2,117
Views
11
CrossRef citations to date
0
Altmetric
Editorials

Lethal influenza infection: Is a macrophage to blame?

&

Abstract

Alveolar macrophages (AMs) are critical for immunity against influenza A virus (IAV) infection. Depletion, hyporeactivity, and disruption of AM development and differentiation are all associated with lethal IAV infection. AMs drive the innate immune response that limits IAV infection. AMs are crucial for steady-state homeostasis of pulmonary surfactant, and in turn surfactant proteins regulate AMs and participate in host defense against IAV. Known factors that are necessary for AM function and differentiation in vivo include surfactant proteins, the growth factor GM-CSF, the hormone receptor PPARγ, and the transcription factors PU.1 and Bach2. Although PU.1 and PPARγ are downstream effectors of GM-CSF, Bach2 works independently. GM-CSF and Bach2-deficient AMs have phenotypes with immature or alternatively activated states of differentiation, respectively, and both extremes are unsuitable for surfactant homeostasis. The activation state of AMs and the local microenvironment may determine the development of symptomatic versus asymptomatic IAV infection in different individuals.

Alveolar macrophages are critical for host resistance to influenza

The indispensable role of alveolar macrophages (AMs) against influenza A virus (IAV) infection is evidenced in several lines of investigation. Depletion of AMs using clodronate liposomes prior to IAV infection led to uncontrolled viral replication and death in mice, pigs, and ferrets Citation[1–3]. Conditional ablation of AMs by diphtheria toxin administration in CD169(Siglec1)-DTR mice resulted in massive immunopathology and death, supporting an essential role of AMs Citation[4]. Lack of AMs in GM-CSF or GM-CSF receptor-deficient mice impaired control of IAV replication and disrupted respiratory gas exchange causing 100% mortality Citation[5]. Expression of GM-CSF under constitutive or inducible promoters in the lungs of GM-CSF-deficient mice conferred full or partial protection against IAV commensurate with activation Citation[6] and local differentiation Citation[7] of AMs. Exogenous delivery of GM-CSF to the lung protected against lethal IAV infection in wild-type mice Citation[8,9]. Disruption of AM function by conditional deletion of PPARγ, a downstream effector of GM-CSF signaling, impaired clearance of apoptotic cells leading to severe pneumonia with high morbidity and mortality, despite adequate induction of humoral and cell-mediated immunity Citation[5]. The role of Bach2 Citation[10], which regulates differentiation of AMs independent of GM-CSF, in pathogenesis of IAV infection has not been determined. Assessing the role of genetics, it was found that the high susceptibility of DBA/2J mice to IAV infection compared to C57BL/6 mice stemmed from hyporeactivity in innate responsiveness of AMs to IAV infection Citation[11]. Restoration of the intrinsic antiviral Mx1 factor, which is deleted or mutated in most inbred mouse strains, failed to enhance protection in the susceptible DBA/2J mice Citation[12], suggesting dysfunction in upstream mechanisms. A transformative finding in establishing the essential role of AMs is that neonatal transfer of wild-type AM precursors to the lungs of GM-CSF βc receptor subunit-deficient mice protected adult mice from lethal IAV infection Citation[5], indicating a direct and primary role of AMs in protection against IAV infection. The local environment in which IAV infection encounters AMs requires further analysis.

AMs: at the interface of pulmonary homeostasis & host defense

Upon entry into the respiratory tract, IAV encounters a highly regulated immune environment designed to eliminate infection and avoid overt inflammation. Early studies demonstrated that AMs maintain a high threshold of immune activation avoiding inflammatory disease from exposure to innocuous antigens being highly efficient in phagocytosis and clearance of airborne agents and by secretion of mediators that suppress adaptive immunity as reviewed earlier Citation[13]. AMs were subsequently found to be critical for the maintenance of surfactant levels through catabolism of excess surfactant proteins (SPs) and lipids Citation[14]. Excessive accumulation of surfactant during IAV infection in GM-CSF and PPARγ-deficient mice leads to respiratory insufficiency Citation[5]. Epithelial cell-derived GM-CSF drives differentiation of AM precursors that seed the lung at early stages of gestation. In postnatal life, AMs are maintained by local proliferation or differentiated from bone marrow precursors that travel to the lung in response to infection or inflammation Citation[5]. SPs contribute to the phenotype of AMs Citation[15]. The ability of mature AMs to orchestrate immune homeostasis in the resting state involves intercellular communication, cell–cell contact of AMs with respiratory epithelial cells, interaction with pulmonary SP-A and SP-D, and possibly other surfactant components as reviewed previously Citation[16,17]. Significantly, genetic polymorphisms in SP-A were associated with increased susceptibility to acute inflammatory injury in patients infected with the 2009 pandemic H1N1 influenza Citation[18], indicating that SP-A shapes the host response of AMs to IAV infection in humans. A question that has not been clearly addressed is whether GM-CSF modifies these homeostatic mechanisms in the course of an immune response. Increased levels of GM-CSF, in the context of IAV infection, may decrease the threshold of immune activation resulting in increased AM responsiveness and protection against infection or result in chronic inflammation Citation[7]. A recent study demonstrated that low levels of IAV induce minimal expression of inflammatory genes but, depending on viral strain, once the virus titer exceeds a certain level, expression of inflammatory genes is strongly induced Citation[19]. At this stage, IAV could disrupt the lung’s homeostatic circuit by depleting AMs; GM-CSF can prevent depletion of AMs by IAV Citation[20]. It can be envisaged that provision of GM-CSF at a time frame when the virus titer is low reduces threshold of virus recognition, facilitating beneficial activation of AMs to eliminate IAV infection before it proliferates to dangerous levels. Furthermore, by rescuing AMs, GM-CSF may promote activation and differentiation of AM precursors encompassing a range of alternative activation states towards resolution of IAV-induced inflammation. In support of this notion, infection with an IAV strain of intermediate virulence was characterized by the appearance of a heterogeneous population of activated AMs and blood-derived lung monocytes with both populations having a mixed M1/M2 phenotype at early stages of infection Citation[21]. On the other hand, it is also reasonable to speculate that at high IAV titer, persistent activation of GM-CSF as induced by highly pathogenic pH1N1 and H5N1 IAV strains Citation[19] may contribute to injurious inflammation and fibrosis by AMs. In this regard, the scavenger receptor MARCO, a known downstream effector of GM-CSF Citation[7], drives macrophage polarization to the M2 alternative macrophage phenotype extreme leading to pulmonary fibrosis in response to injury Citation[22]. Consistent with a pathogenic role, MARCO-deficient mice are resistant to IAV infection Citation[7,23], suggesting MARCO as a potential target for therapy against influenza-induced injury. Virus-induced GM-CSF could also sensitize AM pattern recognition receptors to tissue and virus-derived ligands enhancing inflammation. For example, activation of TLR-7 may result in oxidative injury through activation of the NADPH oxidase Nox2 in AMs Citation[24]; inhibition of Nox2 has been shown to attenuate IAV-induced lung inflammation Citation[25]. In this context, a decoy peptide that blocked activation of multiple toll-like receptors rescued mice from lethal influenza infection Citation[26]. The interaction of these pathways with the Nox2 subunit rac1 downstream of SP-A receptor isoforms in response to IAV infection remains to be investigated Citation[15]. These studies highlight that IAV evades multiple tasks of AMs in initiating, balancing, and resolution of the inflammatory response.

Abortive infection may direct host outcomes

The fate of infected AMs resulting in pathogenesis or resistance to IAV infection has not been established. Infection of AMs with IAV consists of a robust replication cycle of the viral genome and synthesis of viral proteins without packaging and release of new virus, which is termed abortive or non-productive infection. Given the importance of redox state in influenza pathogenesis Citation[25,27] with the myeloid Nox2 as a key mediator of oxidative injury Citation[25], a question for further study is whether Nox2 facilitates abortive replication of the virus in AMs compared to Nox4 which was shown to mediate replication of IAV in epithelial cells Citation[28]. Effective antiviral immunity by AMs rests on induction of adequate levels of Type I interferon and inflammatory mediators, which suppress IAV infection in respiratory epithelial cells and promote adaptive and cell-mediated immunity Citation[29]. It appears that IAV infection results in depletion of AMs Citation[20], although other studies suggest that the AM population remains stable during IAV infection Citation[30]. On the other hand, premature AM apoptosis as induced by the IAV polymerase gene product PB1-F2 Citation[31], or timely apoptosis following production of Type I interferon Citation[29] may lead to pathogenesis or host resistance to IAV infection, respectively. The Type I interferon response by AMs is subject to inhibition by prostaglandin PGE2 produced by alveolar epithelial cells and macrophages during infection Citation[29]. In this context, GM-CSF may be pivotal for suppression of PGE2 production activating the antiviral response of AMs Citation[32]. One consideration that to our knowledge has not been determined is whether abortive IAV infection drives AMs towards alternative states of differentiation in vivo as has been shown in vitro Citation[33]. In this case, Bach2 deficiency results in AMs with an M2 alternative activation state of differentiation independent of GM-CSF/PPARγ signaling pathways Citation[10], suggesting Bach2 as a termination checkpoint for AM differentiation in the lung. The inability of bach2-deficient M2 AMs to catabolize surfactant suggests that additional factors in the local microenvironment modulate differentiation of AMs through Bach2. A similar phenotype was reported earlier in mice with constitutive overexpression of the M2 macrophage differentiation factor IL-4 Citation[34], at a time when the M1/M2 macrophage differentiation model was not widely studied. The crosstalk between GM-CSF, Bach2, and IAV infection in AMs may thus determine progression of the infection towards disease resolution or disease pathogenesis in symptomatic versus asymptomatic IAV infection, respectively.

Concluding remarks

IAV causes highly contagious respiratory infections that spread from the upper to the lower airway and can lead to fatal viral pneumonia. New strains of IAV that easily spread between individuals arise frequently. AMs are essential for host resistance to IAV infection. We propose that AMs also serve as an essential portal for IAV infection, pathogenesis, and transmission. It is evident that IAV induces inflammatory/oxidative pathways to establish infection; such capacity would increase the risk for development of detrimental IAV-induced pneumonia by AMs. Controlled IAV infections of human volunteer cohorts revealed molecular signatures that can differentiate symptomatic from asymptomatic individuals during seasonal influenza Citation[35]; asymptomatic individuals are not restrained by hospitalization and can freely shed infectious virus in the community. That GM-CSF, a key homeostatic factor for AMs, enhances host resistance to influenza supports the notion that IAV exploits the local homeostatic circuitry to establish infection and transmission. Through abortive infection of AMs, IAV is in a position to ‘polarize’ the immune response towards tolerance or pathogenesis depending on inter-individual genetic differences distributed randomly in the population. We propose that understanding the mechanisms of IAV entry and persistence in AMs is of paramount importance for discovering how to prevent spread of deadly IAV infections. The set point that enables disposal of the offending virus and restoration of the lung microenvironment by AMs, as exemplified by treatment with GM-CSF or innate immune activation antagonists, is amenable to therapeutic intervention.

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • Wijburg OL, DiNatale S, Vadolas J, et al. Alveolar macrophages regulate the induction of primary cytotoxic T-lymphocyte responses during influenza virus infection. J Virol 1997;71(12):9450-7
  • Kim HM, Lee YW, Lee KJ, et al. Alveolar macrophages are indispensable for controlling influenza viruses in lungs of pigs. J Virol 2008;82(9):4265-74
  • Kim HM, Kang YM, Ku KB, et al. The severe pathogenicity of alveolar macrophage-depleted ferrets infected with 2009 pandemic H1N1 influenza virus. Virology 2013;444(1-2):394-403
  • Purnama C, Ng SL, Tetlak P, et al. Transient ablation of alveolar macrophages leads to massive pathology of influenza infection without affecting cellular adaptive immunity. Eur J Immunol 2014;44(7):2003-12
  • Schneider C, Nobs SP, Heer AK, et al. Alveolar macrophages are essential for protection from respiratory failure and associated morbidity following influenza virus infection. PLoS Pathog 2014;10(4):e1004053
  • Huang FF, Barnes PF, Feng Y, et al. GM-CSF in the lung protects against lethal influenza infection. Am J Respir Crit Care Med 2011;184(2):259-68
  • Sever-Chroneos Z, Murthy A, Davis J, et al. GM-CSF modulates pulmonary resistance to influenza A infection. Antiviral Res 2011;92(2):319-28
  • Subramaniam R, Hillberry Z, Chen H, et al. Delivery of GM-CSF to Protect against Influenza Pneumonia. PLoS One 2015;10(4):e0124593
  • Huang H, Li H, Zhou P, Ju D. Protective effects of recombinant human granulocyte macrophage colony stimulating factor on H1N1 influenza virus-induced pneumonia in mice. Cytokine 2010;51(2):151-7
  • Nakamura A, Ebina-Shibuya R, Itoh-Nakadai A, et al. Transcription repressor Bach2 is required for pulmonary surfactant homeostasis and alveolar macrophage function. J Exp Med 2013;210(11):2191-204
  • Casanova T, Van de Paar E, Desmecht D, Garigliany MM. Hyporeactivity of Alveolar Macrophages and Higher Respiratory Cell Permissivity Characterize DBA/2J Mice Infected by Influenza A Virus. J Interferon Cytokine Res 2015. [Epub ahead of print]
  • Shin DL, Hatesuer B, Bergmann S, et al. Protection from severe influenza infections in mice carrying the Mx1 influenza resistance gene strongly depends on genetic background. J Virol 2015;89(19):9998-10009
  • Holt PG, Strickland DH, Wikstrom ME, Jahnsen FL. Regulation of immunological homeostasis in the respiratory tract. Nat Rev Immunol 2008;8(2):142-52
  • Yoshida M, Ikegami M, Reed JA, et al. GM-CSF regulates protein and lipid catabolism by alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 2001;280(3):L379-86
  • Yang L, Carrillo M, Wu YM, et al. SP-R210 (Myo18A) Isoforms as Intrinsic Modulators of Macrophage Priming and Activation. PLoS One 2015;10(5):e0126576
  • Hussell T, Bell TJ. Alveolar macrophages: plasticity in a tissue-specific context. Nat Rev Immunol 2014;14(2):81-93
  • Chroneos ZC, Sever-Chroneos Z, Shepherd VL. Pulmonary surfactant: an immunological perspective. Cell Physiol Biochem 2010;25(1):13-26
  • Herrera-Ramos E, Lopez-Rodriguez M, Ruiz-Hernandez JJ, et al. Surfactant protein A genetic variants associate with severe respiratory insufficiency in pandemic influenza A virus infection. Crit Care 2014;18(3):R127
  • Shoemaker JE, Fukuyama S, Eisfeld AJ, et al. An Ultrasensitive Mechanism Regulates Influenza Virus-Induced Inflammation. PLoS Pathog 2015;11(6):e1004856
  • Ghoneim HE, Thomas PG, McCullers JA. Depletion of alveolar macrophages during influenza infection facilitates bacterial superinfections. J Immunol 2013;191(3):1250-9
  • Duan M, Li WC, Vlahos R, et al. Distinct macrophage subpopulations characterize acute infection and chronic inflammatory lung disease. J Immunol 2012;189(2):946-55
  • Murthy S, Larson-Casey JL, Ryan AJ, et al. Alternative activation of macrophages and pulmonary fibrosis are modulated by scavenger receptor, macrophage receptor with collagenous structure. FASEB J 2015;29(8):3527-36
  • Ghosh S, Gregory D, Smith A, Kobzik L. MARCO regulates early inflammatory responses against influenza: a useful macrophage function with adverse outcome. Am J Respir Cell Mol Biol 2011;45(5):1036-44
  • To EE, Broughton BR, Hendricks KS, et al. Influenza A virus and TLR7 activation potentiate NOX2 oxidase-dependent ROS production in macrophages. Free Radic Res 2014;48(8):940-7
  • Vlahos R, Stambas J, Bozinovski S, et al. Inhibition of Nox2 oxidase activity ameliorates influenza A virus-induced lung inflammation. PLoS Pathog 2011;7(2):e1001271
  • Piao W, Shirey KA, Ru LW, et al. A Decoy Peptide that Disrupts TIRAP Recruitment to TLRs Is Protective in a Murine Model of Influenza. Cell Reports 2015;11(12):1941-52
  • Yatmaz S, Seow HJ, Gualano RC, et al. Glutathione peroxidase-1 reduces influenza A virus-induced lung inflammation. Am J Respir Cell Mol Biol 2013;48(1):17-26
  • Amatore D, Sgarbanti R, Aquilano K, et al. Influenza virus replication in lung epithelial cells depends on redox-sensitive pathways activated by NOX4-derived ROS. Cell Microbiol 2015;17(1):131-45
  • Coulombe F, Jaworska J, Verway M, et al. Targeted prostaglandin E2 inhibition enhances antiviral immunity through induction of type I interferon and apoptosis in macrophages. Immunity 2014;40(4):554-68
  • Janssen WJ, Barthel L, Muldrow A, et al. Fas determines differential fates of resident and recruited macrophages during resolution of acute lung injury. Am J Respir Crit Care Med 2011;184(5):547-60
  • Jaworska J, Coulombe F, Downey J, et al. NLRX1 prevents mitochondrial induced apoptosis and enhances macrophage antiviral immunity by interacting with influenza virus PB1-F2 protein. Proc Natl Acad Sci USA 2014;111(20):E2110-19
  • Sorgi CA, Rose S, Court N, et al. GM-CSF priming drives bone marrow-derived macrophages to a pro-inflammatory pattern and downmodulates PGE2 in response to TLR2 ligands. PLoS One 2012;7(7):e40523
  • Zhao X, Dai J, Xiao X, et al. PI3K/Akt signaling pathway modulates influenza virus induced mouse alveolar macrophage polarization to M1/M2b. PLoS One 2014;9(8):e104506
  • Ikegami M, Whitsett JA, Chroneos ZC, et al. IL-4 increases surfactant and regulates metabolism in vivo. Am J Physiol Lung Cell Mol Physiol 2000;278(1):L75-80
  • Huang Y, Zaas AK, Rao A, et al. Temporal dynamics of host molecular responses differentiate symptomatic and asymptomatic influenza a infection. PLoS Genet 2011;7(8):e1002234

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.