Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

REVIEW

C URRENT
OPINION Gut–lung crosstalk during critical illness
Sridesh Nath a, Georgios D. Kitsios a,b,c, and Lieuwe D.J. Bos d,e,
Downloaded from http://journals.lww.com/co-criticalcare by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XM

Purpose of review
Study of organ crosstalk in critical illness has uncovered complex biological communication between
i0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 05/03/2023

different organ systems, but the role of microbiota in organ crosstalk has received limited attention. We
highlight the emerging understanding of the gut--lung axis, and how the largest biomass of the human
body in the gut may affect lung physiology in critical illness.
Recent findings
Disruption of healthy gut microbial communities and replacement by disease-promoting pathogens
(pathobiome) generates a maladaptive transmitter of messages from the gut to the lungs, connected via the
portal venous and the mesenteric lymphatic systems. Gut barrier impairment allows for microbial
translocation (living organisms or cellular fragments) to the lungs. Host-microbiota interactions in the gut
mucosa can also impact lung physiology through microbial metabolite secretion or host-derived messengers
(hormones, cytokines or immune cells). Clinical examples like the prevention of ventilator-associated
pneumonia by selective decontamination of the digestive tract show that the gut--lung axis can be
manipulated therapeutically.
Summary
A growing body of evidence supports the pathophysiological relevance of the gut--lung axis, yet we are
only at the brink of understanding the therapeutic and prognostic relevance of the gut microbiome,
metabolites and host-microbe interactions in critical illness.
Keywords
critical illness, gut microbiome, gut--lung axis, pathobiome

INTRODUCTION nonhuman integral component of the human host,


Organ crosstalk, the complex biological communi- or the so-called forgotten organ of critical illness: the
cation between anatomically distant organs medi- human microbiome.
ated by various signaling factors, is essential for Matching human cells in nearly 1:1 ratio, an
dynamic maintenance of homeostasis. Maladjusted estimated 39 trillion bacteria colonize the external
inter-organ communication in critical illness can and internal surfaces of the human body, including
amplify the systemic negative impact of single organ the skin, gastrointestinal, respiratory and urogenital
dysfunction, beyond what is predicted from the tract, yet the vast majority of bacterial biomass
directly measured physiologic deficits of the failing resides in the gut and especially in the colon [2].
organ. For example, acute kidney injury can lead to A growing body of research in the role of gut micro-
worse pulmonary dysfunction than what is expected biota in health and disease has expanded the
merely due to fluid retention, likely due to humoral/
cellular factors released or retained from the injured a
Department of Medicine, Division of Pulmonary, Allergy and Critical
kidneys and acting distally to the lungs [1]. Simi- Care Medicine, bAcute Lung Injury Center of Excellence, cCenter for
larly, our well intended interventions to restore or Medicine and the Microbiome, University of Pittsburgh, Pittsburgh,
Pennsylvania, dIntensive Care and eLaboratory of Experimental Intensive
replace homeostatic function in one organ system Care and Anesthesiology (LEICA), Amsterdam University Medical
may have unintended consequences in other distal Centers, location AMC, University of Amsterdam, Amsterdam, The
organs, because organ crosstalk occurs in complex Netherlands
ways that are not easy to observe on the clinical Correspondence to Lieuwe D.J. Bos, MD, PhD, Amsterdam UMC
surface. Although extensive research has focused on location AMC, Intensive Care, Meibergdreef 9, 1105AZ Amsterdam,
understanding organ crosstalk in critical illness, The Netherlands. E-mail: l.d.bos@amsterdamumc.nl

involving physiologic, neurohormonal, metabolic These authors contributed equally to this work.
and immune mechanisms, more recent investiga- Curr Opin Crit Care 2023, 29:130–137
tions have started to unravel the role of the DOI:10.1097/MCC.0000000000001015

www.co-criticalcare.com Volume 29  Number 2  April 2023

Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.


Gut--lung crosstalk during critical illness Nath et al.

[3]. On the other end of the ecological spectrum, we


KEY POINTS have the microbe-friendly gut, generously sharing
 Gut microbial communities in critical illness show the dietary nutrients ingested by the human host.
decreased species diversity, loss of obligate anaerobes Progressively increasing in biomass and composi-
and emergence of pathogenic species abundance, tional diversity along the gastrointestinal tract, gut
collectively shaping the profile of a disease- microbiota in the colon constitute an abundant,
promoting pathobiome. diverse and metabolically active community of
 The gut--lung axis is comprised of both microbial and mostly obligate anaerobic bacteria [4].
host-derived messengers, traveling from the gut to the Despite its anatomical remoteness, a gut–lung
Downloaded from http://journals.lww.com/co-criticalcare by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XM

lungs via the routes of the mesenteric lymphatic and the axis is strongly supported by experimental and clin-
portal venous system. ical evidence in health and disease (Fig. 1). The two
i0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 05/03/2023

compartments are connected via the venous (sys-


 Impaired gut barrier integrity and increased
permeability allows for living gut microbes or their temic and portal) and lymphatic circulations. Gut
cellular fragments to reach the lungs and populate the microbiota can use these connecting routes to send
lung microbiome or induce remote host innate ‘‘messages’’ to lung microbiota and human lung
immune responses. cells either in direct ways, via microbial transloca-
tion and engraftment in the lungs, or mostly indi-
 Clinical studies show enrichment of the lung
microbiome with gut-origin bacteria in patients with rect ways, involving a variety of microbial or host
acute respiratory distress syndrome. ‘‘messengers’’ (Fig. 1). Under this expansive gut–
lung crosstalk model, lung function can be impacted
 The clinical success of selective decontamination by remote host-microbiota interactions in the gut,
strategies of the digestive tract in reducing ventilator-
even if gut microbes never breach the gut mucosa.
associated pneumonia and improving clinical outcomes
offers proof-of-concept evidence for the premise of Although there is emerging data for inverse, lung-to-
therapeutic manipulation of the gut--lung axis in gut influences on microbiota, we focus our evidence
critical illness. synthesis on studies investigating the unidirectional
impact of the high biomass gut microbiome on lung
physiology in the context of critical illness, starting
by what is known on gut–lung crosstalk from
concept of ‘‘organ crosstalk’’ to also include the chronic lung disease.
direct and indirect effects of gut microbiota on distal
organ physiology (e.g. gut–brain, gut–lung or gut–
liver axis). In this review, we focus on the role of GUT–LUNG AXIS IN CHRONIC LUNG
the gut microbiome on pulmonary dysfunction in DISEASES
critical illness and summarize recent evidence on Human and animal model studies in chronic respi-
the proposed gut–lung axis. ratory diseases, such as allergies, asthma and chronic
obstructive pulmonary disease (COPD), have largely
shaped our understanding on the gut–lung axis.
GUT AND LUNG MICROBIOTA: DISTANT Epidemiologic observations have linked early life
BUT RELATED factors shaping the gut microbiome, such as mode
Despite their common embryologic origin from the of delivery, lactation or antibiotic exposures, with
foregut and their common pharyngeal passage, the future development of atopic disease and asthma
anatomic development of the respiratory and gas- [5]. Patients with COPD have distinct gut micro-
trointestinal tracts results in two environments biome profiles from healthy controls, with plausible
vastly different in their hospitality for microbiota. causal mechanisms involving alternations of micro-
&
The lungs are the unfriendly environment. The bial metabolism [6 ]. Clinical observations also
healthy lower airways and airspaces are poor in implicate translocation in severe COPD, as patients
nutrient supply for bacteria and are continuously with acute exacerbations have increased small intes-
patrolled by lung macrophages. This inhospitable tinal permeability, while plasma levels of 1,3-b-D-
environment results in a low biomass yet diverse glucan (BDG), a fungal cell-wall constituent, poten-
community of bacteria originating from the oral tially signifying translocating fungi from the gut),
cavity. It is not entirely clear whether this low correlate with lung matrix degradation markers and
&
microbial population density is resident in the worse lung function [7 ,8].
healthy lungs or continuously re-populated from The most thoroughly studied mechanism of the
the upper respiratory tract, yet in each unit of time, gut–lung axis involves the microbial metabolites
microbes are present and play important roles in short-chain fatty acids (SCFAs), such as propionate
tuning resident innate and adaptive immune cells or butyrate [9]. Production of SCFA in the gut

1070-5295 Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved. www.co-criticalcare.com 131

Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.


Gastrointestinal system
Downloaded from http://journals.lww.com/co-criticalcare by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XM
i0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 05/03/2023

FIGURE 1. A schematic summary of the connecting routes and the possible messengers constituting the gut--lung axis in critical
illness. The two compartments are anatomically connected via the venous and lymphatic circulations. Gut barrier disruption due to
ischemia or inflammatory tissue injury in critical illness can allow for translocation of gut microbiota (mostly bacteria or fungi), which
can travel via the venous or lymphatic route and engraft in the lungs, generating a gut-microbiota enriched lung microbiome. Apart
from this prototypical scenario of a microbial gut--lung axis, gut microbiota can affect lung physiology even when the connecting
routes are sterile from living organisms. Cellular fragments from killed microbes (e.g. cell-wall constituents, such as
Lipopolysaccharide or b-D-glucan for bacteria and fungi, respectively or nucleic acids, such as cell-free DNA) or metabolites released
by living bacteria (such as short-chain fatty acids) may reach the lungs and stimulate innate or adaptive immune responses. In an
expanded conceptual model of the gut--lung axis, the two organs can interact even when there is no breach of the intestinal mucosa
by living organisms or microbial components. Host--microbiota interactions in the intestinal mucosa can result in release of damage-
associated molecular patterns (DAMPs, e.g. cell-free human DNA from injured intestinal cells), extracellular vesicles, cytokines,
hormones or even migrating immune cells, which can all reach the lungs and act as host messengers of the host-microbiome
interactions confined within the gut. DAMPs, damage-associated molecular patterns.

132 www.co-criticalcare.com Volume 29  Number 2  April 2023

Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.


Gut--lung crosstalk during critical illness Nath et al.

depends entirely on bacteria. Dietary fiber that is bacteremia, Clostridioides difficile colitis and nosoco-
&&
unfermentable by the host is metabolized by fer- mial pneumonia [16 ,17]. A pathobiome is then the
menting bacteria in the colon to produce SCFAs. necessary (but perhaps not sufficient) condition for
Diets high in fiber and low in animal protein and a maladaptive gut–lung axis in critical illness.
fat further enrich fiber-fermenting bacteria and con-
sequently SCFA production. SCFAs serve as a fuel for
colonic epithelia cells, thereby improving barrier EMPIRICAL EVIDENCE INDICATIVE OF
function and mucosal integrity, but also act as impor- GUT–LUNG AXIS IN CRITICAL ILLNESS
tant immunomodulatory molecules. SCFAs modu- Culture-based surveys in mechanically ventilated
Downloaded from http://journals.lww.com/co-criticalcare by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XM

late allergic and asthma responses by promoting patients have shown that pharyngeal or intestinal
dendritic cell maturation in the bone marrow in ways colonization with gram-negative bacteria predicts
i0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 05/03/2023

that such dendritic cells are ineffective at driving TH2 subsequent ventilator-associated pneumonias
&
cell responses when they migrate to the airways [10]. (VAP) [18 ,19]. Such epidemiologic observations
Pilot clinical studies on asthma patients have shown provided the rationale for preventive decontamina-
the benefit of inulin supplementation (a soluble fiber tion strategies, such as oral care with chlorhexidine
which is fermented by intestinal microbes leading to rinses, or the more aggressive options of selective
SCFA production) in reducing airway inflammation decontamination of the digestive tract (SDD) and
and exhaled nitric oxide levels [11]. selective oropharyngeal decontamination (SOD)
Such gut-derived coordination of adaptive with nonabsorbable antibiotics against Gram-nega-
&& && &&
immune responses requires repeated host–micro- tive bacteria, yeasts, and S. aureus [20 ,21 ,22 ].
biota interactions to impact systemic responses SDD avoids the use of antibiotics with anaerobic
and the immunophenotype of lung resident cells, coverage, with the aim of eliminating gut-derived
as the result of a complex and time-consuming pathogens while preserving commensal anaerobes.
process. Despite their relevance in chronic respira- Head-to-head comparison has shown SDD to be
tory diseases, such mechanisms may not operate superior to SOD in reducing VAP incidence and
similarly in critical illness, which can happen improving clinical outcomes including mortality.
acutely and evolve rapidly, with innate immune Empiric administration of antianaerobic antibiotics
responses mostly at play. We will next review the in critically ill patients was recently shown to be
available clinical and mechanistic evidence of gut– associated with decreased overall survival, increased
lung axis and its impact on innate immunity and risk of VAP, and enrichment by pathogenic Enter-
lung function in critical illness syndromes. obacteriaceae in gut microbial communities and
&&
respiratory cultures diagnostic of VAP [23 ]. This
randomized and observational evidence defies con-
GUT MICROBIOME IN CRITICAL ILLNESS – ventional thinking that VAP mostly develops due to
A FAST COLLAPSE AND A DEFECTIVE contiguous spread of upper respiratory tract patho-
RECONSTRUCTION gens into the lower respiratory tract and highlights
Critical illness has been associated with rapid, seis- the gut as a probable pool of pathogens for secon-
&&
mic changes in gut microbial communities [12]. dary infections in the ICU [21 ]. We will next
These changes include decreased species diversity, review how gut microbiota may escape the confine-
loss of obligate anaerobes, emergence of Proteobac- ments of their anatomical compartment.
terial and other pathogenic species abundance,
which collectively shape the profile of a disease-
promoting pathobiome replacing the healthy gut THE ROUTE FROM THE GUT TO THE
&&
microbiome [13 ]. The drivers of the microbiome LUNGS
collapse and its maladaptive replacement by a path- The two major routes connecting the gut to the lungs
obiome are numerous, including inter-individual are the portal venous and the mesenteric lymphatic
variability in microbiome resilience, immune status, system (Fig. 1). The portal venous system drains
gut perfusion/oxygenation and permeability, as well venous blood from the digestive tract into the liver,
as critical care interventions, such as antibiotics, the major site for clearance of gut-derived microbial
opioids, proton pump inhibitors and nutritional fragments, such as endotoxin, or even translocated
management (tube feedings, total parenteral nutri- bacteria. Kupffer cells, the liver resident macro-
&&
tion) [14 ,15]. Although no commonly agreed def- phages, have highly efficient phagocytic capacity
inition or diagnostic test exist for detection of a gut compared to alveolar and interstitial macrophages
pathobiome or broadly-defined gut dysbiosis, [24], and effectively scavenge the translocating
altered microbiota profiles in critically-ill patients microbial burden. However, the capacity of this filter-
are predictive of secondary infections including ing system can be exceeded (e.g. by excessive

1070-5295 Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved. www.co-criticalcare.com 133

Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.


Gastrointestinal system

bacterial translocation, Kupffer cell dysfunction) or Although, gut translocation is a biologically


by-passed (such as in porto-systemic shunts in highly plausible and experimentally reproducible
patients with cirrhosis), allowing for translocating phenomenon, documenting and quantifying gut
microbes or microbial products to reach the lungs microbial translocation in the human host is a rather
[25,26]. challenging task. First, accessing the portal venous or
The mesenteric lymphatic system facilitates mesenteric lymphatic systems for biospecimen acquis-
absorption of fluid and nutrients from the gut, trans- ition are extremely invasive and technically challeng-
porting lymph to the superior mesenteric lymph ing procedures, effectively infeasible for clinical care or
duct and finally the thoracic duct. The thoracic duct large-scale clinical research [34,35]. Even if the optimal
Downloaded from http://journals.lww.com/co-criticalcare by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XM

enters the systemic (blood) circulation at the junc- samples were available (e.g. portal venous blood or
tion of the left subclavian and internal jugular veins, mesenteric lymph), detection of bacteria with conven-
i0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 05/03/2023

draining into the right atrium and then directly into tional microbiologic cultures is likely insensitive, as
&&
the pulmonary circulation [27,28 ]. Hence, the many of the translocating organisms may be difficult
lungs are the first organs to be exposed to mesenteric to grow anaerobic bacteria [36], whereas the ultra-
lymph and its contents. Mechanistic studies in ani- sensitive culture-independent microbiota sequencing
mal models have highlighted the relevance of the approaches (e.g. 16S rRNA gene amplification) are
mesenteric lymphatic system in the gut–lung axis. vulnerable to contamination risks. Lastly, transloca-
For example, in murine models of acute respiratory tion is likely an episodic and not a steady phenom-
distress syndrome (ARDS) and traumatic/hemorrha- enon, and intermittent sampling can miss it. For
gic shock with severe abdominal infection, ligation example, in experimental shock models, gut perme-
of gut lymph vessels or drainage of gut lymph fluid ability and mesenteric lymph toxicity peaked in the
prevented lung injury [29]. Although both the immediate hours following shock, hence subsequent
venous and lymphatic system are possible conduits assessment of blood or mesenteric lymph may fail to
in a gut–lung axis, the relative contributions of each detect translocating bacteria [37]. Thus, the ability to
route in different syndromes or phases of critical study translocation in the critically ill host in practical
illness are not well defined. and reproducible ways is limited, and we have to rely
on surrogate metrics, such as paired microbiome anal-
ysis of gut and blood samples as well as comparing
TRANSLOCATION OF GUT MICROBES lung and gut microbiota for emergent similarities
In multiorgan failure during critical illness, the gut during critical illness. Recent research in coronavirus
may fail not only in its primary mission of nutrient disease 2019 (COVID-19) has provided proof-of-con-
absorption, but also in maintaining barrier integrity, cept evidence for gut translocation. In mouse models,
i.e. sealing the host circulation from the abundant severe acute respiratory syndrome coronavirus 2
&&
microbiome in the intestinal lumen [30,31 ]. Many (SARS-CoV-2) infection induced gut dysbiosis and
factors can lead to intestinal barrier damage and altered barrier permeability, whereas paired analysis
increased permeability [32,33], including mesen- of gut and blood samples from antibiotic-treated
teric hypoperfusion, increased intra-abdominal COVID-19 patients revealed proliferation of patho-
pressure, hypoxemia, inflammation, microcircula- genic bacterial genera in the gut, as well as increased
tory disturbances, as well as nutritional deprivation risk of bloodstream infections by such pathogenic
&&
with reduced SCFA production, which serve as fuel microbiota [38 ]. Additionally human studies in
for colonic epithelia. On the luminal surface, patients with ARDS have shown that the lung micro-
decreased production, altered composition or biome is enriched with gut-associated bacteria, which
increased degradation of the glycoprotein mucus may have engrafted in the lungs following the venous
allows bacteria to adhere to the epithelial surface or lymphatic route, and not via oropharyngeal aspira-
&& && &
[31 ]. At the epithelial level, the physical barrier is tion [39 ,40 ]. Similarly, in murine models of sepsis,
compromised by increased apoptosis of intestinal the lung microbiome was enriched with bacteria
epithelial cells, impaired renewal due to diminished found in the murine gut such as members of the
proliferation and migration, and altered expression Bacteroidales order, Enterococcus and Lachnospiraceae
&& &
and regulation of tight junction proteins, cumula- species [39 ,40 ].
tively creating a permeable, inefficient physical bar-
&&
rier [31 ]. Such host physiologic derangements
coupled by proliferation of disease-promoting bac- TRANSLOCATION OF MICROBIAL
teria and/or induction of virulence in commensals COMPONENTS AND METABOLITES
&&
organisms [13 ] can lead to translocation of living Apart from living microbes, their cellular constituents
organisms to either the venous circulation and/or or metabolic products may also travel along the gut–
the mesenteric lymphatic system. lung routes and influence host biology. Pathogen-

134 www.co-criticalcare.com Volume 29  Number 2  April 2023

Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.


Gut--lung crosstalk during critical illness Nath et al.

associated molecular patterns (PAMPs) [41], including transport not only microbial, but also gut-derived,
bacterial or fungal cell wall components, lipoproteins, host mediators to the pulmonary circulation. Such
carbohydrates, nucleic acids etc., represent signals host-derived messengers of the gut–lung axis include
recognized by pattern recognition receptors on damage-associated molecular patterns (DAMPs),
immune cells, particularly macrophages, leading to such as cellular fragments of dying cells, cell-free
cell activation and release of inflammatory mediators. nuclear or mitochondrial DNA, histones, ATP etc.,
The prototypical PAMP is lipopolysaccharide (LPS), which can trigger sterile inflammatory responses.
the endotoxin found on the outer cell wall of gram- Furthermore, other host molecules influenced by
negative bacteria, which causes increased neutrophil gut microbiota interactions, such as gut-derived hor-
Downloaded from http://journals.lww.com/co-criticalcare by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XM

migration, higher myeloperoxidase activity, and cyto- mones (e.g. incretins), extracellular vesicles and cyto-
kine levels in the lungs leading to epithelial cell dam- kines can also exert immunomodulatory and
i0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 05/03/2023

&&
age and permeability alveolar edema [42,43]. Among vascular permeability effects in the lungs [28 ,41].
critically ill patients without evidence of invasive The gut–lung axis can be influenced not only by
fungal infection, plasma levels of the fungal PAMP circulating host molecules, but also by migrating
BDG were significantly associated with the marker of immune cells. An important example involves the
intestinal permeability intestinal fatty acid binding interaction between segmented filamentous bacte-
protein-2, the hyper-inflammatory subphenotype of ria (SFB) and mucosal immune responses. T-helper
systemic host responses, higher mortality and longer 17 (Th17) and T-regulatory (Treg) cells are fre-
times for liberation from mechanical ventilation quently found at barrier surfaces such as intestinal
& &&
[7 ,44 ]. Such studies provide indirect evidence of mucosa, lungs and skin where they function to
the role of gut-origin circulating PAMPs on lung phys- protect the host from pathogenic microorganisms
iology and outcomes. and to restrain excessive effector T-cell responses,
&&
In contrast to gut derived PAMPs, microbial respectively [49 ]. Tregs contribute to the resolu-
metabolites, such as SCFAs, bile acids and desamino- tion phase of ARDS, while Th17 cells can cause
tyrosine can have beneficial effects on lung immun- systemic inflammation by producing interleukin
ity. There is evidence that gut derived SCFAs help in (IL)-17, which is elevated in patients with sepsis-
lung clearance of bacteria, modulated by down- related ARDS [50]. Additionally, higher Th17 to Treg
stream effects of G protein-coupled receptor 43 cell ratio in the systemic circulation is associated
(GPR43)- and Late Endosomal/Lysosomal Adaptor, with poorer prognosis in ARDS [51]. SFB are com-
MAPK And MTOR Activator 2 (LAMTOR2)-depend- mensal organisms in the gut, promoting develop-
ent signaling pathways. Activation of GPR43 leads ment of Th17 cells locally for enhanced resistance to
&&
to increased production of reactive oxygen species, intestinal pathogens [11,49 ]. Similarly, SFB can
activation of inflammasome and increased phagocy- regulate the CD4þ T-cell polarization into the
&
tosis during bacterial pneumonia [45 ]. Similarly, Th17 pathway, which when overexpressed can exac-
LAMTOR2-dependent signal pathway promotes erbate pulmonary fungal infections and autoim-
&
macrophage elimination of K. pneumoniae [46 ]. Con- mune lung conditions. This plausible pathway
versely, in murine models of antibiotic-induced dys- linking systemic T cell responses in ARDS and sepsis
biosis with decreased SCFA production, inoculation with gut microbiota represents another dimension
with Respiratory Syncytial Virus results in less effec- of the gut–lung axis that may be amenable to ther-
tive immune response and viral clearance in lungs apeutic manipulation [52].
&
[47 ]. In COVID-19 patients upon admission to the
ICU, reduced levels of fecal microbial metabolites,
such as secondary bile acids and desaminotyrosine, CONCLUSION – THE ROAD TOWARDS
were predictive of more severe respiratory failure and THERAPEUTIC MANIPULATION OF THE
mortality, with postulated impact on T- regulatory GUT–LUNG AXIS
(Treg) cell development and interferon-I signaling The vascular routes connecting the gut and lungs,
&&
[48 ]. The demonstrated protective roles of microbial along with evidence from epidemiologic observa-
metabolites, such as SCFAs and bile acids, suggest tions, clinical studies and animal model experi-
that the optimal gut–lung interaction is not only ments provide compelling evidence supporting
about preventing a pathobiome, but also promoting the presence and function of a gut–lung axis in
a healthy gut microbiome. critical illness. However, we are far from sufficient
understanding of the driving principles to confi-
dently intervene with the gut–lung axis. Even
GUT–MUCOSA INTERACTIONS though clinical interventions targeting an improve-
Following intestinal injury during local or systemic ment in composition and function of the gut micro-
stress, mesenteric lymph or portal venous blood can biome to reduce lung injury and inflammation may

1070-5295 Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved. www.co-criticalcare.com 135

Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.


Gastrointestinal system

seem a logical next step, many of the underlying REFERENCES AND RECOMMENDED
mechanisms need to be further clarified before we READING
Papers of particular interest, published within the annual period of review, have
can make educated decisions on who to target with been highlighted as:
what intervention. & of special interest
&& of outstanding interest
In order to establish the direction of gut–lung
interaction, we will require investigation with tech- 1. Joannidis M, Forni LG, Klein SJ, et al. Lung–kidney interactions in critically ill
niques such as paired metagenomic comparisons of patients: consensus report of the Acute Disease Quality Initiative (ADQI) 21
Workgroup. Intensive Care Med 2020; 46:654–672.
gut and lung microbiota and the use of labeled 2. Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the
‘tracer’ bacteria in gnotobiotic animals. Further- ratio of bacterial to host cells in humans. Cell 2016; 164:337–340.
3. Dickson RP, Erb-Downward JR, Martinez FJ, Huffnagle GB. The microbiome
Downloaded from http://journals.lww.com/co-criticalcare by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XM

more, secondary analysis of randomized controlled and the respiratory tract. Annu Rev Physiol 2016; 78:481–504.
trials or instrumental variable analyses with gut- 4. Gilbert JA, Blaser MJ, Caporaso JG, et al. Current understanding of the human
i0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 05/03/2023

microbiome. Nat Med 2018; 24:392–400.


targeted interventions could further enhance our 5. Hakansson AP, Orihuela CJ, Bogaert D. Bacterial–host interactions: physiology
understanding of the direction of effect. Addition- and pathophysiology of respiratory infection. Physiol Rev 2018; 98:781–811.
6. Bowerman KL, Rehman SF, Vaughan A, et al. Disease-associated gut
ally, to elucidate the route and the most relevant & microbiome and metabolome changes in patients with chronic obstructive
messengers, animal models are likely required due pulmonary disease. Nat Commun 2020; 11:5886.
Study looking at gut microbiome and metabolic profiles in COPD patients. A total
to the difficulty in sampling portal and lymphatic of 28 COPD patients (54% female) and 29 healthy controls were compared. Fecal
systems in critically ill patients. We should also not microbiome analysis showed dysbiosis with certain microbes such as members of
the family Lachnospiraceae correlated with reduced lung function.
only focus on the bacterial kingdom, but remain 7. Gorgone M, Singhvi D, Mehdi Nouraie S, et al. Circulating 1,3-beta-D-glucan
aware that trans-kingdom interactions are likely & is associated with lung function, respiratory symptoms, and mediators of
matrix degradation in chronic obstructive pulmonary disease. Chronic Obstr
important [53]. Pulm Dis (Miami) 2022; 9:325–335.
A common issue in critical care practice is the This study looked at COPD patients and correlation of symptoms, lung function
with 1,3-beta-D-glucan (BDG) a component of fungal cell wall. It was found that
extensive use of often empiric, broad-spectrum higher plasma levels of a pathogen-associated molecular pattern, BDG correlate
antibiotics. Recent retrospective research in crit- with worse lung function, greater respiratory morbidity, and circulating markers of
matrix degradation.
ically ill patients has shown that use of antianaero- 8. Sprooten RTM, Lenaerts K, Braeken DCW, et al. Increased small intestinal
bic antibiotics is associated with decreased overall permeability during severe acute exacerbations of COPD. Respiration 2018;
95:334–342.
survival and increased risk of VAP, with detectable 9. Arpaia N, Campbell C, Fan X, et al. Metabolites produced by commensal
impact on gut microbial communities and lower bacteria promote peripheral regulatory T-cell generation. Nature 2013;
&& 504:451–455.
respiratory tract microbiology [23 ]. Such research 10. Trompette A, Gollwitzer ES, Yadava K, et al. Gut microbiota metabolism of
shows that even when governed by principles of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med
2014; 20:159–166.
targeting and stewardship, clinical choices on anti- 11. Halnes I, Baines KJ, Berthon BS, et al. Soluble fibre meal challenge reduces
biotics from different classes are undertaken with airway inflammation and expression of GPR43 and GPR41 in asthma.
Nutrients 2017; 9:.
limited appreciation of their off-target effects on 12. McDonald D, Ackermann G, Khailova L, et al. Extreme dysbiosis of the
the gut microbiome. microbiome in critical illness. mSphere 2016; 1: Analysis of microbiomes
of 115 critically ill subjects that demonstrated rapid dysbiosis in multiple
Overall, our understanding of the gut–lung axis compartments after ICU admission.
has significantly increased over the recent years. 13. Alverdy JC, Krezalek MA. Collapse of the microbiome, emergence of the
&& pathobiome, and the immunopathology of sepsis. Crit Care Med 2017;
Once the secrets of this relationship are unlocked, 45:337–347.
it may provide novel treatment strategies and can Highly relevant article exploring the novel concept of pathobiome. The authors
introduce a concept of an ecologic collapse which is rapid and leads to emergence
shape our choices in antimicrobial treatment strat- of disease promoting microbe communities called ‘‘pathobiome’’ in critical illness
egies. Translational research is needed to improve such as sepsis.
14. Miniet AA, Grunwell JR, Coopersmith CM. The microbiome and the immune
our understanding of the gut–lung axis as well as && system in critical illness. Curr Opin Crit Care 2021; 27:157–163.
methods of reinstatement of symbiosis. Review article looking at the role of pathobiome in immune responses during
critical illness. Development of pathobiome in the critically ill gut leads to dysre-
gulated immune responses.
Acknowledgements 15. Ashley SL, Sjoding MW, Popova AP, et al. Lung and gut microbiota are altered
by hyperoxia and contribute to oxygen-induced lung injury in mice. Sci Transl
None. Med 2020; 12:.
16. Taur Y, Xavier JB, Lipuma L, et al. Intestinal domination and the risk of
&& bacteremia in patients undergoing allogeneic hematopoietic stem cell trans-
Financial support and sponsorship plantation. Clin Infect Dis 2012; 55:905–914.
Longitudinal research done evaluating use of antibiotics and fecal microbiome
Lieuwe D.J. Bos receives research funding from Amster- samples. Findings of this study showed that intestinal microbial dysbiosis was
dam UMC., Longfonds, ERS, ZonMW and the Innova- associated with higher risk of bacteremia in patients undergoing stem cell
transplantation.
tive Medicine Initiative. Dr Kitsios receives funding 17. Shimasaki T, Seekatz A, Bassis C, et al. Increased relative abundance of
support from the National Institutes of Health: K23- Klebsiella pneumoniae Carbapenemase-producing Klebsiella pneumoniae
within the gut microbiota is associated with risk of bloodstream infection in
HL139987 and R03-HL162655. long-term acute care hospital patients. Clin Infect Dis 2019; 68:2053–2059.
18. Frencken JF, Wittekamp BHJ, Plantinga NL, et al. Associations between
& enteral colonization with Gram-negative bacteria and intensive care unit-
Conflicts of interest acquired infections and colonization of the respiratory tract. Clin Infect Dis
2018; 66:497–503.
Dr Kitsios has received research funding from Karius, Inc. Research in critically ill patients showing the association of gut dysbiosis and
The remaining authors have no conflicts of interest. secondary infection with gram negative bacteremia.

136 www.co-criticalcare.com Volume 29  Number 2  April 2023

Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.


Gut--lung crosstalk during critical illness Nath et al.

19. Torres A, el-Ebiary M, González J, et al. Gastric and pharyngeal flora in 38. Bernard-Raichon L, Venzon M, Klein J, et al. Gut microbiome dysbiosis in
nosocomial pneumonia acquired during mechanical ventilation. Am Rev && antibiotic-treated COVID-19 patients is associated with microbial transloca-
Respir Dis 1993; 148:352–357. tion and bacteremia. Nat Commun 2022; 13:5926.
20. Bos LD, Stips C, Schouten LR, et al. Selective decontamination of the Translational research providing evidence of gut dysbiosis in COVID-19 leading to
&& digestive tract halves the prevalence of ventilator-associated pneumonia translocation and secondary bacteremia, combining animal models of SARS-CoV-
compared to selective oral decontamination. Intensive Care Med 2017; 2 infection and paired gut and blood biospecimen analysis from critically ill COVID-
43:1535–1537. 19 patients.
Single-center study that showed superiority of SDD over SOD. This study 39. Dickson RP, Singer BH, Newstead MW, et al. Enrichment of the lung
compared the occurrence of VAP in patients who either received selective && microbiome with gut bacteria in sepsis and the acute respiratory distress
decontamination of the digestive tract (SDD) or selective oropharyngeal decon- syndrome. Nat Microbiol 2016; 1:16113.
tamination (SOD). The regime of SOD was associated with a twice as high Landmark paper that showed indirect evidence of gut lung bacterial translocation.
occurrence of VAP compared to the SDD regime. This was the first study This research was one of the first to use culture independent techniques such as
comparing SDD to SOD using likelihood scoring for VAP. bacterial DNA isolation in murine models of sepsis as well as in ARDS patients. It
21. Plantinga NL, de Smet AMGA, Oostdijk EAN, et al. Selective digestive and was found that the lung microbiome is enriched with gut associated bacteria in
Downloaded from http://journals.lww.com/co-criticalcare by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XM

&& oropharyngeal decontamination in medical and surgical ICU patients: indivi- both murine models of sepsis and ARDS patients. This paper also showed the
dual patient data meta-analysis. Clin Microbiol Infect 2018; 24:505–513. shortcomings of standard culture techniques in trying to establish gut lung
i0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 05/03/2023

Meta-analysis of 6 studies that compared SDD and SOD in medical and surgical translocation.
patients. SDD and SOD led to improved hospital and ICU survival when compared 40. Panzer AR, Lynch SV, Langelier C, et al. Lung microbiota is related to smoking
to standard care in both patient populations. SDD was also more effective than & status and to development of acute respiratory distress syndrome in critically
SOD in improving hospital and ICU survival. ill trauma patients. Am J Respir Crit Care Med 2018; 197:621–631.
22. Oostdijk EAN, Kesecioglu J, Schultz MJ, et al. Effects of decontamination of Human study demonstrating enrichment of gut-origin bacteria in lung microbial
&& the oropharynx and intestinal tract on antibiotic resistance in ICUs: a rando- communities of patients with ARDS.
mized clinical trial. JAMA 2014; 312:1429–1437. 41. Tang D, Kang R, Coyne CB, et al. PAMPs and DAMPs: signal 0 s that spur
Landmark paper comparing SDD vs SOD. This was a large randomized clinical trial autophagy and immunity. Immunol Rev 2012; 249:158–175.
that looked at primary outcome of antibiotics resistance incidence and secondary 42. Rocco PRM, Nieman GF. ARDS: what experimental models have taught us.
outcomes of day 28 mortality, ICU mortality, hospital mortality, length of stay, and Intensive Care Med 2016; 42:806–810.
rate of candidemia. SDD led to lower day-28 mortality, rectal carriage of antibiotic- 43. Parsons PE, Worthen GS, Moore EE, et al. The association of circulating
resistant gram-negative bacteria, and ICU-acquired bacteremia. endotoxin with the development of the adult respiratory distress syndrome.
23. Chanderraj R, Baker JM, Kay SG, et al. In critically ill patients, antianaerobic Am Rev Respir Dis 1989; 140:294–301.
&& antibiotics increase risk of adverse clinical outcomes. Eur Respir J 2022; 61:. 44. Kitsios GD, Kotok D, Yang H, et al. Plasma 1,3-b-D-glucan levels predict
Retrospective research in critically ill patients showing worse outcomes with use of && adverse clinical outcomes in critical illness. JCI Insight 2021; 6:.
antianaerobic antibiotics. Early treatment with antianaerobic antibiotics was asso- Translational study in critically-ill patients without fungal infections showing the
ciated with increased mortality, increased risk of VAP and gut microbiota enrich- plasma levels of the fungal cell-wall constituent beta-D-glucan were significantly
ment for Enterobacteriaceae. associated with markers of intestinal permeability, the hyper-inflammatory sub-
24. Laskin DL, Weinberger B, Laskin JD. Functional heterogeneity in liver and lung phenotype of systemic host responses and worse outcome.
macrophages. J Leukoc Biol 2001; 70:163–170. 45. Galvão I, Tavares LP, Corrêa RO, et al. The metabolic sensor GPR43 receptor
25. Rimola A, Soto R, Bory F, et al. Reticuloendothelial system phagocytic activity & plays a role in the control of Klebsiella pneumoniae infection in the lung. Front
in cirrhosis and its relation to bacterial infections and prognosis. Hepatology Immunol 2018; 9:142.
1984; 4:53–58. Animal model research looking at a gut microbiome metabolite SCFA’s effect in
26. Triantafyllou E, Gudd CLC, Mawhin M-A, et al. PD-1 blockade improves immune responses in lung. This article showed that gut derived SCFAs play a role
Kupffer cell bacterial clearance in acute liver injury. J Clin Investig 2021; 131:. in control of Klebsiella pneumonia infection in the lung through the effect on
27. Deitch EA. Gut lymph and lymphatics: a source of factors leading to organ metabolic Sensor GPR43 receptor.
injury and dysfunction. Ann N Y Acad Sci 2010; 1207(Suppl 1):E103–E111. 46. Wu T, Li H, Su C, et al. Microbiota-derived short-chain fatty acids promote
28. Ma Y, Yang X, Chatterjee V, et al. The gut–lung axis in systemic inflammation. & LAMTOR2-mediated immune responses in macrophages. mSystems 2020;
&& Role of mesenteric lymph as a conduit. Am J Respir Cell Mol Biol 2021; 5:.
64:19–28. Animal model research looking at a gut microbiome metabolite SCFA’s effect in
A narrative review article addressing mesenteric lymphatic system’s major role in immune responses in lung. This article showed that gut derived SCFAs affect
gut–lung axis. Here the authors go over the evidence on how mesenteric lymphatic immune responses in macrophages via the LAMTOR-2 pathway.
system acts as the gut lung route for microbe, microbial metabolites and fragments 47. Antunes KH, Fachi JL, de Paula R, et al. Microbiota-derived acetate protects
as well as microbe host interaction products. & against respiratory syncytial virus infection through a GPR43-type 1 interferon
29. Jin C, Chen J, Gu J, Zhang W. Gut–lymph–lung pathway mediates sepsis- response. Nat Commun 2019; 10:3273.
induced acute lung injury. Chin Med J 2020; 133:2212–2218. Animal model research examining the antiviral effects of acetate, a gut microbial
30. Reintam Blaser A, Preiser J-C, Fruhwald S, et al. Gastrointestinal dysfunction metabolite in respiratory syncytial virus infections. Acetate supplementation and
in the critically ill: a systematic scoping review and research agenda proposed diet promoting acetate production lead to faster resolution of RSV infection
by the Section of Metabolism, Endocrinology and Nutrition of the European 48. Stutz MR, Dylla NP, Pearson SD, et al. Immunomodulatory fecal metabolites
Society of Intensive Care Medicine. Crit Care 2020; 24:224. && are associated with mortality in COVID-19 patients with respiratory failure.
31. Otani S, Coopersmith CM. Gut integrity in critical illness. J Intensive Care Nat Commun 2022; 13:6615.
&& 2019; 7:17. Translational study in critically ill patients with COVID-19 showing that decreased
A narrative review on factors involved in maintaining gut integrity during home- concentrations of microbial metabolites, such as bile acids and desaminotyrosine,
ostasis and factors leading to development of permeability in critical illness. in baseline fecal samples upon ICU admission were associated with progression of
32. Yoseph BP, Klingensmith NJ, Liang Z, et al. Mechanisms of intestinal barrier respiratory failure and mortality.
dysfunction in sepsis. Shock 2016; 46:52–59. 49. Omenetti S, Pizarro TT. The treg/th17 axis: a dynamic balance regulated by
33. Berger G, Guetta J, Klorin G, et al. Sepsis impairs alveolar epithelial function && the gut microbiome. Front Immunol 2015; 6:639.
by downregulating Na-K-ATPase pump. Am J Physiol Lung Cell Mol Physiol Narrative review article examining the relationship between the gut microbiome
2011; 301:L23–L30. and T reg/Th17 cells.
34. Moore FA, Moore EE, Poggetti R, et al. Gut bacterial translocation via the 50. Ding Q, Liu G-Q, Zeng Y-Y, et al. Role of IL-17 in LPS-induced acute lung
portal vein: a clinical perspective with major torso trauma. J Trauma 1991; injury: an in vivo study. Oncotarget 2017; 8:93704–93711.
31:629–636; discussion 636. 51. Yu Z, Ji M, Yan J, et al. The ratio of Th17/Treg cells as a risk indicator in early
35. Banan B, Wei Y, Simo O, et al. Intestinal lymph collection via cannulation of acute respiratory distress syndrome. Crit Care 2015; 19:82.
the mesenteric lymphatic duct in mice. J Surg Res 2021; 260:399–408. 52. Gauguet S, D’Ortona S, Ahnger-Pier K, et al. Intestinal microbiota of mice
36. Lagier J-C, Khelaifia S, Alou MT, et al. Culture of previously uncultured members influences resistance to Staphylococcus aureus pneumonia. Infect Immun
of the human gut microbiota by culturomics. Nat Microbiol 2016; 1:16203. 2015; 83:4003–4014.
37. Adams CA, Xu DZ, Lu Q, Deitch EA. Factors larger than 100 kD in post- 53. Haak BW, Argelaguet R, Kinsella CM, et al. Integrative transkingdom analysis
hemorrhagic shock mesenteric lymph are toxic for endothelial cells. Surgery of the gut microbiome in antibiotic perturbation and critical illness. mSystems
2001; 129:351–363. 2021; 6.

1070-5295 Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved. www.co-criticalcare.com 137

Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.

You might also like