Abstract
Viral pneumonias remain global health threats, as exemplified in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, requiring novel treatment strategies both early and late in the disease process. We have reported that mice treated before or soon after infection with a combination of inhaled Toll-like receptor (TLR) 2/6 and 9 agonists (Pam2-ODN) are broadly protected against microbial pathogens including respiratory viruses, but the mechanisms remain incompletely understood. The objective of this study was to validate strategies for immune modulation in a preclinical model of viral pneumonia and determine their mechanisms. Mice were challenged with the Sendai paramyxovirus in the presence or absence of Pam2-ODN treatment. Virus burden and host immune responses were assessed to elucidate Pam2-ODN mechanisms of action and to identify additional opportunities for therapeutic intervention. Enhanced survival of Sendai virus pneumonia with Pam2-ODN treatment was associated with reductions in lung virus burden and with virus inactivation before internalization. We noted that mortality in sham-treated mice corresponded with CD8+ T-cell lung inflammation on days 11–12 after virus challenge, after the viral burden had declined. Pam2-ODN blocked this injurious inflammation by minimizing virus burden. As an alternative intervention, depleting CD8+ T cells 8 days after viral challenge also decreased mortality. Stimulation of local innate immunity within the lungs by TLR agonists early in disease or suppression of adaptive immunity by systemic CD8+ T-cell depletion late in disease improves outcomes of viral pneumonia in mice. These data reveal opportunities for targeted immunomodulation to protect susceptible human subjects.
Keywords: immunomodulation, immunopathology, CD8+ T cells, viral pneumonia, inducible epithelial resistance
Viruses are the most frequent cause of community-acquired pneumonia in children and adults, resulting in significant morbidity in vulnerable subjects and exerting a tremendous healthcare burden (1–3). Seasonal influenza and emergent pandemic viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), inflict particular mortality in susceptible individuals, with clinicians frequently lacking effective interventions to improve patient outcomes (4–7). Moreover, in addition to causing acute disease, respiratory virus infections are often complicated by chronic lung pathologies, such as exacerbation and progression of asthma and chronic obstructive pulmonary disease (8–10). Therefore, development of novel therapeutic antiviral strategies is required to effectively prevent and treat respiratory infections and their associated chronic complications (11, 12).
Although lung epithelial cells are the principal targets of most respiratory viruses (13), there is expanding evidence that lung epithelia themselves are capable of generating antimicrobial responses (10, 14, 15). We hypothesized that lung epithelial cells can be harnessed to control virus replication, thereby enhancing acute survival and reducing chronic complications of virus infections (16–19). Our group has previously described the phenomenon of inducible epithelial resistance wherein the lungs’ mucosal defenses can be broadly stimulated to protect against a wide range of respiratory pathogens, including viruses (16–21). This protection is induced by a single inhalation of a combination treatment consisting of Toll-like receptor (TLR) 2/6 and 9 agonists (Pam2-ODN) shortly before or after viral challenge. Although no individual leukocyte populations have been identified as critical for Pam2-ODN–induced resistance, lung epithelial cells are essential to the inducible antiviral response (16). More recently, we have shown prevention of chronic virus-induced asthma in mice treated with Pam2-ODN, but we have not clarified the antiviral mechanisms (22).
In this study, we investigated the mechanisms of Pam2-ODN–enhanced mouse survival of pneumonia caused by a paramyxovirus, Sendai virus (SeV). We found that Pam2-ODN treatment not only reduced lung SeV burden but also decreased epithelial cell injury and host immunopathologic leukocyte responses to SeV infections. Although CD8+ T cells are known to contribute to antiviral immunity, it is shown here that CD8+ T cells contribute substantially to mortality, and this effect can be prevented by Pam2-ODN treatment early in the course of infection or CD8+ depletion late in the course. Furthermore, we demonstrate antiviral mechanisms of inducible epithelial resistance, where virus particles are inactivated before internalization by their epithelial targets.
Some of the results of these studies have been previously reported in the form of a preprint (23).
Methods
Mice
All in vivo experiments were performed using 6- to 10-week-old C57BL/6J mice. All groups in each individual experiment were composed of a single sex. Both sexes were tested in replicates of each experimental model. No sex-dependent differences were detected in any recorded outcome. Mice were handled according to the Institutional Animal Care and Use Committee of MD Anderson Cancer Center, protocol 00000907-RN01.
Cells
Mouse lung epithelial (MLE-15) cells were kindly provided by Jeffrey Whitsett, Cincinnati Children’s Hospital Medical Center. Mouse tracheal epithelial cells were harvested and cultured as previously described (20, 24). See the online supplement for additional details.
TLR Treatments and Viral Challenges
Cells were treated with Pam2CSK4 (2.2 μM) and ODN M362 (0.55 μM) as previously described (20, 21). For all in vivo experiments, mice were pretreated with 10 ml of Pam2CSK4 (4 μM) and ODN M362 (1 μM) by nebulization 1 day before SeV challenge as previously described (20, 21). For in vitro challenges, SeV at multiplicity of infection = 1 was used. Unless otherwise stated, mice were challenged with 1 × 108 plaque-forming units inserted into the oropharynx as described (22). See the online supplement for additional details.
Flow Cytometry
Single cells from disaggregated lungs or cell culture were stained as indicated in the antibody table (Table 1), fixed, and acquired on a BD LSRII (BD Biosciences). See the online supplement for additional details.
Table 1.
Antibodies
Target Epitope | Vendors | Catalog Numbers |
---|---|---|
CD3 | Tonbo | 65-0031-U100 |
CD4 | Tonbo | 60-0042-U100 |
CD8 | Tonbo | 25-0081-U100 |
Live dead | Tonbo | 13-0870-T500 |
CD25 | Biolegend | 102038 |
Foxp3 Treg kit | eBiosciences | 72-5775 |
CD8-depleting Ab | Bioxell | BE0223-A025 |
CD19 | Biolegend | 115507 |
B220 | BD Biosciences | 562922 |
Anti-SeV Ab | MBL International | PD029 |
Ki67 | Invitrogen | MA5-14520 |
cCasp3 | Cell signaling | 9662S |
Definition of abbreviations: Ab = antibody; cCasp3 = cleaved caspase 3; SeV = Sendai virus.
Epithelial Proliferation Assays
Epithelial proliferation was determined by staining lung sections for 5-ethynyl-2′deoxyuridine (EdU) 24 hours after intraperitoneal injection. See the online supplement for details.
CD8+ T-Cell Depletion
Anti–CD8-β antibody (200 μg/mouse, clone 53-5.8; Bioxell) was delivered to mice intraperitoneally at indicated time points. CD8+ T-cell depletion was confirmed by flow cytometry analysis 24 to 48 hours after depletion.
Viral Burden Quantification
Viral burden was determined by qRT-PCR of the Sendai Matrix (M) protein normalized to host housekeeping gene 18SRNA. For in vivo experiments, mouse lungs were collected 5 days after SeV challenge. For in vitro experiments, cell lysates were collected 24 hours after infection, unless otherwise indicated. See the online supplement for additional details.
Viral Attachment Assays
For most enveloped viruses, internalization into epithelial cells is inhibited at 4°C without affecting viral binding to epithelial cells (25–27). MLE-15 cells were infected with SeV at 4°C for 4 hours, washed to remove unattached virus, and then assessed for uninternalized SeV burden using immunofluorescence or flow cytometry. See the online supplement for additional details.
Results
Enhanced Mouse Survival of SeV Infection by Pam2-ODN Treatment
Aerosolized Pam2-ODN treatment 1 day before SeV challenge increased mouse survival of SeV challenge (Figure 1A), similar to the protection observed against lethal influenza pneumonia (16, 19, 20). The survival benefit was associated with reduced lung SeV burden, as measured by SeV M gene expression (Figure 1B). Investigating the natural progression of infection revealed that SeV lung burden was maximal on day 5 and gradually decreased until falling below the limit of quantification by day 11 (Figure 1C). Pam2-ODN pretreatment reduced SeV burden on all assessed days (Figure 1C). Although the lethality of SeV infection was exquisitely dependent on the inoculum size, we strikingly found that peak mortality paradoxically occurred around days 10 to 12 after infection irrespective of inoculum size, despite the fact that SeV is essentially undetectable that long after challenge (Figures 1A, 1C, and 1D).
Figure 1.
Pam2-ODN enhances mouse survival of Sendai virus (SeV) infection and reduces lung virus burden. (A) Survival of mice treated with PBS or combination therapy with Pam2CSK4 and ODN M362 (Pam2-ODN) day before SeV virus challenge. (B) Mouse lung SeV burden 5 days after infection assessed by qPCR for M gene (RQ to 18S) relative to 18S. (C) Time course of lung SeV burden in mice treated with PBS or Pam2-ODN. (D) SeV inoculum–dependent mouse survival. Data shown are from one representative experiment of three independent experiments. n = 10 mice per group in survival plots (A and D). n = 4 mice/group in virus burden experiments. *P < 0.05, Student’s t test and **P < 0.005, Log-rank (Mantel-Cox) test. LOQ = limit of quantification; M = Sendai Matrix; pfu = plaque-forming unit; RQ = relative quantification.
Pam2-ODN Treatment Attenuates SeV-induced Epithelial Injury
This temporal dissociation between peak virus burden and peak mortality led to the hypothesis that SeV-induced mortality may not be exclusively driven by excessive virus burden but may also result from untoward SeV-induced host immune responses. Therefore, the acute changes in mouse lungs following SeV infection were characterized. We found increases in lung epithelial cleaved caspase 3, a marker for programmed cell death, on days 7 to 11 after SeV infection (Figure 2A, upper panel). Virus infection–related epithelial cell injury and death is typically associated with proliferative repair mechanisms (28, 29). Staining the infected mouse lung tissue for Ki67 and EdU revealed maximum signals for both markers in the second week after infection (Figure 2B–2E, upper panel). These events of lung epithelial cell death and proliferation coincided with the peak of mortality (day 12, Figure 1E). Furthermore, hematoxylin and eosin staining of lung tissues infected with SeV showed profound increases in inflammatory cells from days 7 to 10 with evidence of damaged airway and parenchymal tissue (Figure 2F). However, Pam2-ODN pretreatment of mice reduced epithelial cell injury and proliferation (Figures 2A–2E, lower panel). This temporal association of epithelial injury and death after viral clearance supported our hypothesis that mouse mortality caused by SeV infection is due in part to the host immune response to SeV infections.
Figure 2.
Pam2-ODN pretreatment reduces epithelial cell death and proliferation during acute SeV infection. (A) cCasp3- or (B) Ki67-positive cells in mouse lung epithelium after SeV infection (upper panel) with or without Pam2-ODN treatment (lower panel, day 10). EdU-positive cells in (C) axial airways, (D) small airways, and (E) parenchyma after SeV infection (upper panel) with or without Pam2-ODN (lower panel, day 10). (F) Mouse lung histology following SeV challenge with or without Pam2-ODN. n = 5 mice per condition. Data shown are from one representative experiment of two independent experiments. Scale bars, 100 μm. *P < 0.05, Student’s t test. cCasp3 = cleaved caspase 3; EdU = 5-ethynyl-2′deoxyuridine.
Pam2-ODN Attenuates SeV-induced Lymphocytic Lung Inflammation
To explore this hypothesis, the host leukocyte response to SeV infection was characterized. Differential Giemsa staining of BAL cells revealed increased neutrophils on days 2 to 5 and increased macrophages on days 5 to 8 (Figure 3A, left and middle panel, solid gray line) after SeV challenge. Congruent with our prior studies, inhaled treatment with Pam2-ODN in the absence of infection led to a rapid rise in neutrophils that was resolved within 5 days (Figure 3A, dashed line) (30). The neutrophil response to SeV challenge was modestly increased among mice pretreated with Pam2-ODN (Figure 3A, left panel, solid dark line). Pam2-ODN–treated, SeV-challenged mice showed almost no difference in macrophage number compared with PBS-treated, SeV-challenged mice (Figure 3A, middle panel, solid dark line). A rise in lymphocytes was observed on days 8 to 11 in PBS-treated, SeV-challenged mice (Figure 3A, right panel, solid gray line), temporally corresponding with peak mortality. However, Pam2-ODN–treated, SeV-challenged mice displayed significantly reduced lymphocyte numbers at every time point assessed (Figure 3A, right panel, solid dark line). The gating strategy for lymphocyte subsets by flow cytometry is shown in Figure E1 in the online supplement. A modest reduction in CD4+ T cells was observed in Pam2-ODN–treated, SeV-challenged mice compared with PBS-treated, SeV-challenged mice (Figure E2). We also found the percentage of CD19+ B220+ B cells was reduced after SeV infection in comparison with Pam2-ODN–treated and uninfected mice (Figure E2), as has been seen with other viral models (31, 32). However, the biggest difference between groups was in CD8+ T cells, with Pam2-ODN–treated, SeV-challenged mice displaying a significantly lower number and percentage of CD8+ T cells than PBS-treated, SeV-challenged mice (Figures 3B and 3C). Because the greatest difference after Pam2-ODN treatment was in CD8+ T-cell levels and there was a tight correlation between peak mortality and the increase in lung CD8+ T cells on days 8 to 11, we investigated the role of CD8+ T cells in SeV-induced mortality.
Figure 3.
Pam2-ODN pretreatment reduces SeV-induced lung CD8+ T cells. (A) Differential Giemsa staining of BAL cells from mice challenged with SeV with or without Pam2-ODN pretreatment. (B) Flow cytometry for CD8+ T cells from disaggregated mouse lungs 11 days after SeV infection with or without Pam2-ODN. (C) Lung CD8+ T cells 11 days after SeV challenge in mice pretreated with PBS or Pam2-ODN. Data shown are from one representative experiment of three independent experiments for A and of five independent experiments for B and C. *P < 0.05 compared with PBS+SeV, Student’s t test.
Depleting CD8+ T Cells after Viral Clearance Enhances Survival of SeV Infection
To understand the apparent contributions of host immunopathology to mouse outcomes, we depleted CD8+ T cells on day 8—after virus burden was substantially reduced but before peak mouse mortality (Figures 1 and 4A). Mice depleted of CD8+ T cells displayed significantly enhanced survival of SeV challenge compared with mice with intact CD8+ T cells (Figure 4B). Depletion of CD8+ T cells was confirmed by flow cytometry in disaggregated lung cells 10 days after SeV challenge (Figures 4C and E3A). We also assessed lung injury by hematoxylin and eosin staining of lung tissue 10 days after SeV challenge and found increased inflammation and epithelial cell damage in undepleted mice compared with CD8+ T-cell–depleted mice (Figure 4D). This supported our hypothesis that CD8+ T cells contribute to fatal SeV-induced immunopathology.
Figure 4.
Pam2-ODN treatment reduces CD8+ T-cell–associated SeV-induced immunopathology. (A) Experimental outline, (B) survival, and (C) percentage of CD8+ T cells from disaggregated mouse lungs 10 days after SeV challenge following pretreatment with PBS or Pam2-ODN and with or without CD8+ T cells depleted on day 8 of SeV challenge. (D) Lung histology 10 days after SeV challenged with or without Pam2-ODN treatment and/or CD8+ T cells. Data shown are one representative experiment of two independent experiments. Scale bars, 100 μm. n = 16 mice/group for survival in experiment A and n = 4 mice/group in experiment B. ****P < 0.0001 compared with PBS in C, ***P < 0.0005 compared with PBS in B and C, †P < 0.05 compared with CD8 Ab-PBS and *P < 0.05 compared with PBS. Mouse survival analysis in B were analyzed using log-rank, Mantel-Cox test. One-way ANOVA with multiple comparisons was used in C. Ab = antibody.
To assess the role of CD8+ T cells throughout the course of infection, mouse CD8+ T cells were depleted before and during SeV challenge (Figures 4A, E3A, and E3B). This depletion resulted in significantly reduced survival of SeV infection (Figure E3C), compatible with the known antiviral functions of CD8+ T cells (33–35). However, it is notable that Pam2-ODN treatment still significantly enhanced survival of SeV challenge even in the absence of CD8+ T cells (Figure E3C). This finding was congruent with our previous studies showing Pam2-ODN–inducible resistance against bacterial pneumonia despite the lack of mature lymphocytes (Rag1−/−) (16).
Pam2-ODN Treatment Leads to Extracellular Inactivation of Virus Particles
As the antiviral protection consistently correlated with reduced viral burden in vivo, and as the reduced virus burden likely contributes to the reduced CD8+ T-cell levels, we sought to determine how Pam2-ODN–induced responses cause antiviral effects. Assessing the effect of Pam2-ODN on SeV burden in immortalized mouse epithelial cells (MLE-15) and primary mouse tracheal epithelial cells, we found that Pam2-ODN treatment reduced SeV burden at every time point measured, reflecting the inducible antiviral capacity of isolated epithelial cells (Figure E4). Furthermore, we investigated whether the principal Pam2-ODN effect occurred before (extracellular) or after (intracellular) virus internalization into their epithelial targets. SeV inoculation was performed at 4°C, preventing SeV internalization while allowing SeV attachment to epithelial cells (25–27). Using multiple methods to determine the effect of Pam2-ODN on SeV attachment, we found no differences in attachment (Figures 5A–5D). However, even though similar numbers of virus particles were attached to epithelial cells, when these attached virus particles were liberated from the epithelial cell targets, virus particles from Pam2-ODN–treated epithelial cells were less able to subsequently infect other naive epithelial cells (Figures 5E and 5F). As the number of attached virus particles was the same, this difference in SeV burden in cells that received liberated virus particles from PBS- versus Pam2-ODN–treated cells indicated that SeV is inactivated before epithelial internalization.
Figure 5.
Pam2-ODN inhibits SeV without altering attachment. (A) Flow cytometry to measure virus attachment to epithelial cells 4 hours after SeV challenge. (B) Percentage of SeV-positive epithelial cells from A. (C) Representative examples of immunofluorescence for virus attachment. (D) Mean fluorescence intensity of SeV-exposed epithelial cells 4 hours after SeV challenge. (E) Experimental outline showing viral attachment and prevention of virus internalization by epithelial cells. (F) SeV M gene expression in untreated MLE-15 cells (left) or primary tracheal epithelial cells (right) challenged with liberated virus (uninternalized virus particles) from cultures that had been pretreated with PBS or Pam2-ODN before SeV infection 24 hours after transfer of liberated virus to new cells. Data shown are one representative experiment of five independent experiments. *P < 0.05, Student’s t test. A.U. = arbitrary units; MFI = mean fluorescence intensity; MLE = mouse lung epithelial; mTEC = mouse tracheal epithelial cell ns = nonsignificant.
Discussion
In this study, we find that therapeutic stimulation of lung epithelial cells enhances mouse survival of acute SeV infections by both reducing the virus burden and attenuating host immunopathology. Although our group has previously demonstrated inducible resistance against multiple respiratory pathogens including viruses (16–21, 30), this study demonstrates for the first time when in the virus lifecycle the antiviral effects begin (i.e., before internalization).
We observed that PBS-treated mice died of SeV infection after they had almost completely cleared the virus. This observation prompted the hypothesis that host mortality is not the exclusive result of direct viral injury to the lungs but is primarily due to the host response to the virus infection. Supporting this, we found enhanced survival in mice depleted of CD8+ T cells 8 days after infection (Figures 4A and 4B), revealing the importance of balancing the dual functions of CD8+ T cells in antiviral immunity and in causing fatal immunopathology. Our findings further suggest that the surge in CD8+ T cells within the lungs after most virus has been cleared causes physiologic impairment via lung injury and cell death (Figure 4D). These findings are potentially informative in the context of treating viral pneumonia in human subjects, including those suffering lung injury associated with SARS-CoV-2. Of note, the median period from SARS-CoV-2 exposure to respiratory distress is 9 to 13 days, identical to the period from viral inoculation to death from pneumonitis in this study of SeV pneumonia in mice (36). Furthermore, it was recently reported that dexamethasone treatment confers a survival advantage to patients with coronavirus disease (COVID-19) requiring respiratory support and in those recruited after the first week of their illness (36). This suggests a stage of disease dominated by immunopathology (37), rather than active viral replication, similar to our observation in the SeV model 9 days after viral challenge. It will be interesting to test the effects of dexamethasone on CD8+ T-cell activity and host mortality in the SeV model.
Previous reports support the concept of counter-balanced immune protection and immunopathology by CD8+ T cells during virus infections (35, 38–42). These include studies showing that antigen-experienced memory CD8+ T cells enhance respiratory syncytial virus clearance but also mediate severe immunopathology (40, 43). Our findings are also congruent with reports on the role of CD8+ T cells in nonrespiratory viral infection models, such as in West Nile virus infection, where CD8+ T-cell–deficient mice display decreased mortality (41). Although these studies and ours reveal that the harmful effects of CD8+ T-cell–mediated immunopathology can supersede the benefits of T-cell–mediated viral clearance, the question arises of what might be the adaptive value of the vigorous late CD8+ T-cell response. A recent study showing that sites of viral RNA remnants following influenza infection are linked to chronic lung disease in mice may offer an answer (44). Thus, a trade-off may exist between the adaptive value of a vigorous CD8+ T-cell response to prevent chronic lung disease and the acute mortality it can cause. Manipulating this balance therapeutically will need to account for both the benefits and costs of the response.
Together, these observations demonstrate an advantage of early immune stimulation to enhance viral clearance and late immune suppression to prevent immunopathology and enhance overall outcome of respiratory infections. Based on this reasoning and evidence of protection against other coronaviruses (45, 46), clinical trials of the use of Pam2-ODN to prevent or treat early COVID-19 have been launched (NCT04313023, NCT04312997), and we suggest that therapeutic targeting of CD8+ T cells later in COVID-19 be considered.
Although the CD8+ T-cell depletion studies enhanced our understanding of immunopathology in virus infections, much of the survival benefit against SeV infection was mediated by rapid antiviral effects induced by Pam2-ODN. This led us to investigate the mechanisms of these inducible antiviral effects. Given the multiple steps in the virus life cycle, it was not known at what stage Pam2-ODN exerted its antiviral effect. Exploring this, we found no differences in the attachment of SeV to epithelial cells induced by Pam2-ODN treatment (Figures 5A–5D). However, attached virus particles that were liberated from Pam2-ODN–treated cells retained less infective capacity when added to naive epithelial cells, revealing preinternalization virus inactivation by Pam2-ODN treatment (Figures 5E and 5F).
Conclusions
Taken together, these findings provide mechanistic insights into the antiviral responses generated by the lung epithelium and the prevention of host immunopathology that may inform future therapeutics to target immunomodulation as a means to improve the survival of respiratory infections in vulnerable populations.
Supplementary Material
Footnotes
Supported by U.S. National Institutes of Health grants R01 HL117976, DP2 HL123229, and R35 HL144805 (S.E.E.).
Author Contributions: S.W. designed and performed the experiments, analyzed the data, and wrote the manuscript. J.R.F., A.M.J., D.L.G., and J.P.G performed experiments. M.J.T. and B.F.D. conceptualized the project and critically reviewed the data. S.E.E. conceptualized the project, designed experiments, provided critical evaluation of data, and edited the manuscript.
This article has a data supplement, which is accessible from this issue’s table of contents at www.atsjournals.org.
Originally Published in Press as DOI: 10.1165/rcmb.2020-0241OC on August 27, 2020
Author disclosures are available with the text of this article at www.atsjournals.org.
References
- 1.Luckhaupt SE, Sweeney MH, Funk R, Calvert GM, Nowell M, D’Mello T, et al. Influenza-associated hospitalizations by industry, 2009-10 influenza season, United States. Emerg Infect Dis. 2012;18:556–562. doi: 10.3201/eid1804.110337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Jain S, Self WH, Wunderink RG, Fakhran S, Balk R, Bramley AM, et al. CDC EPIC Study Team. Community-acquired pneumonia requiring hospitalization among U.S. Adults. N Engl J Med. 2015;373:415–427. doi: 10.1056/NEJMoa1500245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jain S, Williams DJ, Arnold SR, Ampofo K, Bramley AM, Reed C, et al. CDC EPIC Study Team. Community-acquired pneumonia requiring hospitalization among U.S. children. N Engl J Med. 2015;372:835–845. doi: 10.1056/NEJMoa1405870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Fauci AS, Lane HC, Redfield RR. Covid-19: navigating the uncharted. N Engl J Med. 2020;382:1268–1269. doi: 10.1056/NEJMe2002387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Taubenberger JK, Kash JC, Morens DM. The 1918 influenza pandemic: 100 years of questions answered and unanswered. Sci Transl Med. 2019;11:eaau5485. doi: 10.1126/scitranslmed.aau5485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wang X, Li Y, O’Brien KL, Madhi SA, Widdowson MA, Byass P, et al. Respiratory Virus Global Epidemiology Network. Global burden of respiratory infections associated with seasonal influenza in children under 5 years in 2018: a systematic review and modelling study. Lancet Glob Health. 2020;8:e497–e510. doi: 10.1016/S2214-109X(19)30545-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Li Q, Guan X, Wu P, Wang X, Zhou L, Tong Y, et al. Early transmission dynamics in Wuhan, China, of novel coronavirus-infected pneumonia. N Engl J Med. 2020;382:1199–1207. doi: 10.1056/NEJMoa2001316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Busse WW, Lemanske RF, Jr, Gern JE. Role of viral respiratory infections in asthma and asthma exacerbations. Lancet. 2010;376:826–834. doi: 10.1016/S0140-6736(10)61380-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Folkerts G, Busse WW, Nijkamp FP, Sorkness R, Gern JE. Virus-induced airway hyperresponsiveness and asthma. Am J Respir Crit Care Med. 1998;157:1708–1720. doi: 10.1164/ajrccm.157.6.9707163. [DOI] [PubMed] [Google Scholar]
- 10.Holtzman MJ, Byers DE, Brett JA, Patel AC, Agapov E, Jin X, et al. Linking acute infection to chronic lung disease: the role of IL-33-expressing epithelial progenitor cells. Ann Am Thorac Soc. 2014;11:S287–S291. doi: 10.1513/AnnalsATS.201402-056AW. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Olin JT, Wechsler ME. Asthma: pathogenesis and novel drugs for treatment. BMJ. 2014;349:g5517. doi: 10.1136/bmj.g5517. [DOI] [PubMed] [Google Scholar]
- 12.Shaw DE, Green RH, Bradding P. Asthma exacerbations: prevention is better than cure. Ther Clin Risk Manag. 2005;1:273–277. [PMC free article] [PubMed] [Google Scholar]
- 13.Ibricevic A, Pekosz A, Walter MJ, Newby C, Battaile JT, Brown EG, et al. Influenza virus receptor specificity and cell tropism in mouse and human airway epithelial cells. J Virol. 2006;80:7469–7480. doi: 10.1128/JVI.02677-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Byers DE, Alexander-Brett J, Patel AC, Agapov E, Dang-Vu G, Jin X, et al. Long-term IL-33-producing epithelial progenitor cells in chronic obstructive lung disease. J Clin Invest. 2013;123:3967–3982. doi: 10.1172/JCI65570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Leiva-Juárez MM, Kolls JK, Evans SE. Lung epithelial cells: therapeutically inducible effectors of antimicrobial defense. Mucosal Immunol. 2018;11:21–34. doi: 10.1038/mi.2017.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Cleaver JO, You D, Michaud DR, Pruneda FA, Juarez MM, Zhang J, et al. Lung epithelial cells are essential effectors of inducible resistance to pneumonia. Mucosal Immunol. 2014;7:78–88. doi: 10.1038/mi.2013.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Duggan JM, You D, Cleaver JO, Larson DT, Garza RJ, Guzmán Pruneda FA, et al. Synergistic interactions of TLR2/6 and TLR9 induce a high level of resistance to lung infection in mice. J Immunol. 2011;186:5916–5926. doi: 10.4049/jimmunol.1002122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Evans SE, Tuvim MJ, Fox CJ, Sachdev N, Gibiansky L, Dickey BF. Inhaled innate immune ligands to prevent pneumonia. Br J Pharmacol. 2011;163:195–206. doi: 10.1111/j.1476-5381.2011.01237.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Tuvim MJ, Gilbert BE, Dickey BF, Evans SE. Synergistic TLR2/6 and TLR9 activation protects mice against lethal influenza pneumonia. PLoS One. 2012;7:e30596. doi: 10.1371/journal.pone.0030596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kirkpatrick CT, Wang Y, Leiva Juarez MM, Shivshankar P, Pantaleón García J, Plumer AK, et al. Inducible lung epithelial resistance requires multisource reactive oxygen species generation to protect against viral infections. MBio. 2018;9:e00696. doi: 10.1128/mBio.00696-18. 18.[Published erratum appears in MBio 10:e00496-19.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ware HH, Kulkarni VV, Wang Y, Pantaleón García J, Leiva Juarez M, Kirkpatrick CT, et al. Inducible lung epithelial resistance requires multisource reactive oxygen species generation to protect against bacterial infections. PLoS One. 2019;14:e0208216. doi: 10.1371/journal.pone.0208216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Goldblatt DL, Flores JR, Valverde Ha G, Jaramillo AM, Tkachman S, Kirkpatrick CT, et al. Inducible epithelial resistance against acute Sendai virus infection prevents chronic asthma-like lung disease in mice. Br J Pharmacol. 2020;177:2256–2273. doi: 10.1111/bph.14977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wali S, Flores JR, Jaramillo AM, Goldbatt DL, Pantaleón García J, Tuvim MJ, et al. Immune modulation to improve survival of respiratory virus infections in mice [preprint] bioRxiv. 2020 doi: 10.1165/rcmb.2020-0241OC. [accessed 2020 Apr 18]. Available from: https://www.biorxiv.org/content/10.1101/2020.04.16.045054v1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Leiva-Juárez MM, Ware HH, Kulkarni VV, Zweidler-McKay PA, Tuvim MJ, Evans SE. Inducible epithelial resistance protects mice against leukemia-associated pneumonia. Blood. 2016;128:982–992. doi: 10.1182/blood-2016-03-708511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tscherne DM, Jones CT, Evans MJ, Lindenbach BD, McKeating JA, Rice CM. Time- and temperature-dependent activation of hepatitis C virus for low-pH-triggered entry. J Virol. 2006;80:1734–1741. doi: 10.1128/JVI.80.4.1734-1741.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Haywood AM, Boyer BP. Sendai virus membrane fusion: time course and effect of temperature, pH, calcium, and receptor concentration. Biochemistry. 1982;21:6041–6046. doi: 10.1021/bi00267a003. [DOI] [PubMed] [Google Scholar]
- 27.Tai CJ, Li CL, Tai CJ, Wang CK, Lin LT. Early viral entry assays for the identification and evaluation of antiviral compounds. J Vis Exp. 2015;29:e53124. doi: 10.3791/53124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hines EA, Szakaly RJ, Leng N, Webster AT, Verheyden JM, Lashua AJ, et al. Comparison of temporal transcriptomic profiles from immature lungs of two rat strains reveals a viral response signature associated with chronic lung dysfunction. PLoS One. 2014;9:e112997. doi: 10.1371/journal.pone.0112997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Look DC, Walter MJ, Williamson MR, Pang L, You Y, Sreshta JN, et al. Effects of paramyxoviral infection on airway epithelial cell Foxj1 expression, ciliogenesis, and mucociliary function. Am J Pathol. 2001;159:2055–2069. doi: 10.1016/S0002-9440(10)63057-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Alfaro VY, Goldblatt DL, Valverde GR, Munsell MF, Quinton LJ, Walker AK, et al. Safety, tolerability, and biomarkers of the treatment of mice with aerosolized Toll-like receptor ligands. Front Pharmacol. 2014;5:8. doi: 10.3389/fphar.2014.00008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Bekker V, Scherpbier H, Beld M, Piriou E, van Breda A, Lange J, et al. Epstein-Barr virus infects B and non-B lymphocytes in HIV-1-infected children and adolescents. J Infect Dis. 2006;194:1323–1330. doi: 10.1086/508197. [DOI] [PubMed] [Google Scholar]
- 32.Shearer WT, Easley KA, Goldfarb J, Rosenblatt HM, Jenson HB, Kovacs A, et al. Prospective 5-year study of peripheral blood CD4, CD8, and CD19/CD20 lymphocytes and serum Igs in children born to HIV-1 women. The P(2)C(2) HIV Study Group. J Allergy Clin Immunol. 2000;106:559–566. doi: 10.1067/mai.2000.109433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Cannon MJ, Stott EJ, Taylor G, Askonas BA. Clearance of persistent respiratory syncytial virus infections in immunodeficient mice following transfer of primed T cells. Immunology. 1987;62:133–138. [PMC free article] [PubMed] [Google Scholar]
- 34.Graham BS, Bunton LA, Wright PF, Karzon DT. Role of T lymphocyte subsets in the pathogenesis of primary infection and rechallenge with respiratory syncytial virus in mice. J Clin Invest. 1991;88:1026–1033. doi: 10.1172/JCI115362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Schmidt ME, Varga SM. Cytokines and CD8 T cell immunity during respiratory syncytial virus infection. Cytokine. 2020;133:154481. doi: 10.1016/j.cyto.2018.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Gandhi RT, Lynch JB, Del Rio C. Mild or moderate Covid-19. N Engl J Med. 2020;383:1757–1766. doi: 10.1056/NEJMcp2009249. [DOI] [PubMed] [Google Scholar]
- 37.Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8:420–422. doi: 10.1016/S2213-2600(20)30076-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, et al. RECOVERY Collaborative Group. Dexamethasone in hospitalized patients with Covid-19: preliminary report. N Engl J Med. [online ahead of print] 17 Jul 2020; DOI: 10.1056/NEJMoa2021436. [Google Scholar]
- 39.Duan S, Thomas PG. Balancing immune protection and immune pathology by CD8(+) T-cell responses to influenza infection. Front Immunol. 2016;7:25. doi: 10.3389/fimmu.2016.00025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hou S, Doherty PC, Zijlstra M, Jaenisch R, Katz JM. Delayed clearance of Sendai virus in mice lacking class I MHC-restricted CD8+ T cells. J Immunol. 1992;149:1319–1325. [PubMed] [Google Scholar]
- 41.Schmidt ME, Knudson CJ, Hartwig SM, Pewe LL, Meyerholz DK, Langlois RA, et al. Memory CD8 T cells mediate severe immunopathology following respiratory syncytial virus infection. PLoS Pathog. 2018;14:e1006810. doi: 10.1371/journal.ppat.1006810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang Y, Lobigs M, Lee E, Müllbacher A. CD8+ T cells mediate recovery and immunopathology in West Nile virus encephalitis. J Virol. 2003;77:13323–13334. doi: 10.1128/JVI.77.24.13323-13334.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Connors TJ, Ravindranath TM, Bickham KL, Gordon CL, Zhang F, Levin B, et al. Airway CD8(+) T cells are associated with lung injury during infant viral respiratory tract infection. Am J Respir Cell Mol Biol. 2016;54:822–830. doi: 10.1165/rcmb.2015-0297OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Cannon MJ, Openshaw PJ, Askonas BA. Cytotoxic T cells clear virus but augment lung pathology in mice infected with respiratory syncytial virus. J Exp Med. 1988;168:1163–1168. doi: 10.1084/jem.168.3.1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Keeler SP, Agapov EV, Hinojosa ME, Letvin AN, Wu K, Holtzman MJ. Influenza A virus infection causes chronic lung disease linked to sites of active viral RNA remnants. J Immunol. 2018;201:2354–2368. doi: 10.4049/jimmunol.1800671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Evans SE, Xu Y, Tuvim MJ, Dickey BF. Inducible innate resistance of lung epithelium to infection. Annu Rev Physiol. 2010;72:413–435. doi: 10.1146/annurev-physiol-021909-135909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Evans SE, Tseng CK, Scott BL, Höök AM, Dickey BF. Inducible epithelial resistance against coronavirus pneumonia in mice. Am J Respir Cell Mol Biol. 2020;63:540–541. doi: 10.1165/rcmb.2020-0247LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.