Acinetobacter baumannii Gastrointestinal Colonization Is Facilitated by Secretory IgA Which Is Reductively Dissociated by Bacterial Thioredoxin A

Multidrug-resistant Acinetobacter baumannii is among the most common causes of infectious complications associated with combat-related trauma in military personnel serving overseas. However, little is currently known about its pathogenesis. While the gastrointestinal (GI) tract has been found to be...

Full description

Bibliographic Details
Main Authors: Patrick M. Ketter, Jieh-Juen Yu, M. Neal Guentzel, Holly C. May, Rishein Gupta, Mark Eppinger, Karl E. Klose, J. Seshu, James P. Chambers, Andrew P. Cap, Bernard P. Arulanandam
Format: Article
Language:English
Published: American Society for Microbiology 2018-07-01
Series:mBio
Subjects:
Online Access:https://doi.org/10.1128/mBio.01298-18
id doaj-ed3c8c00189945aab68add9d521fec6f
record_format Article
spelling doaj-ed3c8c00189945aab68add9d521fec6f2021-07-02T10:52:05ZengAmerican Society for MicrobiologymBio2150-75112018-07-0194e01298-1810.1128/mBio.01298-18Acinetobacter baumannii Gastrointestinal Colonization Is Facilitated by Secretory IgA Which Is Reductively Dissociated by Bacterial Thioredoxin APatrick M. KetterJieh-Juen YuM. Neal GuentzelHolly C. MayRishein GuptaMark EppingerKarl E. KloseJ. SeshuJames P. ChambersAndrew P. CapBernard P. ArulanandamMultidrug-resistant Acinetobacter baumannii is among the most common causes of infectious complications associated with combat-related trauma in military personnel serving overseas. However, little is currently known about its pathogenesis. While the gastrointestinal (GI) tract has been found to be a major reservoir for A. baumannii, as well as to potentially contribute to development of multidrug resistance, no studies have addressed the mechanisms involved in gut colonization. In this study, we address this critical gap in knowledge by first assessing the interaction between secretory IgA (SIgA), the principal humoral immune defense on mucosal surfaces, and the A. baumannii clinical isolate Ci79. Surprisingly, SIgA appeared to enhance A. baumannii GI tract colonization, in a process mediated by bacterial thioredoxin A (TrxA), as evidenced by reduction of bacterial attachment in the presence of TrxA inhibitors. Additionally, a trxA targeted deletion mutant (ΔtrxA) showed reduced bacterial burdens within the GI tract 24 h after oral challenge by in vivo live imaging, along with loss of thiol-reductase activity. Surprisingly, not only was GI tract colonization greatly reduced but the associated 50% lethal dose (LD50) of the ΔtrxA mutant was increased nearly 100-fold in an intraperitoneal sepsis model. These data suggest that TrxA not only mediates A. baumannii GI tract colonization but also may contribute to pathogenesis in A. baumannii sepsis following escape from the GI tract under conditions when the intestinal barrier is compromised, as occurs with cases of severe shock and trauma.Acinetobacter baumannii is an emerging bacterial pathogen recently classified as a serious threat to U.S. and global health by both the Centers for Disease Control and Prevention and the World Health Organization. It also is one of the leading causes of combat-related infections associated with injured military personnel serving overseas. Little is known regarding mechanisms of gastrointestinal tract colonization despite this site being shown to serve as a reservoir for multidrug-resistant (MDR) A. baumannii isolates. Here, we establish that secretory IgA, the major immunoglobulin of mucosal surfaces, promotes A. baumannii GI tract colonization via bacterial thioredoxin A as evidenced through significant reduction in colonization in IgA-deficient animals. Additionally, bacterial colonization and mortality were significantly reduced in animals challenged with a thioredoxin A-deficient A. baumannii mutant. Combined, these data suggest that thioredoxin A is a novel virulence factor, for which antithioredoxin therapies could be developed, for this important multidrug-resistant pathogen.https://doi.org/10.1128/mBio.01298-18Acinetobacterbacterial virulencegastrointestinal infectionSIgAthioredoxin
collection DOAJ
language English
format Article
sources DOAJ
author Patrick M. Ketter
Jieh-Juen Yu
M. Neal Guentzel
Holly C. May
Rishein Gupta
Mark Eppinger
Karl E. Klose
J. Seshu
James P. Chambers
Andrew P. Cap
Bernard P. Arulanandam
spellingShingle Patrick M. Ketter
Jieh-Juen Yu
M. Neal Guentzel
Holly C. May
Rishein Gupta
Mark Eppinger
Karl E. Klose
J. Seshu
James P. Chambers
Andrew P. Cap
Bernard P. Arulanandam
Acinetobacter baumannii Gastrointestinal Colonization Is Facilitated by Secretory IgA Which Is Reductively Dissociated by Bacterial Thioredoxin A
mBio
Acinetobacter
bacterial virulence
gastrointestinal infection
SIgA
thioredoxin
author_facet Patrick M. Ketter
Jieh-Juen Yu
M. Neal Guentzel
Holly C. May
Rishein Gupta
Mark Eppinger
Karl E. Klose
J. Seshu
James P. Chambers
Andrew P. Cap
Bernard P. Arulanandam
author_sort Patrick M. Ketter
title Acinetobacter baumannii Gastrointestinal Colonization Is Facilitated by Secretory IgA Which Is Reductively Dissociated by Bacterial Thioredoxin A
title_short Acinetobacter baumannii Gastrointestinal Colonization Is Facilitated by Secretory IgA Which Is Reductively Dissociated by Bacterial Thioredoxin A
title_full Acinetobacter baumannii Gastrointestinal Colonization Is Facilitated by Secretory IgA Which Is Reductively Dissociated by Bacterial Thioredoxin A
title_fullStr Acinetobacter baumannii Gastrointestinal Colonization Is Facilitated by Secretory IgA Which Is Reductively Dissociated by Bacterial Thioredoxin A
title_full_unstemmed Acinetobacter baumannii Gastrointestinal Colonization Is Facilitated by Secretory IgA Which Is Reductively Dissociated by Bacterial Thioredoxin A
title_sort acinetobacter baumannii gastrointestinal colonization is facilitated by secretory iga which is reductively dissociated by bacterial thioredoxin a
publisher American Society for Microbiology
series mBio
issn 2150-7511
publishDate 2018-07-01
description Multidrug-resistant Acinetobacter baumannii is among the most common causes of infectious complications associated with combat-related trauma in military personnel serving overseas. However, little is currently known about its pathogenesis. While the gastrointestinal (GI) tract has been found to be a major reservoir for A. baumannii, as well as to potentially contribute to development of multidrug resistance, no studies have addressed the mechanisms involved in gut colonization. In this study, we address this critical gap in knowledge by first assessing the interaction between secretory IgA (SIgA), the principal humoral immune defense on mucosal surfaces, and the A. baumannii clinical isolate Ci79. Surprisingly, SIgA appeared to enhance A. baumannii GI tract colonization, in a process mediated by bacterial thioredoxin A (TrxA), as evidenced by reduction of bacterial attachment in the presence of TrxA inhibitors. Additionally, a trxA targeted deletion mutant (ΔtrxA) showed reduced bacterial burdens within the GI tract 24 h after oral challenge by in vivo live imaging, along with loss of thiol-reductase activity. Surprisingly, not only was GI tract colonization greatly reduced but the associated 50% lethal dose (LD50) of the ΔtrxA mutant was increased nearly 100-fold in an intraperitoneal sepsis model. These data suggest that TrxA not only mediates A. baumannii GI tract colonization but also may contribute to pathogenesis in A. baumannii sepsis following escape from the GI tract under conditions when the intestinal barrier is compromised, as occurs with cases of severe shock and trauma.Acinetobacter baumannii is an emerging bacterial pathogen recently classified as a serious threat to U.S. and global health by both the Centers for Disease Control and Prevention and the World Health Organization. It also is one of the leading causes of combat-related infections associated with injured military personnel serving overseas. Little is known regarding mechanisms of gastrointestinal tract colonization despite this site being shown to serve as a reservoir for multidrug-resistant (MDR) A. baumannii isolates. Here, we establish that secretory IgA, the major immunoglobulin of mucosal surfaces, promotes A. baumannii GI tract colonization via bacterial thioredoxin A as evidenced through significant reduction in colonization in IgA-deficient animals. Additionally, bacterial colonization and mortality were significantly reduced in animals challenged with a thioredoxin A-deficient A. baumannii mutant. Combined, these data suggest that thioredoxin A is a novel virulence factor, for which antithioredoxin therapies could be developed, for this important multidrug-resistant pathogen.
topic Acinetobacter
bacterial virulence
gastrointestinal infection
SIgA
thioredoxin
url https://doi.org/10.1128/mBio.01298-18
work_keys_str_mv AT patrickmketter acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
AT jiehjuenyu acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
AT mnealguentzel acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
AT hollycmay acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
AT risheingupta acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
AT markeppinger acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
AT karleklose acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
AT jseshu acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
AT jamespchambers acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
AT andrewpcap acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
AT bernardparulanandam acinetobacterbaumanniigastrointestinalcolonizationisfacilitatedbysecretoryigawhichisreductivelydissociatedbybacterialthioredoxina
_version_ 1721331735101177856