<<

Antibiotic therapy–induced collateral damage: IgA takes center stage in pulmonary host defense

Juergen Lohmeyer, … , Rory E. Morty, Susanne Herold

J Clin Invest. 2018;128(8):3234-3236. https://doi.org/10.1172/JCI122032.

Commentary

The use of broad-spectrum in empirical therapy is a lifesaving strategy for patients in intensive care. At the same time, antibiotics dramatically increase the risk for nosocomial , such as hospital‑acquired pneumonia caused by Pseudomonas aeruginosa, and other -resistant . In this issue of theJ CI, Robak and colleagues identified a mechanism by which depletion of resident gut and lung microbiota by antibiotic treatment results in secondary IgA deficiency and impaired anti–P. aeruginosa host defense. Impaired defenses could be improved by substitution of polyclonal IgA via the intranasal route in a mouse model of pneumonia. Importantly, antibiotic treatment caused lung IgA deficiency that involved reduced TLR-dependent production of a proliferation-inducing ligand (APRIL) and B –activating factor (BAFF) in intensive care unit patients. These patients might therefore benefit from future strategies to increase pulmonary IgA levels.

Find the latest version: https://jci.me/122032/pdf COMMENTARY The Journal of Clinical Investigation Antibiotic therapy–induced collateral damage: IgA takes center stage in pulmonary host defense

Juergen Lohmeyer,1 Rory E. Morty,1,2 and Susanne Herold1,3 1Department of Internal II, University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany. 2Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Bad Nauheim, Germany. 3Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.

biota changes induced by ABx treatment inhibit immunoglobulin A (IgA) produc- The use of broad-spectrum antibiotics in empirical antimicrobial therapy tion in the lung, thereby increasing suscep- is a lifesaving strategy for patients in intensive care. At the same time, tibility of ABx-treated patients to antibiotics dramatically increase the risk for nosocomial infections, such with P. aeruginosa (Figure 1). as hospital‑acquired pneumonia caused by Pseudomonas aeruginosa, and other antibiotic-resistant bacteria. In this issue of the JCI, Robak and Microbiota-host immunity colleagues identified a mechanism by which depletion of resident gut and cross-talk lung microbiota by antibiotic treatment results in secondary IgA deficiency How do the findings of Robak et al. help and impaired anti–P. aeruginosa host defense. Impaired defenses could expand our understanding of the interplay be improved by substitution of polyclonal IgA via the intranasal route in between the microbiota and host defense a mouse model of pneumonia. Importantly, antibiotic treatment caused in lung infection, and thereby promote lung IgA deficiency that involved reduced TLR-dependent production of a development of new prophylactic or thera- proliferation-inducing ligand (APRIL) and B cell–activating factor (BAFF) in peutic strategies? It is well established that intensive care unit patients. These patients might therefore benefit from antimicrobial therapies inevitably cause future strategies to increase pulmonary IgA levels. dramatic and long-lasting collateral dam- age to the diverse populations of com- mensal bacterial species that are part of a patient’s intestinal microbiota. However, Antibiotic use and risk of risk factor for the normal to recent investigations have further revealed nosocomial infection turn into a so-called pathobiome, which is that composition of the microbiota also Broad-spectrum antibiotics (ABx) form diagnosed as hospital-acquired pneumo- crucially impacts systemic and pulmonary the backbone of empiric antimicrobial nia (HAP) and often associated with out- innate immune responses during bacterial treatment regimens, which are lifesav- growth of P. aeruginosa or other antibiotic- and viral infections (13–15). Considerably ing for those with severe infections such resistant bacteria (6, 7). Recent estimates less is known about how the microbiota as pneumonia, peritonitis, or sepsis, and indicate that in Europe, 2.6 million cases ecosystem and its depletion by ABx impact which constitute an integral part of treat- of hospital-acquired infections occur each adaptive immune responses. The gut ment guidelines (1). However, apart from year, and that HAP accounts for the high- microbiota is critical for regulating intesti- driving the selection of multiresistant est burden and number of deaths nal IgA production, as IgA-secreting cells , ABx severely disrupt the host (8). HAP dramatically increases both the and IgA production are almost absent in microbiome, which has been increasingly length of the hospital stay and health care the gut of germ-free mice (16). Moreover, recognized as a critical factor in shaping costs, and is associated with a mortality of microbial signals are known to activate host immunity (2). Antibiotic stewardship up to 13% (9). TLRs on intestinal epithelial cells and DCs and improved point-of-care microbial The increased susceptibility of critical- to induce production of the crucial B cell diagnostics increasingly facilitate the opti- ly ill patients to infection with multidrug survival signals APRIL and BAFF, which mized use of specific antibiotics (3), but in resistant (MDR) gram-negative bacteria in turn, promote IgA production by plas- critically ill patients the empiric use of ABx was originally attributed to generation of ma cells (17–19). In addition, microbiota- will remain indispensable. Point-preva- a permissive niche with selection of MDR derived short-chain fatty acids have lence studies have documented that to pathogens that were not covered by the recently been demonstrated to positively date 64%–71% of all patients in intensive ABx regimen (10, 11). In this issue, Robak regulate IgA production (20). care units (ICUs) receive antibiotics (4, 5). and colleagues (12) identify an additional Previous antimicrobial therapy is a major mechanism by which lung and gut micro- IgA-mediated pulmonary host defense Related Article: p. 3535 What makes IgA so special for lung host defense? Immunoglobulin A has a criti- Conflict of interest: The authors have declared that no conflict of interest exists. cal role in immune defense, particularly Reference information: J Clin Invest. 2018;128(8):3234–3236. https://doi.org/10.1172/JCI122032. at mucosal surfaces, and IgA is specially

3234 jci.org Volume 128 Number 8 August 2018 The Journal of Clinical Investigation COMMENTARY

Figure 1. Broad-spectrum antibiotic treatment exerts severe collateral damage by inhibiting microbiota-induced secretory IgA synthesis and IgA- dependent lung host defense toward P. aeruginosa. The lung and gut commensal microbiota induce IgA production at mucosal surfaces involving TLR-, APRIL-, and BAFF-dependent signaling. Lung secretory IgA (sIgA) binds P. aeruginosa and reduces host susceptibility to P. aeruginosa pneumonia. ABx treatment destroys luminal microbiota and severely reduces sIgA production by lung IgA-secreting plasma cells, thereby impairing anti-pseudomonas host defense, which can be reestablished by transnasal administration of sIgA.

equipped to undertake this task through had not been investigated if and how dis- However, it is also conceivable that the the unique structural attributes of the turbances to this mechanism contribute to induction of IgA generation occurs in the IgA heavy chain and by virtue of its abil- the enhanced susceptibility to lung infec- intestinal tract and that IgA+ plasma cells ity to polymerize (16). Notably, more IgA tions following antibiotic treatment. The subsequently migrate to the lung, given antibodies are synthesized in mammals findings by Robak et al. explain how deple- that migration of plasma cells between per day than antibodies of all other iso- tion of the resident microbiota affects lung lung and intestine has been previously types combined. IgA plays a pivotal role host defense against P. aeruginosa and described (22). Second, the cellular com- in maintaining a homeostatic relation- represent a fundamental step in filling this partment responsible for the TLR-depen- ship between the host and the resident knowledge gap. To our knowledge, this is dent APRIL production and the specific microbiota of the intestine (21). Produc- the first study to demonstrate that deple- TLR member involved remain unknown. tion of high-affinity and antigen‑specific tion of resident microbiota by ABx inhib- Third, the mechanism by which IgA exerts IgA in Peyer’s patches and mesenteric its TLR-dependent production of APRIL, antimicrobial properties directed against lymph nodes has been documented to be resulting in secondary IgA deficiency in P. aeruginosa (and most likely other bac- T cell dependent (16), whereas low-affin- the lungs of both mice and ICU patients terial species) in the lung is unknown, ity polyreactive IgA responses are mainly and increases susceptibility to subsequent but likely is similar to the gut, where IgA induced in isolated lymphoid follicles P. aeruginosa infection (Figure 1). There achieves neutralization of invasive patho- and in subepithelial B cells in a T cell– are several essential unresolved questions gens by various mechanisms, such as independent manner (19). Both low- and that arise from this work. First, what is the immobilization, opsonization, or killing high-affinity IgA regulate the composi- cellular source of TLR-dependent APRIL, (23, 24). Knowledge about such molecular tion of the intestinal microbiota by coat- BAFF, and IgA production? The relevant mechanisms will be crucial to select the ing many of the bacterial components in cell might in fact be in the lung, as suggest- most appropriate preparations for passive the intestinal lumen, in order to maintain ed by a recent study that demonstrated IgA supplementation. Finally, the specific intestinal homeostasis (21). that lung CD103+ and CD24+CD11b+ DCs features of respiratory and/or intestinal In contrast to the gut, it was not known induce IgA class-switch recombination by microbiota (for example, distinct bacte- whether microbiota-dependent IgA pro- activating B cells through T cell–depen- rial taxa) required for the induction of duction also maintained lung microbi- dent or –independent pathways (22). This protective immunity in the lung, and the ome homeostasis and whether IgA was process involved microbial stimuli act- potential of to rescue this IgA- required for antibacterial defense in the ing through MyD88/TRIF-mediated TLR dependent antibacterial defense, remain lungs of mice and humans. Moreover, it signaling and TGF-β receptor signaling. to be defined.

jci.org Volume 128 Number 8 August 2018 3235 COMMENTARY The Journal of Clinical Investigation

Conclusions and future the German Center for Lung Research colonization resistance against intestinal patho- directions (DZL), and by the Max Planck Society. gens. Nat Rev Immunol. 2013;13(11):790–801. 11. Pamer EG. Resurrecting the intestinal micro- Current attempts to target the affected biota to combat antibiotic-resistant pathogens. microbiome for infection prevention Address correspondence to: Juergen Loh- Science. 2016;352(6285):535–538. or even treatment primarily intervene meyer, Department of Medicine II, Justus- 12. Robak OH, et al. Antibiotic treatment–induced at points upstream to reconstitute the Liebig-University, Klinikstr. 33, 35392 Gies- secondary IgA deficiency enhances susceptibil- microbial flora or/and microbial‑derived sen, Germany. Phone 49.641.985.57061; ity to Pseudomonas aeruginosa pneumonia. J Clin Invest. 2018;128(8):3535–3545. signals using live microbiota (probiotics) Email: [email protected]. 13. Clarke TB, Davis KM, Lysenko ES, Zhou AY, or even fecal transplantation; however, uni-giessen.de. Yu Y, Weiser JN. Recognition of peptidoglycan these strategies face considerable regula- from the microbiota by Nod1 enhances systemic tory hurdles (11). The pathway outlined 1. Ferrer R, et al. Empiric antibiotic treatment innate immunity. Nat Med. 2010;16(2):228–231. by Robak et al. (12) now offers an attrac- reduces mortality in severe sepsis and septic 14. Ichinohe T, et al. Microbiota regulates immune shock from the first hour: results from a guide- defense against respiratory tract tive downstream intervention to compen- line-based performance improvement program. A infection. Proc Natl Acad Sci U S A. sate for disrupted microbiome-related Crit Care Med. 2014;42(8):1749–1755. 2011;108(13):5354–5359. defects in humoral defense. To optimize 2. Honda K, Littman DR. The microbiota in adap- 15. Abt MC, et al. Commensal bacteria calibrate the a passive IgA supplementation approach tive immune homeostasis and disease. Nature. activation threshold of innate antiviral immu- toward clinical , the potential 2016;535(7610):75–84. nity. Immunity. 2012;37(1):158–170. 3. Campion M, Scully G. Antibiotic use in the 16. Pabst O. New concepts in the generation of different immunoglobulin prepara- intensive care unit: optimization and de-esca- and functions of IgA. Nat Rev Immunol. tions and routes of administration as lation [published online ahead of print Janu- 2012;12(12):821–832. preemptive or therapeutic interventions ary 1, 2018]. J Intensive Care Med. https://doi. 17. Tezuka H, et al. Regulation of IgA production by for HAP need to be evaluated in both org/10.1177/0885066618762747. naturally occurring TNF/iNOS-producing den- experimental models and clinical trials. 4. Zarb P, et al. The European Centre for Disease dritic cells. Nature. 2007;448(7156):929–933. Prevention and Control (ECDC) pilot point 18. Litinskiy MB, et al. DCs induce CD40- Although the location of TLR-dependent survey of healthcare-associated independent immunoglobulin class switch- microbiota sensing and APRIL, BAFF, infections and antimicrobial use. Euro Surveill. ing through BLyS and APRIL. Nat Immunol. and IgA induction remains to be defined, 2012;17(46):20316. 2002;3(9):822–829. the pathway identified by Robak et al. 5. Vincent JL, et al. International study of the prev- 19. He B, et al. Intestinal bacteria trigger T cell- also holds promise for the development alence and outcomes of infection in intensive independent immunoglobulin A(2) class switch- care units. JAMA. 2009;302(21):2323–2329. ing by inducing epithelial-cell secretion of the of novel strategies that rely on preserv- 6. Shindo Y, et al. Risk factors for drug-resistant cytokine APRIL. Immunity. 2007;26(6):812–826. ing or restoring IgA function under ABx pathogens in community-acquired and health- 20. Kim M, Qie Y, Park J, Kim CH. Gut microbial treatment. IgA restoration could be care-associated pneumonia. Am J Respir Crit metabolites fuel host antibody responses. Cell achieved by engagement of potentially Care Med. 2013;188(8):985–995. Host Microbe. 2016;20(2):202–214. druggable candidate pathways using suit- 7. Venier AG, et al. Identifying new risk factors 21. Macpherson AJ, Yilmaz B, Limenitakis JP, for Pseudomonas aeruginosa pneumonia in Ganal-Vonarburg SC. IgA function in relation able agonists of the still-undefined TLR intensive care units: experience of the French to the intestinal microbiota. Annu Rev Immunol. signaling pathway or the APRIL/BAFF national surveillance, REA-RAISIN. J Hosp 2018;36:359–381. signaling axis. Such treatments are not Infect. 2011;79(1):44–48. 22. Ruane D, et al. Microbiota regulate the ability only envisioned to improve outcomes of 8. Cassini A, et al. Burden of six healthcare- of lung dendritic cells to induce IgA class- P. aeruginosa–induced HAP, but might associated infections on European population switch recombination and generate protective health: estimating -based disability- gastrointestinal immune responses. J Exp Med. also be applied to reduce chronic P. aeru- adjusted life years through a population 2016;213(1):53–73. ginosa colonization in those at risk, such prevalence-based modelling study. PLoS Med. 23. Simmons CP, et al. Central role for B lymphocytes as patients with cystic fibrosis. 2016;13(10):e1002150. and CD4+ T cells in immunity to infection by 9. Melsen WG, et al. Attributable mortality of the attaching and effacing Citrobacter Acknowledgments ventilator-associated pneumonia: a meta- rodentium. Infect Immun. 2003;71(9):5077–5086. analysis of individual patient data from ran- 24. Rollenske T, et al. Cross-specificity of protec- This work was supported by the Deutsche domised prevention studies. Lancet Infect Dis. tive human antibodies against Klebsiella Forschungsgemeinschaft (DFG) through 2013;13(8):665–671. pneumoniae LPS O-antigen. Nat Immunol. grants EXC147 and CRU309 (P2, P8, P6), 10. Buffie CG, Pamer EG. Microbiota-mediated 2018;19(6):617–624.

3236 jci.org Volume 128 Number 8 August 2018