smoke extracts inhibit TLR ligand-induced inflammation via inactivation of NF-kB and activation of histone deacetylase in macrophages (INM7P.353) DNA methylation in nasal epithelial cells from smokers: identification of ULBP3-related effects Cigarette smoke modulates expression of human rhinovirus-induced airway epithelial host defense genes Cigarette smoke suppresses type I interferon-mediated antiviral immunity in lung fibroblast and epithelial cells Inhibition of IFN-gammadependent antiviral airway epithelial defense by cigarette smoke Cigarette smoke dampens antiviral signaling in small airway epithelial cells by disrupting TLR3 cleavage Diesel exhaust enhances influenza virus infections in respiratory epithelial cells compromises antiviral immunity in influenza infection by inhibiting activation of NLRP3 inflammasome and expression of interferon-b Particulate matter (PM10) enhances RNA virus infection through modulation of innate immune responses Oxidative stress and air pollution exposure Air pollution and its effects on the immune system Role of oxidative stress on diesel-enhanced influenza infection in mice Leukocyte trafficking to the lungs and beyond: lessons from influenza for COVID-19 Flavored e-cigarette liquids and cinnamaldehyde impair respiratory innate immune cell function Chemicals in diesel exhaust particles generate reactive oxygen radicals and induce apoptosis in macrophages Nasal epithelial cells as surrogates for bronchial epithelial cells in airway inflammation studies Brushed nasal epithelial cells are a surrogate for bronchial epithelial CFTR studies Comparison of nasal and bronchial epithelial cells obtained from patients with COPD