ArticlePDF AvailableLiterature Review

Stress Induces Endotoxemia and Low-Grade Inflammation by Increasing Barrier Permeability

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

Chronic non-communicable diseases (NCDs) are the leading causes of work absence, disability, and mortality worldwide. Most of these diseases are associated with low-grade inflammation. Here, we hypothesize that stresses (defined as homeostatic disturbances) can induce low-grade inflammation by increasing the availability of water, sodium, and energy-rich substances to meet the increased metabolic demand induced by the stressor. One way of triggering low-grade inflammation is by increasing intestinal barrier permeability through activation of various components of the stress system. Although beneficial to meet the demands necessary during stress, increased intestinal barrier permeability also raises the possibility of the translocation of bacteria and their toxins across the intestinal lumen into the blood circulation. In combination with modern life-style factors, the increase in bacteria/bacterial toxin translocation arising from a more permeable intestinal wall causes a low-grade inflammatory state. We support this hypothesis with numerous studies finding associations with NCDs and markers of endotoxemia, suggesting that this process plays a pivotal and perhaps even a causal role in the development of low-grade inflammation and its related diseases.
Content may be subject to copyright.
May 2015 | Volume 6 | Article 2231
HYPOTHESIS AND THEORY
published: 15 May 2015
doi: 10.3389/mmu.2015.00223
Frontiers in Immunology | www.frontiersin.org
Edited by:
Heiko Mühl,
University Hospital Goethe University
Frankfurt, Germany
Reviewed by:
Giamila Fantuzzi,
University of Illinois at Chicago, USA
Fulvio D’Acquisto,
Queen Mary University of London, UK
Kottarappat N. Dileepan,
The University of Kansas Medical
Center, USA
*Correspondence:
Karin de Punder,
Charité University Medicine
Berlin, Hufelandweg 14, 10117
Berlin, Germany
karin.de-punder@charite.de
Specialty section:
This article was submitted to
Inammation, a section of the journal
Frontiers in Immunology
Received: 13February2015
Accepted: 24April2015
Published: 15May2015
Citation:
de PunderK and PruimboomL
(2015) Stress induces endotoxemia
and low-grade inammation by
increasing barrier permeability.
Front. Immunol. 6:223.
doi: 10.3389/mmu.2015.00223
Stress induces endotoxemia and
low-grade inammation by
increasing barrier permeability
Karin de Punder
1,
2* and Leo Pruimboom
2
1 Institute of Medical Psychology, Charité University Medicine, Berlin, Germany, 2 Natura Foundation, Numansdorp,
Netherlands
Chronic non-communicable diseases (NCDs) are the leading causes of work absence,
disability, and mortality worldwide. Most of these diseases are associated with low-grade
inammation. Here, we hypothesize that stresses (dened as homeostatic disturbances)
can induce low-grade inammation by increasing the availability of water, sodium, and
energy-rich substances to meet the increased metabolic demand induced by the stressor.
One way of triggering low-grade inammation is by increasing intestinal barrier permeability
through activation of various components of the stress system. Although benecial to meet
the demands necessary during stress, increased intestinal barrier permeability also raises
the possibility of the translocation of bacteria and their toxins across the intestinal lumen
into the blood circulation. In combination with modern life-style factors, the increase in
bacteria/bacterial toxin translocation arising from a more permeable intestinal wall causes
a low-grade inammatory state. We support this hypothesis with numerous studies nding
associations with NCDs and markers of endotoxemia, suggesting that this process plays
a pivotal and perhaps even a causal role in the development of low-grade inammation
and its related diseases.
Keywords: endotoxemia, endotoxin, inammation, intestinal permeability, lipopolysaccharide, stress, tight
junction
Introduction
Inammation is the response of the innate immune system triggered by stimuli like microbial pathogens
and injury. Acute systemic inammation such as in sepsis, trauma, burns, and surgery is characterized
by a quick increase in plasma levels (up to 100-fold) of pro-inammatory cytokines and acute phase
proteins, while in low-grade inammation, there is a sustained but only two to threefold increase
in circulation pro-inammatory mediators (1). Chronic low-grade inammation is characteristic
for many non-communicable diseases (NCDs) including diabetes type II, cardiovascular disorders,
autoimmune diseases, chronic fatigue syndrome, depression, and neurodegenerative pathologies,
but until now the exact mechanism behind the elevated levels of inammatory mediators found in
these conditions is not well understood (25).
Inammation can be induced by the binding of pathogen-associated molecular patterns (PAMPs)
to toll-like receptors (TLRs), which are expressed on dierent cells types including immune cells,
adipocytes, and endothelial cells. e most extensively studied PAMP is lipopolysaccharide (LPS)
or endotoxin (the terms LPS and endotoxin will be used interchangeably throughout the rest of
the article), a major cell wall component of Gram-negative bacteria, which is normally present
May 2015 | Volume 6 | Article 2232
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
in the human circulation in very low concentrations. It has
been hypothesized that most of this circulating LPS is derived
from the gut, since the gut-microbiota is the biggest source of
Gram-negative bacteria-derived LPS. However, LPS found in the
circulation could also be derived from Gram-negative bacteria
residing in the oral cavity, respiratory, and genitourinary tracts,
or can be food-derived (68). Under certain circumstances, there
can be an increase of endotoxin translocation across the intestinal
barrier, leading to mildly increased concentrations in the blood
circulation. is process has been associated with several NCDs,
like depression (9), chronic fatigue syndrome (10), chronic heart
failure (11), type 2 diabetes (12), autism (13), non-alcoholic fatty
liver disease (NAFLD) (14), and inammatory bowel disease (IBD)
(15), diseases that are all linked to chronic systemic low-grade
inammation, indicating that endotoxemia could be an important
contributor in the development of these conditions.
Here, we hypothesize that stress-induction leads to a more
permeable intestinal wall intended to facilitate an increase in the
availability of water, sodium, and energy-rich substances necessary
to meet the increased metabolic demand induced by the stressor.
Modern life-style factors, such as long-term psychosocial stress
and components of our “Western” diet constantly challenge the
stress-axis and further compromise intestinal barrier function,
resulting in endotoxemia, low-grade inammation, and its related
diseases. We support our hypothesis by describing literature sur-
rounding stress- and immune system-activation processes and
their relation to gut barrier function and explain how life-style
choices impact all these systems. In addition, we present a vast
amount of literature describing associations with NCDs and
markers of endotoxemia. Overall, we conclude that stress-induced
disrupted barrier function in parallel with elevated circulating
endotoxin levels may underlie disease onset and progression and
should be considered much more than just a risk factor for chronic
disease; it could be a cause.
Bacterial Toxins Activate the Immune
System via TLRs
Lipopolysaccharide, the major cell wall component of Gram-
negative bacteria, is characterized by its capacity to induce
inammation, fever, shock, and death (1). Additionally in recent
years, other cell wall components of Gram-negative and -positive
bacteria have been recognized to have endotoxic properties (16),
but these will not be further addressed in the rest of the paper.
Endotoxins are released from bacteria during infection or as a
consequence of bacterial lysis. Although both whole bacteria
and bacterial toxins can translocate transcellular or paracellular
into the lymph, blood, and mesenteric lymph nodes, it is still not
precisely clear if the presence of endotoxin in the blood circulation
(endotoxemia) also presents whole bacteria translocation across
the intestinal wall (17).
Inammation can be induced by the binding of LPS to TLR4.
e lipid-A moiety of LPS interacts with the LPS-sensing machin-
ery composed of TLR4, myeloid dierential protein 2, CD14, and
LPS-binding protein (LBP). LBP transports and delivers circulating
aggregates of LPS to lipoproteins, resulting in hepatic clearance, or
delivers LPS to CD14 (the membrane-bound or secreted, soluble
form of this molecule), leading to TLR4 activation. TLR4 activation
activates two transcription factors, activator protein (AP)-1 and
nuclear factor κB (NF-κB) (18, 19), and stimulates the production
of pro-inammatory mediators such as prostaglandin 2 (PGE2)
(20), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6,
interferon (IFN)-γ, and the acute phase protein, C-reactive protein
(CRP) (19). Simultaneously, an uncontrolled pro-inammatory
reaction is prevented by the induction of TLR4, NF-κB, and AP-1
signaling inhibitors, which are probably involved in creating
endotoxin tolerance (21). LPS tolerance is dened as a reduced
responsiveness to a LPS challenge following a rst encounter of
endotoxin (22). It has been suggested that the dose of LPS exposure
is important for determining the switch between LPS tolerance and
priming. For example, in macrophages, high LPS concentrations
induced a robust pro-inammatory response in parallel with the
activation of inhibitory feedback mechanisms. Lower concentra-
tions of LPS, like those observed in NCDs, removed transcriptional
suppressors on the promoters of pro-inammatory genes and
induced a mild but persistent expression of pro-inammatory
mediators (21, 23).
Intestinal Barrier Function
The Paracellular Pathway is Important for Water,
Mineral, and Nutrient Uptake
e intestinal barrier allows for the regulated uptake of water,
minerals, and nutrients and protects the gut lumen from damage
due to harmful substances. Components can cross the epithelial
barrier by active transport and endocytosis (transcellular) or via
the paracellular route. Because hydrophilic solutes are limited to
cross lipid membranes of epithelial cells, the paracellular route is
an important and major route for the transport of water, solutes,
and minerals across the intestinal barrier (24, 25). Active glucose,
sodium, and water uptake is mediated by the activity of sodium-
dependent glucose co-transporters (SGLTs) (26). e transcellular
absorption of glucose and sodium and the resulting basolateral
disposition of glucose and sodium by these transporters opens
up the paracellular pathway structure and allows the passive ow
of water and small nutrients by creating an osmotic gradient (27).
Intestinal permeability is a measure of the barrier function
of the gut and relates to the paracellular space surrounding the
brush border surface of the enterocytes and the junctional com-
plexes (28). e junctional complex, containing tight junctions,
adherens junctions, and desmosomes is an important regulator of
the paracellular pathway and allows the passage of water, solutes,
and ions, but under normal conditions provides a barrier to larger
molecules (28, 29). e claudin family of junctional transmem-
brane proteins has a substantial eect on paracellular permeability.
While one group of sealing claudins makes the paracellular barrier
less permeable, the other group of claudins is known to increase
paracellular permeability by the formation of pores that increase
permeability for small solutes (30, 31). e expression of claudin
proteins varies between tissues, explaining the variances in per-
meability of tight junctions among tissues (27). e paracellular
pathway can be divided into the pore and non-pore pathway. e
pore pathway is mainly controlled by the expression of claudins,
while the non-pore pathway is more sensitive to cytoskeletal
May 2015 | Volume 6 | Article 2233
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
disruptions (30). Cytoskeletal rearrangements can be induced
by phosphorylation of the regulatory myosin light chain
(MLC), induced by MLC-kinase (MLCK). Phosphorylation
of the MLC facilitates myosin binding to actin and therefore
aids in cytoskeletal contractility. MLCK can be activated by
cytokines such as TNF-α, causing increases in tight junction
permeability by actomyosin contraction and reorganization
of the tight junction (32, 33). In addition, SGLT1 activation
and associated increases in tight junction permeability are
also paralleled with phosphorylation of MLC, indicating that
MLCK is an important mediator in tight junction and paracel-
lular permeability regulation (25, 34) (Figure1).
Increased intestinal permeability has been associated with
autoimmune diseases, such as type 1 diabetes (35), rheumatoid
arthritis, multiple sclerosis (36), and diseases related to chronic
inflammation, like IBD (36, 37), asthma (38), chronic fatigue
syndrome, and depression (10, 39). It has been hypothesized
that chronic intestinal hyper-permeability results in a pro-
inflammatory phenotype induced by the enhanced paracel-
lular translocation of microbial (and dietary) antigens across
the gut barrier (40).
Stress Increases Permeability of the
Intestinal Barrier
Stressful stimuli activate the sympathetic nervous system (SNS)
and hypothalamic–pituitary–adrenal (HPA)-axis. Activation of
both systems increases the availability of water, minerals, and
energy-rich substances in order to meet with the body’s metabolic
FIGURE 1 | MLC phosphorylation increases intestinal permeability.
Activation of the SNS increases intestinal permeability by stimulating the
activity of SGLT1 on epithelial cells. Activation of SGLT1 is paralleled by MLC
phosphorylation by MLCK, inducing actomyosin contraction and
reorganization of the tight junction. The resulting increase in paracellular
permeability raises the possibility of translocation of bacteria and/or their
toxins across the more permeable gut barrier. Pro-inammatory cytokines
produced by activated immune cells residing in the lamina propria further
increase intestinal permeability by activating MLCK. JC, junctional complex.
demand (41, 42). e SNS responds instantly to physical and
psychological stress by reallocating energy into dierent organs by
neuronal regulation of heart rate, blood ow, release of catecho-
lamines (adrenalin and noradrenalin) from the adrenal medulla
(43), and stimulation of the renin–angiotensin–aldosterone
system (44), involved in retention of water and sodium from the
kidneys. In addition to the kidneys, water and sodium reabsorp-
tion can also be achieved at the level of the intestine. e intestinal
wall is innervated by adrenergic sympathetic nerve bers that
upon stimulation increase water and sodium absorption (45, 46),
which is paralleled by increases in intestinal permeability. e
SNS-induced increase in permeability is likely mediated by β2-
adrenergic receptors expressed on epithelial cells (47). Activation
of the β2-adrenergic-receptors stimulated SGLT1-mediated glu-
cose absorption from the gut (48, 49) and the resulting basolateral
disposition of glucose and sodium by these transporters opens
up the paracellular pathway (27) (Figure 1). Not surprisingly,
blockage of the SNS by means of thoracic epidural anesthesia
resulted in the blockage of the endotoxin-induced increase in
intestinal permeability in rats (50).
Activation of the HPA-axis leads to the release of glucocor-
ticoids that potentiate some of the actions of catecholamines.
Essential to this response are the neurons in the paraventricular
nucleus of the hypothalamus expressing corticotropin-releasing
hormone (CRH) and other co-secretagogues, such as arginine
vasopressin (AVP) and oxytocin, both involved in the regulation of
water homeostasis. AVP and CRH trigger the immediate release of
adrenocorticotropic hormone (ACTH) from the anterior pituitary,
which in turn induces the release of glucocorticoids and to some
extend mineralocorticoids from the adrenal cortex, stimulating
gluconeogenesis and increasing sodium and water retention,
respectively (51, 52). Intestinal permeability is regulated by several
components of the HPA-axis.
In epithelial HT-29 monolayers, exposure to CRH resulted in
an increased response to LPS as reected by a decrease in transepi-
thelial resistance and a signicant increase in the expression of the
pore forming protein, claudin-2. Interestingly enough, these eects
were mediated by an increase in TLR4 expression, an observation
that could be repeated in mice treated with the water-avoid stressor
(53). TLR4 activation resulted in the activation of the transcription
factor NF-κB, which has specic binding sites in the claudin-2 gene
promoter (54), indicating that in epithelial cells CRH aects both
intestinal permeability and inammatory pathways.
In rats, exposure to restricted stress or swimming stress
increased intestinal permeability throughout the whole intestinal
tract as measured by the fractional secretion of the urinary recovery
of sucrose (reecting gastric permeability), the lactulose–mannitol
ratio (as a marker for small intestinal permeability), and sucralose
(reecting both small intestinal and colonic permeability) (55).
Other experimental animal stress models such as thermal injury
or early maternal deprivation induced the development of gastric
ulcers, altered gastrointestinal motility and ion secretion, and
increased intestinal permeability [reviewed by Caso etal. (56)].
SGLT1 expression was markedly increased in the rat jejunum and
ileum aer 8weeks of restraint stress. ese ndings were paral-
leled with an increase in intestinal lymphocytic inltration and
adrenal gland weight gain (26). e up-regulation of the SGLT1
May 2015 | Volume 6 | Article 2234
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
is probably necessary to meet with the increased water, sodium,
and nutrient demand, induced by chronic stress (42).
e eect of acute stress on intestinal permeability was also
investigated in humans (57). In healthy volunteers subjected to a
public speech test, high cortisol-responders displayed increased
intestinal permeability as measured by the lactulose–mannitol
ratio. Exogenous CRH administration also increased intestinal
permeability, yet the CRH-induced hyper-permeability could be
suppressed by the mast cell stabilizer disodium cromoglycate. Mast
cell stabilization before the public speech test also did not alter
intestinal permeability, however, it should be noted that in this
experiment, a control group was not included. Nevertheless, these
results identify CRH as an important factor in the stress-induced
alterations of the intestinal barrier function. ese alterations
seemed to be mediated by intestinal mast cells that upon activa-
tion secrete pro-inammatory mediators like IFN-γ and TNF-α.
A variety of pro-inammatory cytokines increases epithelial and
endothelial paracellular permeability by modulating the structure
of the tight junction and by inducing cytoskeletal disruptions via
activation of MLCK (32, 34, 58) (Figure1). For example, IFN-
γ increased epithelial permeability of T84 monolayers to large
molecules (10 kDa). Interestingly, the IFN-γ-induced increase
in permeability also up-regulated the passage of FITC-labeled-
endotoxin by 10-fold (59).
Neuroendocrine–Immune Interactions
e complex neuroendocrine–immune interactions are
evidenced by the fact that emotional stressors inuence the
immune response and that pure immunological stimuli impact
on cognitive performance (60). Inammatory mediators activate
the HPA-axis with the purpose to provoke disease behavior and
redirect energy-rich nutrients toward the immune system (61).
Cytokines have been shown to increase nutrient availability to meet
with the inammation-dependent increased metabolic demand.
For example, the cytokine IL-1α increased whole body glucose
metabolism on a central level (62) and cytokines like IL-6, TNF-α,
IL-1, and IFN independently evoke a HPA-axis response (6365).
Immune mediators can communicate with the brain via several
pathways. By stimulating aerent sensory nerve bers, by entering
the brain via the circumventricular organs or by binding to cerebral
blood vessel endothelium, immune mediators eectively redirect
energy-rich substrates toward the immune system (41, 42).
Besides inammatory cytokines, prostaglandins synthesized
via the cyclooxygenase system play a central role in inammation
and HPA-axis activation. Zimomra etal. (65) demonstrated that
in rats the initial activation of the HPA-axis by LPS is mediated by
prostaglandins, like PGE2, while inammatory cytokines maintain
corticosterone levels at later time-points. In this study, it was sug-
gested that prostaglandins stimulated corticosterone release in a
direct manner, since the peak in circulating corticosterone levels
was observed long before the peak in circulating ACTH. is idea
was conrmed by a study in rodents, showing that PGE2 directly
stimulated the release of glucocorticoids from the adrenal gland (66).
In human adrenal cells expressing TLR2 and TLR4, LPS stimulation
resulted in the release of cortisol. is eect was mediated by PGE2,
since inhibition of cyclooxygenase-2 attenuated cortisol release (67).
As indicated, TLR4 activation stimulates the release of PGE2
by immune cells, adipocytes, endothelial, epithelial, and prob-
ably also adrenal cells (68), inducing the peripheral release of
glucocorticoids from the adrenal gland (66). PGE2 also activates
glucocorticoid production through activation of the HPA-axis at
the level of the hypothalamus and the pituitary (69). Macrophages,
homing in blood vessels in the cranium, are directly activated by
danger signals such as LPS. Activation of these special macrophages
induces the production of PGE2 which directly stimulates the
paraventricular nucleus of the hypothalamus, leading to higher
production of glucocorticoids, which should probably protect
against possible inammation of the brain (69).
Acute Stress Stimulates Pro-Inammatory
Pathways by Increasing Intestinal
Permeability
Acute stress modulates the immune response and changes immune
cell distribution. ese neuroendocrine eects on the immune
system are mediated by stress-hormones released from the
adrenal gland, by direct innervation of sympathetic nerve bers
into lymphoid organs and by stress hormone receptors expressed
on immune cells, like glucocorticoid receptors (GRs) and α- and
β-adrenergic receptors (7072). It has been suggested that by
mobilizing immune cells, the stress response, also known as the
“ght–ight reaction,” prepares the immune system for oncoming
challenges (70).
In addition, acute stress increases circulating pro-inam-
matory mediators (7375). In subjects exposed to acute stress,
NF-κB was up-regulated in peripheral blood mononuclear cells
in parallel with elevated levels of circulating catecholamines and
glucocorticoids (76). Until now, it is not completely understood
what causes this pro-inammatory response. Glucocorticoids
mostly have an inhibitory eect on inammatory pathways and
catecholamines a rather modulating than activating inuence
on the immune system (71, 72, 77), however, it has been shown
that activation of the β-adrenergic receptor by noradrenalin (but
not adrenalin) increased NF-κB binding to DNA in monocytes
invitro (76). A recent study in rodents showed that acute stress-
induced neuro-inammation could be prevented by a pre-stress
treatment with antibiotics or an inhibitor of MLCK. In addition,
these treatments prevented stress-induced hyper-permeability
and endotoxemia, indicating that it is not the stress-factor itself
producing a pro-inammatory response of the immune system,
but the fact that stress increases barrier permeability and the
translocation of endotoxin. Pre-stress probiotic treatment with
Lactobacillus farciminis had similar eects, which could be
explained by its ability to enhance intestinal barrier function
(78). In agreement with these results, it could be hypothesized
that the (short-lasting) pro-inammatory activity in humans
observed during acute stress is initiated by a stress-induced
increase in intestinal permeability, mediated by the SNS and
components of the HPA-axis, and resulting in higher levels
of translocating endotoxin interacting with TLRs on immune
cells, adipocytes, and epithelial cells. A schematic overview of
the complex neuroendocrine–immune interactions and their
relation to gut barrier function are displayed in Figure2.
FIGURE 2 | The complex neuroendocrine–immune interactions and
their relation to gut barrier function. Stressors, including inammatory
mediators, activate the SNS and HPA-axis. Activation of the HPA-axis
stimulates neurons in the paraventricular nucleus of the hypothalamus to
secrete CRH and AVP that trigger the release of ACTH from the anterior
pituitary, resulting in the secretion of corticosteroids from the adrenal cortex.
CRH has been shown to affect intestinal permeability. SNS activation results
in the release of catecholamines from the adrenal medulla. The intestinal wall
is innervated by adrenergic sympathetic nerve bers that upon stimulation
increase water, sodium, and glucose absorption, paralleled by increased
intestinal permeability. The resulting increase in translocation of endotoxin
across the intestinal barrier can stimulate immune cells in the underlying
lamina propria to secrete pro-inammatory cytokines and prostaglandins like
PGE2. Inammatory mediators communicate with the brain by stimulating
afferent sensory nerve bers, by entering the brain via the circumventricular
organs or by binding to cerebral blood vessel endothelium. Continuous
stress-induced impairment of the intestinal barrier creates a vicious circle
whereby inammatory cytokines will persistently activate the SNS and
HPA-axis resulting in barrier disruption, increased endotoxin translocation,
and a pro-inammatory state.
May 2015 | Volume 6 | Article 2235
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
Chronic Stress Dysregulates the HPA-Axis
and Changes Immune Function
Chronic psychological stress is known to dysregulate the
immune system. These alterations are accompanied by low-
grade inflammation, delayed wound healing, and increased
susceptibility to infectious diseases (79). Chronic stress leads
to hypercortisolemia (77), long-term permeability of barriers,
endotoxemia, and low-grade inflammation (our hypothesis and
theory). Normally, the release of glucocorticoids puts a limit on
the maximum activity of the immune system; however, chronic
HPA-axis stimulation can result in glucocorticoid resistance
at the level of the immune system, making it insensitive to
its inhibitory and modulatory actions (2). This process is
observed in several conditions (including conditions related
to psychosocial stress), whereby immune cells from patients
are less responsive to the inhibitory actions of glucocorticoids
on cytokine release and cell proliferation after stimulation
in vitro (8083). In addition, chronic stress induces a shift
in the production of type 1 cytokines toward type 2 cytokine
production. It can be deducted that by this mechanism, the part
of the immune system involved in the clearance of extracellular
bacteria and bacterial toxins (the type 2 response) is prevented
from being suppressed and protection against ongoing micro-
bial infiltration (endotoxemia) is guaranteed, while the type 1
response, involved in clearance of intracellular pathogens (like
viruses) is inhibited (71, 84).
Life-Style-Related Factors Induce
Endotoxemia
The fact that stress increases barrier permeability and thereby
enhances the availability of water, sodium, and nutrients,
makes sense from an evolutionary perspective. However, the
question arises if the accompanied translocation of bacteria
and their toxins should also be considered beneficial for the
host. We sp eculate that when the composition of the microbiota
is physiological, and barrier opening is short-lasting, acute
stress will not produce low-grade inflammation. However,
modern people suffer from new multi-factorial stressors,
such as chronic psychosocial stress and the consumption
of a “Western diet,” which constantly challenge the stress-
axis, alter microbiota composition, and thereby compromise
intestinal barrier function. This next section discusses how
modern life-style factors impact the gut–brain–immune-axis
and promote endotoxemia, low-grade inflammation, and its
related diseases.
Gut-Microbiota Modulate Stress-Axis and Inuence
Gut Barrier Function
Large dierences in the composition of the gut-microbiota and an
overall reduction in microbial diversity are observed in Western
populations when compared to traditional Hunter-gatherers or
people from rural Africa (85, 86). ese environment and diet-
induced changes in gut-microbiota have been connected to an
increased susceptibility to chronic diseases, like IBD, obesity, and
type 1 and type 2 diabetes (87). e gut-microbiota inuences
inammatory (88) and metabolic processes (89) and has been
shown to inuence the development of the HPA-axis and immune
system (90, 91). For example, exposure to LPS during developmen-
tal periods can exaggerate the HPA-axis and immune response to
stress (92, 93), but also the absence of bacteria can induce these
eects. Animals raised in germ-free environments showed an exag-
gerated HPA-axis response, which was normalized by colonization
with fecal matter from specically germ-free animals or by the
administration of the Gram-positive Bidobacter ium infantis (94).
Viceversa, exposure to social stress changed the composition of
the gut-microbiota in mice (95, 96) and prenatal stress altered the
May 2015 | Volume 6 | Article 2236
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
microbiome in rhesus monkeys by reducing the overall numbers of
the Gram-positive Bidobacteria and Lactobacilli (97), indicating
that chronic stress aected the composition of the gut microbiome.
Stress inuences gut motility, secretions, and mucin production,
thereby altering the habitat of resident bacteria, promoting changes
in the composition of the gut microbiome (98), and allowing the
growth of pathogenic bacteria (99).
Increasing evidence supports an important role for microbiota
on the homeostasis of the intestinal barrier. Certain strains of the
Gram-positive Lactobacilli decreased intestinal permeability in
several animal and human disease models (78, 100). B. infantis
reduced intestinal permeability (as assessed by 70-kDa uorescein
isothiocyanate–dextran transmucosal ux) and ameliorated symp-
toms in a neonatal necrotizing enterocolitis mouse model (101).
Further evidence indicating the inuence of the gut-microbiota
on intestinal permeability was presented in detoxifying alcoholic-
dependent subjects: lower levels of Ruminococcaceae and higher
abundance of Lachnospiraceae (Dorea) and Blautia were associ-
ated with increased intestinal permeability (102). In addition,
higher levels of certain pathogenic bacteria can increase intestinal
permeability by disrupting the epithelial barrier and triggering
cell death and inammation. ese bacteria have the ability to
bind and/or translocate through endothelial and microfold cells
and have been shown to secrete toxins or other eector molecules
via specialized secretion systems. Although the exact mechanisms
are not well described, most pathogenic gut bacteria including
Escherichi a coli, Helicobac ter pylori, Staphylococcus aureus, Cholera
Pseudomonas uorescens, Pseudomonas aeruginosa, Yersinia
enterocolitica, Campylobacter jejuni, and Salmonella ty phimurium
alter paracellular permeability by disassembling tight junctions
and generating cytoskeleton changes by increasing inammation
[reviewed by Barreau etal. (103)]. As an example, a strain of E.
coli, normally present in the human gut, induced focal leaks in
colonic epithelial monolayers and in rat distal colon by using
α-hemolysin, allowing for its paracellular translocation across
the epithelial layer (104).
High-Caloric and High-Fat Diets Induce Inammation
and Increase Circulating Endotoxin Levels
Compared to healthy individuals, patients suering from obesity
have higher circulating endotoxin levels together with greater levels
of circulating pro-inammatory cytokines and insulin resistance
(105). Food intake can produce post-prandial immune activation
and elevate endotoxin levels when a meal is high in calories (106)
or has a high fat content (6, 107109).
Rodents fed a 4-week high-fat diet (72% fat) showed a con-
stant elevation in circulating endotoxin levels, while in control
animals, endotoxin levels only increased during feeding hours.
Furthermore, a high-fat diet produced fasting glycemia, insulin
resistance, general weight gain, and weight gain of the liver and
visceral and subcutaneous adipose tissue. In addition, adipose
tissue F4/80-positive cells (indicating the inltration of mac-
rophages), markers of inammation, and liver triglyceride content
were increased. Interestingly, almost similar eects were observed
in mice subcutaneously infused with LPS (resulting in similar
circulating LPS levels as observed in the high-fat fed mice). ese
eects were mediated by TLR4, since mice lacking CD14, which
is important for the recognition of LPS to this receptor, showed a
delayed response to a high-fat diet or LPS injections (107).
In healthy humans, a 910 calories high-fat and high-carbohy-
drate meal resulted in increased circulating endotoxin levels and
elevated levels of LBP in parallel with higher inammatory markers
and increased protein expression of TLR2 and TLR4 in isolated
leukocytes. A meal high in fruits and ber did not induce these
eects (108). Plasma endotoxin levels, pro-inammatory markers,
and leukocyte TLR4 expression increased aer the intake of cream
(300 calories), while the intake of 300 calories of glucose resulted
only in a pro-inammatory response and the intake of orange juice
and water showed none of these eects (110). In healthy individu-
als, plasma endotoxin levels increased about 50% aer the intake
of a high-fat meal (900 calories) (6) and 4weeks consumption of
a Western-style diet raised plasma endotoxin activity levels by
71% (111).
How exactly the intake of a high-caloric meal increases cir-
culating endotoxin levels is still unclear but has been explained
by several mechanisms [reviewed by Kelly etal. (112)]. One of
these suggested mechanisms is that the introduction of a high-fat
diet modulates the expression of genes involved in the barrier
function in epithelial cells, thereby directly compromising
the integrity of the tight junction (113). Another explanation
could be that a high-caloric/high-fat meal induces high levels
of insulin and leptin, hormones that directly activate the SNS
(114, 115). Moreover, insulin enhances SGLT1-mediated glucose
absorption (116). Activation of the SGLT1 and the SNS leads
to increased permeability of the gut barrier, which may induce
the observed post-prandial endotoxemia (our hypothesis and
theory).
Gliadin Compromises the Integrity of Tight
Junctions
e intake of wheat and other cereal grains has been implicated
in the development of inammation-related diseases, by inducing
inammation and increasing intestinal permeability (40). Gliadin,
a component of gluten, has been demonstrated to increase perme-
ability in human Caco-2 intestinal epithelial cells by reorganizing
actin laments and altering expression of junctional complex
proteins (117). Several studies by the group of Fasano eta l. showed
that the binding of gliadin to the chemokine receptor CXCR3
on epithelial IEC-6 and Caco-2 cells releases zonulin, a protein
that directly compromises the integrity of the junctional complex
(118, 119).
Alcohol Consumption Increases Intestinal
Permeability
Alcohol consumption is an important risk factor for disease and
is one of the major causes of chronic liver disease. Increased
intestinal permeability has been observed during chronic alcohol
consumption. In an animal model of chronic alcoholic liver
disease, alcohol feeding for 8weeks increased intestinal perme-
ability (120). In humans, alcohol-dependence induced changes
in the gut-microbiota composition that were associated with
increased intestinal permeability (102). Furthermore, increased
intestinal permeability and higher circulating endotoxin levels
were observed in patients with chronic alcohol abuse (121123).
May 2015 | Volume 6 | Article 2237
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
Exercise-Induced Heat-Stress Increases Intestinal
Permeability
Exercise increases body temperature, reduces intestinal blood ow
(reallocated to the muscles and cardiac system), and increases
intestinal permeability by activating the SNS and HPA-axis.
Already in 1992, Oktedalen etal. (124) showed that marathon
runners displayed a signicant increase in intestinal permeability.
In addition, studies have indicated that strenuous exercise induced
higher circulating endotoxin levels and activated the immune
system (125128). Further evidence of exercise- and heat-induced
increased intestinal permeability, leading to gastrointestinal com-
plaints in people engaging in physical activity, has been recently
reviewed (129).
Endotoxemia is Associated with Diseases
Related to Chronic Inammation
Multiple human studies have emerged that nd associations with
NCDs and markers of endotoxemia. Even aging, associated with
higher sympathetic nerve activity (130) and higher circulating
inammatory mediators like IL-6, has been linked to higher
plasma concentrations of LPS and LBP (131). In further support
of our theory, in this section, an overview is given of human stud-
ies nding changes in levels of endotoxin or endotoxin-related
markers in NCDs (Tab l e 1 ).
Metabolic Syndrome
Metabolic syndrome is accompanied by an increased risk for
NAFLD, obesity, type 2 diabetes, and cardiovascular diseases.
All of these conditions are related to and even predicted by
increased sympathetic nerve activity (154) and a dysregulated
HPA-axis (155). Higher circulating endotoxin and LBP levels
are associated with risk factors of the metabolic syndrome, like
insulin resistance, obesity, dyslipidemia, and chronic inflam-
mation (132135). Patients suffering from NAFLD exhibited
significantly higher serum endotoxin levels in contrast to
healthy controls (14). Farhardi et al. (136) indicated that
elevated plasma endotoxin levels in these patients were related
to an impaired intestinal barrier function, because, only in the
patient group, the intake of a permeability stressor (aspirin)
increased the 0–24h urinary excretion of sucralose (a marker
of whole-gut permeability). Furthermore, augmented plasma
LBP levels in concert with increased plasma levels of TNF-α
were observed in obese NAFLD patients compared to healthy
controls (137).
Elevated circulating levels of endotoxin and LBP were
detected in type 2 diabetics (12, 133, 138140). Compared
to healthy controls, obese individuals and type 2 diabetics
showed higher endotoxin levels after the intake of a high-fat
meal. Increased endotoxin levels were observed in all chal-
lenged individuals, yet higher endotoxin levels were seen in
individuals suffering from metabolic illnesses, suggesting an
increased intestinal permeability in these patients (141). This
was further indicated by a recent study showing that increased
serum levels of endotoxin, IL-6, and TNF-α were found in type
2 diabetic patients compared to healthy individuals. The level
of endotoxin was positively related to zonulin, a marker for
intestinal permeability (12).
A large cohort of patients with coronary artery disease identi-
ed increased serum LBP levels to be associated with total and
cardiovascular mortality (144). Moreover, circulating LBP levels
were associated with carotid intima media thickness (a marker
of atherosclerosis), obesity, insulin resistance, and high-sensitive
CRP (145).
Patients suering from chronic heart failure with aggravated
renal function displayed increased circulating endotoxin levels
and an impairment of the intestinal barrier (11). Wiedermann
etal. (146) showed that subjects with the highest levels of circu-
lating endotoxin (90th percentile) had a threefold increased risk
of incident atherosclerosis. Higher serum endotoxin and pro-
inammatory cytokine concentrations were seen in patients with
edematous chronic heart disease compared to stable patients
and healthy controls. Intriguingly, aer short-term diuretic
treatment, circulating endotoxin concentrations decreased in
edematous patients (147). Diuretic treatment [like angiotensin-
converting enzyme (ACE) inhibitors] ameliorated intestinal
inammation, perhaps by impacting on intestinal permeability
through interference with the renin–angiotensin–aldosterone
system. Several components of this system (renin, ACE, and
angiotensin II) have been shown to stimulate pro-inammatory
pathways (44, 156).
Inammatory Bowel Disease
Ulcerative colitis and Crohn’s disease are intestinal inammatory
disorders, also known as IBD, which have been causally linked
to chronic psychological stress (157), altered immune function,
changes in the gut-microbiota, increased intestinal permeability,
and endotoxemia (158). For example, increased plasma endotoxin
and LBP levels were measured in both patient groups, but were
more pronounced in patients with active disease compared to
inactive disease and were associated with disease severity (148).
In addition, detectable plasma endotoxin levels and higher plasma
levels of LBP were more frequently observed in IBD patients
compared to controls (15, 149) and were correlated with disease
severity and circulating TNF-α levels (150).
Psychiatric Diseases
Over the last decade, the role of the gut–brain axis has emerged as
an important mediator in the development of psychiatric and mood
disorders (159). Moreover, higher endotoxin levels and intestinal
barrier dysfunction are observed in several of these conditions. For
example, Parkinson’s patients exhibited increased total intestinal
permeability and a more intense staining for E. coli LPS and oxida-
tive stress markers in intestinal sigmoid mucosa samples. However,
in these patients, endotoxin levels resembled control samples
and serum LBP concentrations were lower compared to healthy
individuals (151). Higher serum endotoxin levels are associated
with severe autism, sporadic amyotrophic lateral sclerosis, and
Alzheimer’s dise ase (13, 152). Furthermore, increased IgA and IgM
responses against LPS of commensal bacteria were seen in chronic
fatigue syndrome (10) and depression (9). Intriguingly, chronic
oral infection of periodontitis was associated with Alzheimer’s
May 2015 | Volume 6 | Article 2238
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
disease where higher antibody levels against oral pathogens were
observed years before the onset of symptoms in people suering
from Alzheimer’s disease (153), suggesting there was an increased
translocation of bacteria and/or bacterial toxins from the mouth
into the bloodstream.
Conclusion
Chronic low-grade inammation is an eminent feature of NCDs.
In addition, many studies report increased circulating endotoxin
levels and increased gut permeability in patients suering from
these conditions. As reviewed in this paper, stress-induced
increases in intestinal permeability, in combination with modern
life-style factors, raise the possibility of translocation of bacteria
and/or their toxins across the more permeable gut barrier. e
resulting, long lasting, endotoxemia should be considered much
more than just a risk factor for chronic disease; it could be a cause.
Notwithstanding the fact that the exact origin and sequence of
events involved in development of NCDs remain to be unsolved,
evidence indicates that a disrupted barrier function in parallel
with elevated circulating endotoxin levels may underlie disease
onset and progression. For this reason, therapies aimed at restoring
intestinal barrier function, life-style changes, and stress manage-
ment should be considered important strategies in preventing and
attenuating the pro-inammatory state observed in NCDs.
Acknowledgments
We would like to thank Alicia Lammerts van Bueren for the edito-
rial work on the manuscript.
TABLE 1 | Associations found between markers of endotoxemia and disease.
Reference Disease Marker(s) of endotoxemia Effect
(132) Metabolic syndrome Serum LPS LPS levels correlated positively with
symptoms of metabolic syndrome
(133) Obesity-related insulin resistance Serum LBP LBP levels increased
(134) Psoriasis/metabolic syndrome Serum LBP LBP levels only increased in psoriasis
patients with metabolic syndrome
(135) Obesity Plasma LBP LBP levels increased
(136) NAFLD Plasma LPS LPS levels increased
(14) NAFLD Serum LPS LPS levels increased
(137) Obesity/NAFLD Plasma LBP LBP levels increased
(122) Liver disease Plasma LPS LPS levels increased
(12) Type 2 diabetes Serum LPS LPS levels increased
(138) Type 2 diabetes Plasma LPS LPS levels increased
(139) Type 2 diabetes Serum LPS LPS levels increased
(140) Diabetes Serum LPS LPS levels increased
(141) Type 2 diabetes, impaired glucose
tolerance
Serum LPS LPS levels increased
(142) Cardiovascular diseases Serum LPS, serum IgA/IgG against
oral bacteria
LPS levels increased, no differences
in IgA/IgG levels
(143) Coronary artery disease Plasma LBP LBP levels increased
(144) Coronary artery disease Serum LBP LBP levels increased
(145) Arteriosclerosis Serum LBP LBP levels increased
(146) Arteriosclerosis Plasma LBP LBP levels increased
(11) Chronic heart failure (edematous) Plasma LPS LPS levels increased in edematous
vs. non-edematous patients. No
differences between all patients vs.
controls
(147) Chronic heart disease (edematous) Plasma LPS LPS levels increased in edematous
vs. non-edematous patients
(148) IBD Serum LPS, LBP, sCD14 LPS, LBP, sCD14 levels increased
(149) IBD Plasma LPS, LBP, sCD15, endoCAbs No differences in levels of LPS,
sCD14, and endoCAbs. LBP levels
increased
(150) IBD Plasma LPS, endoCAbs LPS and endoCAbs levels increased
with disease severity
(151) Parkinson’s disease Serum LBP, E. coli LPS inltration in
intestinal tissue
LBP levels decreased, increased LPS
inltration in intestinal tissue
(13) Autism Serum LPS, sCD14 LPS levels increased, no differences
in sCD14 levels
(152) Sporadic amyotrophic lateral sclerosis,
Alzheimer’s disease
Plasma LPS LPS levels increased
(9) Depression Serum IgA/IgM against intestinal bacteria IgA/IgM levels increased
(10) Chronic fatigue syndrome Serum IgA/IgM against intestinal bacteria IgA/IgM levels increased
(153) Alzheimer’s disease Serum IgG against oral bacteria IgG levels increased
May 2015 | Volume 6 | Article 2239
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
References
1. Andreasen AS, Krabbe KS, Krogh-Madsen R, Taudorf S, Pedersen BK, Moller
K. Human endotoxemia as a model of systemic inammation. Curr Med Chem
(2008) 15(17):1697–705. doi:10.2174/092986708784872393
2. Bosma-den Boer MM, van Wetten ML, Pruimboom L. Chronic inamma-
tory diseases are stimulated by current lifestyle: how diet, stress levels and
medication prevent our body from recovering. Nutr Metab (2012) 9(1):32.
doi:10.1186/1743-7075-9-32
3. Eckel RH, Alberti KG, Grundy SM, Zimmet PZ. e metabolic syndrome. Lancet
(2010) 375(9710):181–3. doi:10.1016/S0140-6736(09)61794-3
4. Raison CL, Capuron L, Miller AH. Cytokines sing the blues: inammation and the
pathogenesis of depression. Tren ds Immunol (2006) 27(1):24–31. doi:10.1016/j.
it.2005.11.006
5. Ruiz-Nunez B, Pruimboom L, Dijck-Brouwer DA, Muskiet FA. Lifestyle and
nutritional imbalances associated with Western diseases: causes and conse-
quences of chronic systemic low-grade inammation in an evolutionary context.
J Nutr Biochem (2013) 24(7):1183–201. doi:10.1016/j.jnutbio.2013.02.009
6. Erridge C, Attina T, Spickett CM, Webb DJ. A high-fat meal induces low-grade
endotoxemia: evidence of a novel mechanism of postprandial inammation.
Am J Clin Nutr (2007) 86(5):1286–92.
7. Moreira AP, Texeira TF, Ferreira AB, Peluzio Mdo C, Alfenas Rde C. Inuence
of a high-fat diet on gut microbiota, intestinal permeability and metabolic
endotoxemia. Br J Nutr (2012) 108(5):801–9. doi:10.1017/S0007114512001213
8. Vaishnavi C. Translocation of gut ora and its role in sepsis. Indian J Med
Microbiol (2013) 31(4):334–42. doi:10.4103/0255-0857.118870
9. Maes M, Mihaylova I, Leunis JC. Increased serum IgM antibodies directed
against phosphatidyl inositol (Pi) in chronic fatigue syndrome (CFS) and major
depression: evidence that an IgM-mediated immune response against Pi is one
factor underpinning the comorbidity between both CFS and depression. Neuro
Endocrinol Lett (2007) 28(6):861–7.
10. Maes M, Mihaylova I, Leunis JC. Increased serum IgA and IgM against LPS of
enterobacteria in chronic fatigue syndrome (CFS): indication for the involvement
of Gram-negative enterobacteria in the etiology of CFS and for the presence of
an increased gut-intestinal permeability. J Aect Disord (2007) 99(1–3):237–40.
doi:10.1016/j.jad.2006.08.021
11. Sandek A, Bjarnason I, Volk HD, Crane R, Meddings JB, Niebauer J, et al. Studies
on bacterial endotoxin and intestinal absorption function in patients with chronic
heart failure. Int J Cardiol (2012) 157(1):80–5. doi:10.1016/j.ijcard.2010.12.016
12. Jayashree B, Bibin YS, Prabhu D, Shanthirani CS, Gokulakrishnan K, Lakshmi
BS, et al. Increased circulatory levels of lipopolysaccharide (LPS) and zonulin
signify novel biomarkers of proinammation in patients with type 2 diabetes.
Mol Cell Biochem (2014) 388(1–2):203–10. doi:10.1007/s11010-013-1911-4
13. Emanuele E, Orsi P, Boso M, Broglia D, Brondino N, Barale F, et al. Low-grade
endotoxemia in patients with severe autism. Neurosci Lett (2010) 471(3):162–5.
doi:10.1016/j.neulet.2010.01.033
14. Harte AL, da Silva NF, Creely SJ, McGee KC, Billyard T, Youssef-Elabd EM, et al.
Elevated endotoxin levels in non-alcoholic fatty liver disease. J Ina mm (2010)
7:15. doi:10.1186/1476-9255-7-15
15. Caradonna L, Amati L, Lella P, Jirillo E, Caccavo D. Phagocytosis, killing,
lymphocyte-mediated antibacterial activity, serum autoantibodies, and
plasma endotoxins in inammatory bowel disease. Am J Gastroenterol (2000)
95(6):1495–502. doi:10.1111/j.1572-0241.2000.02085.x
16. Wang JE, Dahle MK, McDonald M, Foster SJ, Aasen AO, iemermann C.
Peptidoglycan and lipoteichoic acid in gram-positive bacterial sepsis: receptors,
signal transduction, biological eects, and synergism. Shock (2003) 20(5):402–14.
doi:10.1097/01.shk.0000092268.01859.0d
1 7. Giannelli V, Di Gregorio V, Iebba V, Giusto M, Schippa S, Merli M, et al. Microbiota
and the gut-liver axis: bacterial translocation, inammation and infection in
cirrhosis. World J Gastroenterol (2014) 20(45):16795–810. doi:10.3748/wjg.v20.
i45.16795
18. Bohannon JK, Hernandez A, Enkhbaatar P, Adams WL, Sherwood ER. e immu-
nobiology of toll-like receptor 4 agonists: from endotoxin tolerance to immuno-
adjuvants. Shock (2013) 40(6):451–62. doi:10.1097/SHK.0000000000000042
19. Neves AL, Coelho J, Couto L, Leite-Moreira A, Roncon-Albuquerque R Jr.
Metabolic endotoxemia: a molecular link between obesity and cardiovascular
risk. J Mol Endocrinol (2013) 51(2):R51–64. doi:10.1530/JME-13-0079
20. Weinlich R, Bortoluci KR, Chehab CF, Serezani CH, Ulbrich AG, Peters-
Golden M, et al. TLR4/MYD88-dependent, LPS-induced synthesis of PGE2
by macrophages or dendritic cells prevents anti-CD3-mediated CD95L
upregulation in T cells. Cell Death Dier (2008) 15(12):1901–9. doi:10.1038/
cdd.2008.128
21. Maitra U, Gan L, Chang S, Li L. Low-dose endotoxin induces inammation by
selectively removing nuclear receptors and activating CCAAT/enhancer-binding
protein delta. J Immunol (2011) 186(7):4467–73. doi:10.4049/jimmunol.1003300
22. Cavaillon JM, Adib-Conquy M. Bench-to-bedside review: endotoxin tolerance
as a model of leukocyte reprogramming in sepsis. Crit Care (2006) 10(5):233.
doi:10.1186/cc5055
23. Deng H, Maitra U, Morris M, Li L. Molecular mechanism responsible for
the priming of macrophage activation. J Biol Chem (2013) 288(6):3897–906.
doi:10.1074/jbc.M112.424390
24. Kapus A, Szaszi K. Coupling between apical and paracellular transport processes.
Biochem Cell Biol (2006) 84(6):870–80. doi:10.1139/o06-202
25. Turner JR, Rill BK, Carlson SL, Carnes D, Kerner R, Mrsny RJ, et al. Physiological
regulation of epithelial tight junctions is associated with myosin light-chain
phosphorylation. Am J Physiol (1997) 273(4 Pt 1):C1378–85.
26. Lee CY. Chronic restraint stress induces intestinal inammation and alters the
expression of hexose and lipid transporters. Clin Exp Pharmacol Physiol (2013)
40(6):385–91. doi:10.1111/1440-1681.12096
27. Turner JR. Show me the pathway! Regulation of paracellular permeability
by Na(+)-glucose cotransport. Adv Drug Deliv Rev (2000) 41(3):265–81.
doi:10.1016/S0169-409X(00)00046-6
28. Turner JR. Intestinal mucosal barrier function in health and disease. Nat Rev
Immunol (2009) 9(11):799–809. doi:10.1038/nri2653
29. S chneeberger EE, Lynch RD. e tight junction: a multifunctional complex. Am
J Physiol Cell Physiol (2004) 286(6):C1213–28. doi:10.1152/ajpcell.00558.2003
30. Anderson JM, Van Itallie CM. Physiology and function of the tight junction.
Cold Spring Harb Perspect Biol (2009) 1(2):a002584. doi:10.1101/cshperspect.
a002584
31. Overgaard CE, Daugherty BL, Mitchell LA, Koval M. Claudins: control of barrier
function and regulation in response to oxidant stress. Antioxid Redox Signal
(2011) 15(5):1179–93. doi:10.1089/ars.2011.3893
32. Graham WV, Wang F, Clayburgh DR, Cheng JX, Yoon B, Wang Y, et al. Tumor
necrosis factor-induced long myosin light chain kinase transcription is regulated
by dierentiation-dependent signaling events. Characterization of the human
long myosin light chain kinase promoter. J Biol Chem (2006) 281(36):26205–15.
doi:10.1074/jbc.M602164200
33. Shen L, Black ED, Witkowski ED, Lencer WI, Guerriero V, Schneeberger EE, et
al. Myosin light chain phosphorylation regulates barrier function by remodeling
tight junction structure. J Cell Sci (2006) 119(Pt 10):2095–106. doi:10.1242/
jcs.02915
34. Cunningham KE, Turner JR. Myosin light chain kinase: pulling the strings
of epithelial tight junction function. Ann N Y Acad Sci (2012) 1258:34–42.
doi:10.1111/j.1749-6632.2012.06526.x
35. Secondulfo M, Iafusco D, Carratu R, deMagistris L, Sapone A, Generoso M,
et al. Ultrastructural mucosal alterations and increased intestinal permeability
in non-celiac, type I diabetic patients. Dig Liver Dis (2004) 36(1):35–45.
doi:10.1016/j.dld.2003.09.016
36. Fasano A. Leaky gut and autoimmune diseases. Clin Rev Allergy Immunol (2012)
42(1):71–8. doi:10.1007/s12016-011-8291-x
37. Keita AV, Soderholm JD. The intestinal barrier and its regulation by
neuroimmune factors. Neurogastroenterol Motil (2010) 22(7):718–33.
doi:10.1111/j.1365-2982.2010.01498.x
38. Hijazi Z, Molla AM, Al-Habashi H, Muawad WM, Molla AM, Sharma PN.
Intestinal permeability is increased in bronchial asthma. Arch Dis Child (2004)
89(3):227–9. doi:10.1136/adc.2003.027680
39. Maes M, Kubera M, Leunis JC, Berk M, Geard M, Bosmans E. In depres-
sion, bacterial translocation may drive inflammatory responses, oxidative
and nitrosative stress (O&NS), and autoimmune responses directed against
O&NS-damaged neoepitopes. Acta Psychiatr Scand (2013) 127(5):344–54.
doi:10.1111/j.1600-0447.2012.01908.x
40. de Punder K, Pruimboom L. e dietary intake of wheat and other cereal grains
and their role in inammation. Nutrients (2013) 5(3):771–87. doi:10.3390/
nu5030771
May 2015 | Volume 6 | Article 22310
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
41. Straub RH. Evolutionary medicine and chronic inammatory state – known and
new concepts in pathophysiology. J Mol Med (2012) 90(5):523–34. doi:10.1007/
s00109-012-0861-8
42. Straub RH. TRPV1, TRPA1, and TRPM8 channels in inammation, energy
redirection, and water retention: role in chronic inammatory diseases with
an evolutionary perspective. J Mol Med (2014) 92(9):925–37. doi:10.1007/
s00109-014-1175-9
43. Triposkiadis F, Karayannis G, Giamouzis G, Skoularigis J, Louridas G, Butler J.
e sympathetic nervous system in heart failure physiology, pathophysiology,
and clinical implications. J Am Coll Cardiol (2009) 54(19):1747–62. doi:10.1016/j.
jacc.2009.05.015
44. Groeschel M, Braam B. Connecting chronic and recurrent stress to vascular
dysfunction: no relaxed role for the renin-angiotensin system. Am J Physiol
Renal Physiol (2011) 300(1):F1–10. doi:10.1152/ajprenal.00208.2010
45. Brunsson I, Eklund S, Jodal M, Lundgren O, Sjovall H. e eect of vasodilatation
and sympathetic nerve activation on net water absorption in the cat’s small intestine.
Acta Physiol Scand (1979) 106(1):61–8. doi:10.1111/j.1748-1716.1979.tb06370.x
46. Levens NR, Peach MJ, Carey RM. Interactions between angiotensin peptides
and the sympathetic nervous system mediating intestinal sodium and water
absorption in the rat. J Clin Invest (1981) 67(4):1197–207. doi:10.1172/JCI110135
47. Lange S, Delbro DS. Adrenoceptor-mediated modulation of Evans blue dye
permeation of rat small intestine. Dig Dis Sci (1995) 40(12):2623–9. doi:10.1007/
BF02220451
48. Aschenbach JR, Borau T, Gabel G. Glucose uptake via SGLT-1 is stimulated
by beta(2)-adrenoceptors in the ruminal epithelium of sheep. J Nutr (2002)
132(6):1254–7.
49. Ishikawa Y, Eguchi T, Ishida H. Mechanism of beta-adrenergic agonist-in-
duced transmural transport of glucose in rat small intestine. Regulation of
phosphorylation of SGLT1 controls the function. Biochim Biophys Acta (1997)
1357(3):306–18. doi:10.1016/S0167-4889(97)00043-8
50. Schaper J, Wagner A, Enigk F, Brell B, Mousa SA, Habazettl H, et al. Regional
sympathetic blockade attenuates activation of intestinal macrophages and
reduces gut barrier failure. Anesthesiology (2013) 118(1):134–42. doi:10.1097/
ALN.0b013e3182784c93
51. de K loet ER, Joels M, Holsboer F. Stress and the brain: from adaptation to disease.
Nat Rev Neurosci (2005) 6(6):463–75. doi:10.1038/nrn1683
52. Hattangady NG, Olala LO, Bollag WB, Rainey WE. Acute and chronic regu-
lation of aldosterone production. Mol Cell Endocrinol (2012) 350(2):151–62.
doi:10.1016/j.mce.2011.07.034
53. Yu Y, L iu ZQ, Liu XY, Yang L, Geng XR, Yang G, et al. Stress-derived corticotro-
pin releasing factor breaches epithelial endotoxin tolerance. PLoS One (2013)
8(6):e65760. doi:10.1371/journal.pone.0065760
54. Suzuki T, Yoshinaga N, Tanabe S. Interleukin-6 (IL-6) regulates claudin-2
expression and tight junction permeability in intestinal epithelium. J Biol Chem
(2011) 286(36):31263–71. doi:10.1074/jbc.M111.238147
55. Meddings JB, Swain MG. Environmental stress-induced gastrointestinal perme-
ability is mediated by endogenous glucocorticoids in the rat. Gastroenterology
(2000) 119(4):1019–28. doi:10.1053/gast.2000.18152
56. Caso JR, Leza JC, Menchen L. e eects of physical and psychological stress
on the gastro-intestinal tract: lessons from animal models. Curr Mol Med (2008)
8(4):299–312. doi:10.2174/156652408784533751
57. Vanuytsel T, van Wanrooy S, Vanheel H, Vanormelingen C, Verschueren S,
Houben E, et al. Psychological stress and corticotropin-releasing hormone
increase intestinal permeability in humans by a mast cell-dependent mechanism.
Gut (2013) 63(8):1293–9. doi:10.1136/gutjnl-2013-305690
58. Capaldo CT, Nusrat A. Cytokine regulation of tight junctions. Biochim Biophys
Acta (2009) 1788(4):864–71. doi:10.1016/j.bbamem.2008.08.027
59. Watson CJ, Hoare CJ, Garrod DR, Carlson GL, Warhurst G. Interferon-gamma
selectively increases epithelial permeability to large molecules by activating
dierent populations of paracellular pores. J Cell Sci (2005) 118(Pt 22):5221–30.
doi:10.1242/jcs.02630
60. Wersching H, Duning T, Lohmann H, Mohammadi S, Stehling C, Fobker M,
et al. Serum C-reactive protein is linked to cerebral microstructural integrity
and cognitive function. Neurology (2010) 74(13):1022–9. doi:10.1212/
WNL.0b013e3181d7b45b
61. Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inamma-
tion to sickness and depression: when the immune system subjugates the brain.
Nat Rev Neurosci (2008) 9(1):46–56. doi:10.1038/nrn2297
62. Lang CH, Molina PE, Yousef KA, Tepper PG, Abumrad NN. Role of IL-1 alpha
in central nervous system immunomodulation of glucoregulation. Brain Res
(1993) 624(1–2):53–60. doi:10.1016/0006-8993(93)90059-V
63. Dunn AJ. Cytokine activation of the HPA axis. Ann N Y Acad Sci (2000)
917:608–17. doi:10.1111/j.1749-6632.2000.tb05426.x
64. Goebel MU, Baase J, Pithan V, Exton M, Saller B, Schedlowski M, et al.
Acute interferon beta-1b administration alters hypothalamic-pituitary-ad-
renal axis activity, plasma cytokines and leukocyte distribution in healthy
subjects. Psychoneuroendocrinology (2002) 27(8):881–92. doi:10.1016/
S0306-4530(01)00099-3
65. Zimomra ZR, Portereld VM, Camp RM, Johnson JD. Time-dependent medi-
ators of HPA axis act ivation following live Escherichia coli. Am J Physiol Regul
Integr Comp Physiol (2011) 301(6):R1648–57. doi:10.1152/ajpregu.00301.2011
66. Mohn CE, Fernandez-Solari J, De Laurentiis A, Prestilippo JP, de la Cal C, Funk
R, et al. e rapid release of corticosterone from the adrenal induced by ACTH
is mediated by nitric oxide acting by prostaglandin E2. Proc Natl Acad Sci U S
A (2005) 102(17):6213–8. doi:10.1073/pnas.0502136102
67. Vakharia K, Hinson JP. Lipopolysaccharide directly stimulates cortisol
secretion by human adrenal cells by a cyclooxygenase-dependent mechanism.
Endocrinology (2005) 146(3):1398–402. doi:10.1210/en.2004-0882
68. Zacharowski K, Zacharowski PA, Koch A, Baban A, Tran N, Berkels R, et al.
Toll-like receptor 4 plays a crucial role in the immune-adrenal response to
systemic inammatory response syndrome. Proc Natl Acad Sci U S A (2006)
103(16):6392–7. doi:10.1073/pnas.0601527103
69. Serrats J, Schiltz JC, Garcia-Bueno B, van Rooijen N, Reyes TM, Sawchenko PE.
Dual roles for perivascular macrophages in immune-to-brain signaling. Neuron
(2010) 65(1):94–106. doi:10.1016/j.neuron.2009.11.032
70. Dhabhar FS, Malarkey WB, Neri E, McEwen BS. Stress-induced redistribution
of immune cells – from barracks to boulevards to battleelds: a tale of three
hormones – Curt Richter award winner. Psychoneuroendocrinology (2012)
37(9):1345–68. doi:10.1016/j.psyneuen.2012.05.008
71. Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES. e sympathetic nerve – an
integrative interface between two supersystems: the brain and the immune
system. Pharmacol Rev (2000) 52(4):595–638.
72. Kohm AP, Sanders VM. Norepinephrine and beta 2-adrenergic receptor
stimulation regulate CD4+ T and B lymphocyte function invitro and invivo.
Pharmacol Rev (2001) 53(4):487–525.
73. Prather AA, Carroll JE, Fury JM, McDade KK, Ross D, Marsland AL. Gender
dierences in stimulated cytokine production following acute psychological
stress. Brain Behav Immun (2009) 23(5):622–8. doi:10.1016/j.bbi.2008.11.004
74. Steptoe A, Hamer M, Chida Y. e eects of acute psychological stress on
circulating inammatory factors in humans: a review and meta-analysis. Brain
Behav Immun (2007) 21(7):901–12. doi:10.1016/j.bbi.2007.03.011
75. Yamakawa K, Matsunaga M, Isowa T, Kimura K, Kasugai K, Yoneda M, et al.
Transient responses of inammatory cytokines in acute stress. Biol Psychol (2009)
82(1):25–32. doi:10.1016/j.biopsycho.2009.05.001
76. Bierhaus A, Wolf J, Andrassy M, Rohleder N, Humpert PM, Petrov D, et
al. A mechanism converting psychosocial stress into mononuclear cell
activation. Proc Natl Acad Sci U S A (2003) 100(4):1920–5. doi:10.1073/
pnas.0438019100
77. Barnes PJ, Adcock IM. Glucocorticoid resistance in inammatory diseases.
Lancet (2009) 373(9678):1905–17. doi:10.1016/S0140-6736(09)60326-3
78. Ait-Belgnaoui A, Durand H, Cartier C, Chaumaz G, Eutamene H, Ferrier L,
et al. Prevention of gut leakiness by a probiotic treatment leads to attenuated
HPA response to an acute psychological stress in rats. Psychoneuroendocrinology
(2012) 37(11):1885–95. doi:10.1016/j.psyneuen.2012.03.024
79. Glaser R, Kiecolt-Glaser JK. Stress-induced immune dysfunction: implications
for health. Nat Rev Immunol (2005) 5(3):243–51. doi:10.1038/nri1571
80. Huang CJ, Acevedo EO, Mari DC, Randazzo C, Shibata Y. Glucocorticoid
inhibition of leptin- and lipopolysaccharide-induced interlukin-6 production
in obesity. Brain Behav Immun (2013). doi:10.1016/j.bbi.2013.10.004
81. Maranville JC, Micic D, Hanauer SB, Di Rienzo A, Kupfer SS. Invitro sensitivity
assays and clinical response to glucocorticoids in patients with inammatory bowel
disease. J Crohns Colitis (2014) 8(11):1539–47. doi:10.1016/j.crohns.2014.06.013
82. Pariante CM. Glucocorticoid receptor function invitro in patients with major
depression. Stress (2004) 7(4):209–19. doi:10.1080/10253890500069650
83. Sauer J, Polack E, Wikinski S, Holsboer F, Stalla GK, Arzt E. e glucocor-
ticoid sensitivity of lymphocytes changes according to the activity of the
May 2015 | Volume 6 | Article 22311
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
hypothalamic-pituitary-adrenocortical system. Psychoneuroendocrinology (1995)
20(3):269–80. doi:10.1016/0306-4530(94)00058-I
84. Glaser R. Stress-associated immune dysregulation and its importance for human
health: a personal history of psychoneuroimmunology. Brain Behav Immun
(2005) 19(1):3–11. doi:10.1016/j.bbi.2004.06.003
85. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, et
al. Impact of diet in shaping gut microbiota revealed by a comparative study
in children from Europe and rural Africa. Proc Natl Acad Sci U S A (2010)
107(33):14691–6. doi:10.1073/pnas.1005963107
86. Schnorr SL, Candela M, Rampelli S, Centanni M, Consolandi C, Basaglia G, et
al. Gut microbiome of the Hadza hunter-gatherers. Nat Commun (2014) 5:3654.
doi:10.1038/ncomms4654
87. Brown K, DeCoe D, Molcan E, Gibson DL. Diet-induced dysbiosis of the
intestinal microbiota and the eects on immunity and disease. Nutrients (2012)
4(8):1095–119. doi:10.3390/nu4081095
88. van Baarlen P, Troost FJ, van Hemert S, van der Meer C, de Vos WM, de Groot
PJ, et al. Dierential NF-kappaB pathways induction by Lactobacillus plantarum
in the duodenum of healthy humans correlating with immune tolerance. Proc
Natl Acad Sci U S A (2009) 106(7):2371–6. doi:10.1073/pnas.0809919106
89. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, et al. Gut microbiota
from twins discordant for obesity modulate metabolism in mice. Science (2013)
341(6150):1241214. doi:10.1126/science.1241214
90. Dinan TG, Cryan JF. Regulation of the stress response by the gut microbiota:
implications for psychoneuroendocrinology. Psychoneuroendocrinology (2012)
37(9):1369–78. doi:10.1016/j.psyneuen.2012.03.007
91. Karrow NA. Activation of the hypothalamic-pituitary-adrenal axis and auto-
nomic nervous system during inammation and altered programming of the
neuroendocrine-immune axis during fetal and neonatal development: lessons
learned from the model inammagen, lipopolysaccharide. Brain Behav Immun
(2006) 20(2):144–58.
92. Sominsky L, Fuller EA, Bondarenko E, Ong LK, Averell L, Nalivaiko E, et al.
Functional programming of the autonomic nervous system by early life immune
exposure: implications for anxiety. PLoS One (2013) 8(3):e57700. doi:10.1371/
journal.pone.0057700
93. Walker AK, Nakamura T, Hodgson DM. Neonatal lipopolysaccharide exposure
alters central cytokine responses to stress in adulthood in Wistar rats. Stress
(2010) 13(6):506–15. doi:10.3109/10253890.2010.489977
94. Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu XN, et al. Postnatal
microbial colonization programs the hypothalamic-pituitary-adrenal system
for stress response in mice. J Physiol (2004) 558(Pt 1):263–75. doi:10.1113/
jphysiol.2004.063388
95. Bailey MT, Dowd SE, Galley JD, Hufnagle AR, Allen RG, Lyte M. Exposure to a
social stressor alters the structure of the intestinal microbiota: implications for
stressor-induced immunomodulation. Brain Behav Immun (2011) 25(3):397–407.
doi:10.1016/j.bbi.2010.10.023
96. Galley JD, Nelson MC, Yu Z, Dowd SE, Walter J, Kumar PS, et al. Exposure to a
social stressor disrupts the community structure of the colonic mucosa-associated
microbiota. BMC Microbiol (2014) 14:189. doi:10.1186/1471-2180-14-189
97. Bailey MT, Coe CL. Maternal separation disrupts the integrity of the intestinal
microora in infant rhesus monkeys. Dev Psychobiol (1999) 35(2):146–55.
doi:10.1002/(SICI)1098-2302(199909)35:2<146::AID-DEV7>3.3.CO;2-7
98. de Jonge WJ. e gut’s little brain in control of intestinal immunity. ISRN
Gastroenterol (2013) 2013:630159. doi:10.1155/2013/630159
99. Bien J, Palagani V, B ozko P. e intestinal microbiota dysbiosis and Clostridium
dicile infection: is there a relationship with inammatory bowel disease? erap
Adv Gastroenterol (2013) 6(1):53–68. doi:10.1177/1756283X12454590
100. Ahrne S, Hagslatt ML. Eect of lactobacilli on paracellular permeability in the
gut. Nutrients (2011) 3(1):104–17. doi:10.3390/nu3010104
101. Bergmann KR, Liu SX, Tian R, Kushnir A, Turner JR, Li HL, et al. Bidobacteria
stabilize claudins at tight junctions and prevent intestinal barrier dysfunction
in mouse necrotizing enterocolitis. Am J Pathol (2013) 182(5):1595–606.
doi:10.1016/j.ajpath.2013.01.013
102. Leclercq S, Matamoros S, Cani PD, Neyrinck AM, Jamar F, Starkel P, et al.
Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of
alcohol-dependence severity. Proc Natl Acad Sci U S A (2014) 111(42):E4485–93.
doi:10.1073/pnas.1415174111
103. B arreau F, Hugot JP. Intestinal barrier dysfunction triggered by invasive bacteria.
Curr Opin Microbiol (2014) 17:91–8. doi:10.1016/j.mib.2013.12.003
104. Troeger H, Richter JF, Beutin L, Gunzel D, Dobrindt U, Epple HJ, et al.
Escherichia coli alpha-haemolysin induces focal leaks in colonic epithelium: a
novel mechanism of bacterial translocation. Cell Microbiol (2007) 9(10):2530–40.
doi:10.1111/j.1462-5822.2007.00978.x
105. Moreno-Navarrete JM, Manco M, Ibanez J, Garcia-Fuentes E, Ortega F,
Gorostiaga E, et al. Metabolic endotoxemia and saturated fat contribute to
circulating NGAL concentrations in subjects with insulin resistance. Int J Obes
(2010) 34(2):240–9. doi:10.1038/ijo.2009.242
106. Amar J, Burcelin R, Ruidavets JB, Cani PD, Fauvel J, Alessi MC, et al. Energy
intake is associated with endotoxemia in apparently healthy men. Am J Clin Nutr
(2008) 87(5):1219–23.
107. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic
endotoxemia initiates obesity and insulin resistance. Diabetes (2007) 56(7):1761–
72. doi:10.2337/db06-1491
10 8. Ghanim H, Abuaysheh S, Sia CL, Korzeniewski K, Chaudhuri A, Fernandez-Real
JM, et al. Increase in plasma endotoxin concentrations and the expression of
toll-like receptors and suppressor of cytokine signaling-3 in mononuclear cells
aer a high-fat, high-carbohydrate meal: implications for insulin resistance.
Diabetes Care (2009) 32(12):2281–7. doi:10.2337/dc09-0979
109. Herieka M, Erridge C. High-fat meal induced postprandial inammation. Mol
Nutr Food Res (2014) 58(1):136–46. doi:10.1002/mnfr.201300104
110. Deopurkar R, Ghanim H, Friedman J, Abuaysheh S, Sia CL, Mohanty P, et
al. Dierential eects of cream, glucose, and orange juice on inammation,
endotoxin, and the expression of toll-like receptor-4 and suppressor of cytokine
signaling-3. Diabetes Care (2010) 33(5):991–7. doi:10.2337/dc09-1630
111. Pendyala S, Walker JM, Holt PR. A high-fat diet is associated with endotox-
emia that originates from the gut. Gastroenterology (2012) 142(5):1100–1e2.
doi:10.1053/j.gastro.2012.01.034
112. Kelly CJ, Colgan SP, Frank DN. Of microbes and meals: the health con-
sequences of dietary endotoxemia. Nutr Clin Pract (2012) 27(2):215–25.
doi:10.1177/0884533611434934
113. Cani PD, Delzenne NM, Amar J, Burcelin R. Role of gut microora in the
development of obesity and insulin resistance following high-fat diet feeding.
Pathol Biol (2008) 56(5):305–9. doi:10.1016/j.patbio.2007.09.008
114. Cassaglia PA, Hermes SM, Aicher SA, Brooks VL. Insulin acts in the arcuate
nucleus to increase lumbar sympathetic nerve activity and baroreex function
in rats. J Physiol (2011) 589(Pt 7):1643–62. doi:10.1113/jphysiol.2011.205575
115. Simonds SE, Cowley MA. Hypertension in obesity: is leptin the culprit? Tre nds
Neurosci (2013) 36(2):121–32. doi:10.1016/j.tins.2013.01.004
116. Stumpel F, Scholtka B, Jungermann K. Stimulation by portal insulin of intestinal
glucose absorption via hepatoenteral nerves and prostaglandin E2 in the isolated,
jointly perfused small intestine and liver of the rat. Ann N Y Acad Sci (2000)
915:111–6. doi:10.1111/j.1749-6632.2000.tb05232.x
117. Sander GR, Cummins AG, Henshall T, Powell BC. Rapid disruption of
intestinal barrier function by gliadin involves altered expression of apical
junctional proteins. FEBS Lett (2005) 579(21):4851–5. doi:10.1016/j.
febslet.2005.07.066
1 18. Drago S, El Asmar R, Di Pierro M, Grazia Clemente M, Tripathi A, Sapone A, et al.
Gliadin, zonulin and gut permeability: eects on celiac and non-celiac intestinal
mucosa and intestinal cell lines. Scand J Gastroenterol (2006) 41(4):408–19.
doi:10.1080/00365520500235334
119. Lammers KM, Lu R, Brownley J, Lu B, Gerard C, omas K, et al. Gliadin
induces an increase in intestinal permeability and zonulin release by binding
to the chemokine receptor CXCR3. Gastroenterology (2008) 135(1):194–204e3.
doi:10.1053/j.gastro.2008.03.023
120. Chen P, Starkel P, Turner JR, Ho SB, Schnabl B. Dysbiosis-induced intestinal
inammation activates TNFRI and mediates alcoholic liver disease in mice.
Hepatology (2014) 61(3):883–94. doi:10.1002/hep.27489
121. Bode C, Kugler V, Bode JC. Endotoxemia in patients with alcoholic and
non-alcoholic cirrhosis and in subjects with no evidence of chronic liver
disease following acute alcohol excess. J Hepatol (1987) 4(1):8–14. doi:10.1016/
S0168-8278(87)80003-X
122. Fukui H, Brauner B, Bode JC, Bode C. Plasma endotoxin concentrations
in patients with alcoholic and non-alcoholic liver disease: reevaluation
with an improved chromogenic assay. J Hepatol (1991) 12(2):162–9.
doi:10.1016/0168-8278(91)90933-3
123. Parlesak A, Schafer C, Schutz T, Bode JC, B ode C. Increased intestinal permea-
bility to macromolecules and endotoxemia in patients with chronic alcohol abuse
May 2015 | Volume 6 | Article 22312
de Punder and Pruimboom
Endotoxemia and low-grade inammation
Frontiers in Immunology | www.frontiersin.org
in dierent stages of alcohol-induced liver disease. J Hepatol (2000) 32(5):742–7.
doi:10.1016/S0168-8278(00)80242-1
124. Oktedalen O, Lunde OC, Opstad PK, Aabakken L, Kvernebo K. Changes in
the gastrointestinal mucosa aer long-distance running. Scand J Gastroenterol
(1992) 27(4):270–4. doi:10.3109/00365529209000073
125. Ashton T, Young IS, Davison GW, Rowlands CC, McEneny J, Van Blerk C, et
al. Exercise-induced endotoxemia: the eect of ascorbic acid supplementation.
Free Radic Biol Med (2003) 35(3):284–91. doi:10.1016/S0891-5849(03)00309-5
126. Jeukendrup AE, Vet-Joop K, Sturk A, Stegen JH, Senden J, Saris WH, et al.
Relationship between gastro-intestinal complaints and endotoxemia, cytokine
release and the acute-phase reaction during and aer a long-distance triathlon
in highly trained men. Clin Sci (2000) 98(1):47–55. doi:10.1042/CS19990258
127. Pals KL, Chang RT, Ryan AJ, Gisol CV. Eect of running intensity on intestinal
permeability. J Appl Physiol (1997) 82(2):571–6.
1 28. Selkirk GA, McLellan TM, Wright HE, Rhind SG. Mild endotoxemia, NF-kappaB
translocation, and cytokine increase during exertional heat stress in trained
and untrained individuals. Am J Physiol Regul Integr Comp Physiol (2008)
295(2):R611–23. doi:10.1152/ajpregu.00917.2007
129. de Oliveira EP, Burini RC, Jeukendrup A. Gastrointestinal complaints during
exercise: prevalence, etiology, and nutritional recommendations. Sports Med
(2014) 44(Suppl 1):S79–85. doi:10.1007/s40279-014-0153-2
130. Hart EC, Charkoudian N. Sympathetic neural regulation of blood pressure: inu-
ences of sex and aging. Physiology (2014) 29(1):8–15. doi:10.1152/physiol.00031.2013
131. Ghosh S, Lertwattanarak R, Garduno JD, Galeana JJ, Li J, Zamarripa F, et al.
Elevated muscle TLR4 expression and metabolic endotoxemia in human aging.
J Gerontol A Biol Sci Med Sci (2014) 70(2):232–46. doi:10.1093/gerona/glu067
132. L assenius MI, Pietilainen KH, Kaartinen K, Pussinen PJ, Syrjanen J, Forsblom C,
et al. Bacterial endotoxin activity in human serum is associated with dyslipidemia,
insulin resistance, obesity, and chronic inammation. Diabetes Care (2011)
34(8):1809–15. doi:10.2337/dc10-2197
133. Moreno-Navarrete JM, Ortega F, Serino M, Luche E, Waget A, Pardo G, et al.
Circulating lipopolysaccharide-binding protein (LBP) as a marker of obesity-re-
lated insulin resistance. Int J Obes (2012) 36(11):1442–9. doi:10.1038/ijo.2011.256
134. Romani J, Caixas A, Escote X, Carrascosa JM, Ribera M, Rigla M, et al.
Lipopolysaccharide-binding protein is increased in patients with psoriasis with
metabolic syndrome, and correlates with C-reactive protein. Clin Exp Dermatol
(2013) 38(1):81–4. doi:10.1111/ced.12007
135. Sun L, Yu Z, Ye X, Zou S, Li H, Yu D, et al. A marker of endotoxemia is associated
with obesity and related metabolic disorders in apparently healthy Chinese.
Diabetes Care (2010) 33(9):1925–32. doi:10.2337/dc10-0340
136. Farhadi A, Gundlapalli S, Shaikh M, Frantzides C, Harrell L, Kwasny MM,
et al. Susceptibility to gut leakiness: a possible mechanism for endotox-
emia in non-alcoholic steatohepatitis. Liver Int (2008) 28(7):1026–33.
doi:10.1111/j.1478-3231.2008.01723.x
137. Ruiz AG, Casafont F, Crespo J, Cayon A, Mayorga M, Estebanez A, et al.
Lipopolysaccharide-binding protein plasma levels and liver TNF-alpha gene
expression in obese patients: evidence for the potential role of endotoxin in the
pathogenesis of non-alcoholic steatohepatitis. Obes Surg (2007) 17(10):1374–80.
doi:10.1007/s11695-007-9243-7
138. Al-Attas OS, Al-Daghri NM, Al-Rubeaan K, da Silva NF, Sabico SL, Kumar S,
et al. Changes in endotoxin levels in T2DM subjects on anti-diabetic therapies.
Cardiovasc Diabetol (2009) 8:20. doi:10.1186/1475-2840-8-20
139. Creely SJ, McTernan PG, Kusminski CM, Fisher FM, Da Silva NF, Khanolkar
M, et al. Lipopolysaccharide activates an innate immune system response in
human adipose tissue in obesity and type 2 diabetes. Am J Physiol Endocrinol
Metab (2007) 292(3):E740–7. doi:10.1152/ajpendo.00302.2006
140. Pussinen PJ, Havulinna AS, Lehto M, Sundvall J, Salomaa V. Endotoxemia is
associated with an increased risk of incident diabetes. Diabetes Care (2011)
34(2):392–7. doi:10.2337/dc10-1676
141. Harte AL, Varma MC, Tripathi G, McGee KC, Al-Daghri NM, Al-Attas OS, et al.
High fat intake leads to acute postprandial exposure to circulating endotoxin in
type 2 diabetic subjects. Diabetes Care (2012) 35(2):375–82. doi:10.2337/dc11-1593
142. Pussinen PJ, Tuomisto K, Jousilahti P, Havulinna AS, Sundvall J, Salomaa V.
Endotoxemia, immune response to periodontal pathogens, and systemic
inammation associate with incident cardiovascular disease events. Arterioscler
romb Vasc Biol (2007) 27(6):1433–9. doi:10.1161/ATVBAHA.106.138743
143. Lepper PM, Schumann C, Triantalou K, Rasche FM, Schuster T, Frank H, et al.
Association of lipopolysaccharide-binding protein and coronary artery disease
in men. J Am Coll Cardiol (2007) 50(1):25–31. doi:10.1016/j.jacc.2007.02.070
144. Lepper PM, Kleber ME, Grammer TB, Homann K, Dietz S, Winkelmann
BR, et al. Lipopolysaccharide-binding protein (LBP) is associated with total
and cardiovascular mortality in individuals with or without stable coronary
artery disease – results from the Ludwigshafen risk and cardiovascular
health study (LURIC). Atherosclerosis (2011) 219(1):291–7. doi:10.1016/j.
atherosclerosis.2011.06.001
145. Serrano M, Moreno-Navarrete JM, Puig J, Moreno M, Guerra E, Ortega F, et
al. Serum lipopolysaccharide-binding protein as a marker of atherosclerosis.
Atherosclerosis (2013) 230(2):223–7. doi:10.1016/j.atherosclerosis.2013.07.004
146. Wiedermann CJ, Kiechl S, Dunzendorfer S, Schratzberger P, Egger G,
Oberhollenzer F, et al. Association of endotoxemia with carotid atherosclerosis
and cardiovascular disease: prospective results from the Bruneck study. J Am
Coll Cardiol (1999) 34(7):1975–81. doi:10.1016/S0735-1097(99)00448-9
147. Niebauer J, Volk HD, Kemp M, Dominguez M, Schumann RR, Rauchhaus M, et
al. Endotoxin and immune activation in chronic heart failure: a prospective cohort
study. Lancet (1999) 353(9167):1838–42. doi:10.1016/S0140-6736(98)09286-1
148. Pastor Rojo O, Lopez San Roman A, Albeniz Arbizu E, de la Hera Martinez
A, Ripoll Sevillano E, Albillos Martinez A. Serum lipopolysaccharide-binding
protein in endotoxemic patients with inammatory bowel disease. Inamm
Bowel Dis (2007) 13(3):269–77. doi:10.1002/ibd.20019
149. Funderburg NT, Stubbleeld Park SR, Sung HC, Hardy G, Clagett B, Ignatz-
Hoover J, et al. Circulating CD4(+) and CD8(+) T cells are activated in inam-
matory bowel disease and are associated with plasma markers of inammation.
Immunology (2013) 140(1):87–97. doi:10.1111/imm.12114
150. Gardiner KR, Halliday MI, Barclay GR, Milne L, Brown D, Stephens S, et al.
Signicance of systemic endotoxemia in inammatory bowel disease. Gut (1995)
36(6):897–901. doi:10.1136/gut.36.6.897
151. Forsyth CB, Shannon KM, Kordower JH, Voigt RM, Shaikh M, Jaglin JA, et al.
Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein
staining and endotoxin exposure markers in early Parkinson’s disease. PLoS One
(2011) 6(12):e28032. doi:10.1371/journal.pone.0028032
152. Zhang R, Miller RG, Gascon R, Champion S, Katz J, Lancero M, et al. Circulating
endotoxin and systemic immune activation in sporadic amyotrophic lateral
sclerosis (sALS). J Neuroimmunol (2009) 206(1–2):121–4. doi:10.1016/j.
jneuroim.2008.09.017
153. Sparks Stein P, Steen MJ, Smith C, Jicha G, Ebersole JL, Abner E, et al. Serum
antibodies to periodontal pathogens are a risk factor for Alzheimer’s disease.
Alzheimer Dement (2012) 8(3):196–203. doi:10.1016/j.jalz.2011.04.006
154. Licht CM, de Geus EJ, Penninx BW. Dysregulation of the autonomic nervous
system predicts the development of the metabolic syndrome. J Clin Endocrinol
Metab (2013) 98(6):2484–93. doi:10.1210/jc.2012-3104
155. Kazakou P, Kyriazopoulou V, Michalaki M, Ierodiakonou V, Psyrogiannis
A, Habeos I. Activated hypothalamic pituitary adrenal axis in patients with
metabolic syndrome. Horm Metab Res (2012) 44(11):839–44. doi:10.105
5/s-0032-1311632
156. Garg M, Angus PW, Burrell LM, Herath C, Gibson PR, Lubel JS. Review
article: the pathophysiological roles of the renin-angiotensin system in
the gastrointestinal tract. Aliment Pharmacol Ther (2012) 35(4):414–28.
doi:10.1111/j.1365-2036.2011.04971.x
157 . Mawdsley JE, Rampton DS. Psychological stress in IBD: new insights into patho-
genic and therapeutic implications. Gut (2005) 54(10):1481–91. doi:10.1136/
gut.2005.064261
158. Caradonna L, Amati L, Magrone T, Pellegrino NM, Jirillo E, Caccavo D. Enteric
bacteria, lipopolysaccharides and related cytokines in inammatory bowel
disease: biological and clinical signicance. J Endotoxin Res (2000) 6(3):205–14.
doi:10.1177/09680519000060030101
159. Dinan TG, Stanton C, Cryan JF. Psychobiotics: a novel class of psychotropic.
Biol Psychiatry (2013) 74(10):720–6. doi:10.1016/j.biopsych.2013.05.001
Conict of Interest Statement: e authors declare that the research was conducted
in the absence of any commercial or nancial relationships that could be construed
as a potential conict of interest.
Copyright © 2015 de Punder and Pruimboom. is is an open-access article distributed
under the terms of the Creative Commons Attribution License (CC BY). e use, distri-
bution and reproduction in other forums is permitted, provided the original author(s) or
licensor are credited and that the original publication in this journal is cited, in accordance
with accepted academic practice. No use, distribution or reproduction is permitted which
does not comply with these terms.
... Thus, if the disruption of inflammatory responses in association with chronic systemic inflammation is a common factor among many pathologies, such as diabetes [39,58], obesity [58,59], sepsis [60], and others [22,48,61], it is reasonable to hypothesize that in rotating-shift workers, given enough time, similar changes in functional outcomes of cellmediated immunity may eventually develop and explain the increased risk of infection among this population. ...
... Studies have found that systemic inflammation may trigger the reprogramming of innate and even adaptive immune responses [61,73]. One of the mechanisms by which reprogramming of immune responses occurs is known as endotoxin tolerance, which may help explain the seemingly paradoxical observation of impaired responses to LPS stimulation by an overactive immune system [74][75][76]. ...
Article
Full-text available
Shift work is a common labor practice affecting nearly 30% of the U.S. workforce. Long-term, rotating-shift work is particularly harmful to health. Persistent sleep deprivation in shift workers, among other factors, facilitates the development of a state of subclinical but chronic systemic inflammation with a high incidence and prevalence of infections and inflammation-related pathologies, suggesting an underlying disruption of immune responses. However, despite this state of chronic immune activation, cell-mediated inflammatory responses in rotating-shift workers are poorly understood. Here, we used lipopolysaccharide (LPS) to stimulate peripheral blood mononuclear cells (PBMCs) isolated from rotating-shift workers and healthy day-shift workers and investigate their immune responses. The results showed that PBMCs from rotating-shift workers had a dampened inflammatory response. Specifically, the secretion of LPS-induced TNF-α in culture supernatants was significantly reduced compared to the response found in PBMCs from day-shift workers. However, anti-inflammatory responses, reflected by the secretion of LPS-induced IL-10, were indistinguishable between PBMCs from day-shift and rotating-shift workers. In addition, the correlation between the plasma concentration of lipopolysaccharide-binding protein (LBP, a marker of systemic inflammation) and LPS-induced responses was disrupted only in rotating-shift workers, suggesting that in this group, an impaired mechanism that weakens the relationship between pro- and anti-inflammatory signaling may underlie the hypo-responsiveness of PBMCs. Our results suggest that persistent subclinical systemic inflammation in rotating-shift workers disrupts cell-mediated immunity, increasing the risk of infection and other inflammation-related pathologies in this population.
... Due to the humoral (e.g., catecholamine, cortisol) and nervous connection (adrenergic sympathetic nerve fibers) between the gut and brain, both axes (SAM and HPA) could contribute to increasing the paracellular permeability in elevating the diffusion of water and ions needed. Therefore, this could also allow the transfer of endotoxins that could in turn trigger low-grade inflammation [152,153]. Interestingly, in animal models, blockage of the sympathetic nervous system leads to a decrease in the permeability induced by endotoxins [154]. Further studies in humans need to be conducted due to the heterogeneity of markers and psychological stressors used, but evidence still shows that stress could also be involved in gut barrier disruption [150,152,153]. ...
... Interestingly, in animal models, blockage of the sympathetic nervous system leads to a decrease in the permeability induced by endotoxins [154]. Further studies in humans need to be conducted due to the heterogeneity of markers and psychological stressors used, but evidence still shows that stress could also be involved in gut barrier disruption [150,152,153]. ...
Article
Full-text available
Women exhibit unique vulnerabilities in health, especially regarding mental health and neurodegenerative diseases. Biological, hormonal, and metabolic differences contribute to sex-specific risks that remain underrepresented in clinical studies. Diseases such as major depressive disorder (MDD) and Alzheimer’s disease (AD) are more prevalent in women and may be influenced by hormonal transitions, particularly during menopause. Chronic low-grade inflammation is emerging as a shared mechanism underlying both conditions, and this inflammatory state can be worsened by dietary habits. During menopause, mood and sleep disturbances can influence dietary behavior, leading to enhanced snacking and consumption of high-glycemic and comfort foods. Such foods, low in nutritional value, promote weight gain and elevated inflammatory markers. Their consumption combined (or not) with a preexisting Western diet pattern—already linked to inflammation—could reinforce systemic inflammation involving the gut–brain axis. Moreover, the symptoms “per se” could act on inflammation as well. Peripheral inflammation may cross the blood–brain barrier, sustaining mood disorders and promoting neurodegenerative changes. Finally, MDD and AD are both associated with conditions such as obesity and diabetes, which occur more frequently in women. The review highlights how menopause-related changes in mood, sleep, and diet may heighten susceptibility to mental and neurodegenerative diseases.
... In addition, stress-induced "leaky gut" allows LPS and other microbial products into the bloodstream, causing systemic inflammation that contributes to metabolic and neuroinflammatory disorders, such as obesity, type 2 diabetes, anxiety, and depression. [75][76][77] Anxiety and depression, two of the most common mental health disorders, are closely linked to changes in the gut microbiota. Individuals with these conditions often show reduced microbial diversity and a decline in beneficial bacteria, such as Lactobacillus and Bifidobacterium, which play a role in producing neurotransmitters critical for mood regulation. ...
... Individuals with these conditions often show reduced microbial diversity and a decline in beneficial bacteria, such as Lactobacillus and Bifidobacterium, which play a role in producing neurotransmitters critical for mood regulation. [71][72][73][74][75] Chronic low-grade inflammation has been implicated in the pathophysiology of both anxiety and depression, as it interferes with neurotransmitter production and signaling, particularly serotonin, a key regulator of mood, appetite, and sleep. 69,78 Dysbiosis impacts the production and regulation of several key neurotransmitters. ...
Article
Full-text available
The gut–brain axis represents a dynamic, bidirectional communication system linking the central nervous system and the gastrointestinal (GI) tract through neural, hormonal, and immune pathways. A growing body of research highlights the role of gut microbiota in modulating neuroinflammatory responses, neurotransmitter production, and vagal nerve signaling. However, while numerous reviews have addressed gut microbiota’s impact on health and disease, few have systematically compared findings across different study models or emphasized recent advances in therapeutic interventions. Hence, this review consolidates present research on the interplay between gut microbiota, psychological conditions (stress, anxiety, and depression), and GI disorders (irritable bowel syndrome and inflammatory bowel disease). This review explores the latest methodologies, including genomic approaches and metabolomics, to understand microbial signatures in health and disease. Furthermore, it examines therapeutic interventions, including probiotics, pre-biotics, dietary modifications, and fecal microbiota transplantation, and their efficacy across different studies. The gut–brain axis plays a crucial role in health and disease through complex interactions between gut microbiota, psychological conditions, and GI disorders. By consolidating recent research and advancements in therapeutic interventions, this review provides a comprehensive perspective on emerging methodologies and potential treatments. A deeper understanding of these mechanisms may pave the way for more effective, personalized therapeutic strategies.
... This neuroinflammation may be explained by changes in the intestine that generate localized inflammation, which, over time, could progress into systemic inflammation (Thevaranjan et al., 2017;Haruwaka et al., 2019), potentially manifesting as neuroinflammation. Moreover, neuroinflammation could be triggered by the passage of endotoxins from the intestine into the bloodstream, which may eventually reach the brain (De Punder and Pruimboom, 2015). Additionally, alterations in intestinal permeability have been linked to changes in the permeability of the blood-brain barrier (BBB), leading to the activation of glial cells (microglia and astrocytes) to protect the BBB (Haruwaka et al., 2019). ...
... It has been proposed that alterations in the intestinal barrier may contribute to anxiety and depression by triggering neuroinflammation through the passage of endotoxins into the bloodstream (De Punder and Pruimboom, 2015). Over time, intestinal changes can lead to systemic inflammation, manifesting as neuroinflammation (Thevaranjan et al., 2017;Haruwaka et al., 2019), which can affect synaptic plasticity (Doan et al., 2015;Lima Giacobbo et al., 2019). ...
Preprint
Early life stress (ELS) can significantly influence vulnerability to the development of psychiatric disorders in adulthood. One of the most widely used preclinical models for investigating ELS is maternal separation with early weaning (MSEW), which mimics early-life neglect. The objective of this study was to evaluate the impact of ELS induced by MSEW on the emotional behaviour of male and female mice, as well as its relationship with intestinal permeability and neuroinflammatory markers in the hippocampus. Our results show that MSEW leads to increased anxiety-like behaviours in the adulthood, particularly in females, and exacerbates depression-like behaviours and anhedonia in both sexes. Notably, increased intestinal permeability was observed, which correlated with higher anxiety and depression-like responses, suggesting that gut health plays a crucial role in emotional regulation. These alterations in intestinal permeability were long-lasting, indicating persistent effects on gut function following ELS. Additionally, we observed that MSEW animals showed higher BDNF expression in the hippocampus, particularly in males. However, we did not find significant differences in the long-term survival of adult-born hippocampal cells, as measured by BrdU+ labeling. Furthermore, upon exposure to MSEW, both sexes showed increased NF-κB protein levels. However, only MSEW male mice exhibited changes in TNF-α and BDNF levels, suggesting a sex-specific regulatory mechanism in response to chronic stress. The novel contribution of this study is its exploration of intestinal permeability as a mechanism linking ELS to emotional and behavioural dysregulation, particularly anxiety and depression. By showing a long-lasting increase in intestinal permeability and its correlation with mood disorders, our study extends the gut-brain axis hypothesis to ELS. Additionally, the inclusion of both male and female mice offers a more comprehensive understanding of the sex-specific effects of early stress, often overlooked in other studies. These findings suggest that intestinal permeability could serve as a biomarker for stress-related psychiatric conditions.
... Endotoxemia, defined as the presence of bacterial endotoxin in the blood, is a significant contributor to acute death and the progression of chronic diseases in humans and animals. This inflammatory condition results from the translocation of endotoxins, such as lipopolysaccharide (LPS)-containing fragments from the outer membrane of gram-negative bacteria in the gastrointestinal tract, mammary gland, or urogenital tract into systemic circulation following stress or damage to mucosal barriers (1,2). Interactions between LPS and toll-like receptors (TLR) found on dendritic cells, macrophages, neutrophils, and non-immune cells, such as adipocytes, stimulate the production of reactive oxygen species, pro-inflammatory cytokines, and lipid-based mediators of inflammation (3). ...
Article
Full-text available
Introduction Endotoxemia, marked by the presence of bacterial lipopolysaccharide (LPS) in the bloodstream, induces acute inflammation and is implicated in both mortality and chronic disease across species. LPS stimulates lipolysis and activates cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP450) enzymes, promoting the synthesis of bioactive lipid mediators known as oxylipins (OXL). However, the dynamics of OXL production during systemic inflammation remain poorly defined, particularly in large animals. Methods To investigate OXL responses to endotoxemia, mature Holstein cows were administered intravenous infusions of either LPS or sterile saline (SAL). Plasma samples were collected at baseline (PRE), 2 hours post-infusion (+2H), and 12 hours post-infusion (+12H). OXL profiles were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Complementary in vitro experiments were conducted using bovine adipocytes exposed to LPS to assess adipocyte-specific OXL release. Results LPS-treated cows exhibited classical signs of endotoxemia, including tachycardia, fever, and tachypnea. Plasma OXL profiling revealed significant alterations in arachidonic acid (AA)- and eicosapentaenoic acid (EPA)-derived pathways. Notably, LPS infusion led to persistent increases in COX- and LOX-derived pro-inflammatory OXL, including thromboxane B₂ and hydroxyeicosatetraenoic acids (HETEs), alongside transient elevations in EPA- and docosahexaenoic acid (DHA)-derived pro-resolving mediators. In vitro, LPS stimulation of adipocytes increased the release of AA-based 5-HETE, 6-keto-PGF₁α, linoleic acid (LA)-based 13-HODE, and DHA-based 19,20-DiHDPA. Discussion These findings indicate that LPS induces robust activation of pro-inflammatory OXL pathways with limited and transient engagement of pro-resolving lipid mediators. The imbalance may contribute to sustained or dysregulated inflammation. Our study provides novel insights into both systemic and adipocyte-specific OXL dynamics during endotoxemia and highlights their potential as biomarkers and therapeutic targets for modulating inflammation.
... Dietinduced obesity disrupts the gut barrier and allows LPS entry, impairing insulin signalling and promoting metabolic dysfunction. Elevated circulating endotoxin levels also correlate with metabolic syndrome risk factors like dyslipidaemia (de Punder & Pruimboom, 2015), corroborating the idea that chronic inflammation is linked to some extent to metabolic maladaptation. This emerging field underscores the intricate interplay between immune responses and metabolic regulation, revealing that immune cells function not only as responders to infection and injury but also as key regulators of metabolic processes. ...
Article
Full-text available
Inflammation is vital for defence against injury and infection, but excessive inflammation can lead to tissue damage and disease. The central nervous system (CNS) helps regulate immune responses through neuroendocrine pathways, such as the hypothalamic–pituitary–adrenal axis and the anti‐inflammatory reflex, which limit systemic inflammation. Immune responses require significant metabolic energy and the liver adapts by increasing glucose production and mobilizing fatty acids. This immune‐metabolic coordination is mediated by cytokines and metabolic regulators. This review explores how the anti‐inflammatory reflex modulates the interplay between inflammation and metabolism during endotoxaemia, with a focus on the haem oxygenase 1/carbon monoxide (HO1/CO) pathway. Carbon monoxide, a byproduct of HO1 activity, acts as a key signalling molecule that reduces inflammation, supports mitochondrial function and protects tissues. Understanding this pathway provides new insights into potential therapeutic strategies for treating inflammatory and metabolic disorders by targeting neuroimmune‐metabolic communication networks.
Article
Full-text available
Exposure to stress during sensitive developmental periods comes with long term consequences for neurobehavioral outcomes and increases vulnerability to psychopathology later in life. While we have advanced our understanding of the mechanisms underlying the programming effects of early-life stress (ES), these are not yet fully understood and often hard to target, making the development of effective interventions challenging. In recent years, we and others have suggested that nutrition might be instrumental in modulating and possibly combatting the ES-induced increased risk to psychopathologies and neurobehavioral impairments. Nutritional strategies are very promising as they might be relatively safe, cheap and easy to implement. Here, we set out to comprehensively review the existing literature on nutritional interventions aimed at counteracting the effects of ES on neurobehavioral outcomes in preclinical and clinical settings. We identified eighty six rodent and ten human studies investigating a nutritional intervention to ameliorate ES-induced impairments. The human evidence to date, is too few and heterogeneous in terms of interventions, thus not allowing hard conclusions, however the preclinical studies, despite their heterogeneity in terms of designs, interventions used, and outcomes measured, showed nutritional interventions to be promising in combatting ES-induced impairments. Furthermore, we discuss the possible mechanisms involved in the beneficial effects of nutrition on the brain after ES, including neuroinflammation, oxidative stress, hypothalamus-pituitary-adrenal axis regulation and the microbiome-gut-brain axis. Lastly, we highlight the critical gaps in our current knowledge and make recommendations for future research to move the field forward.
Article
Full-text available
Liver disease is associated with qualitative and quantitative changes in the intestinal microbiota. In cirrhotic patients the alteration in gut microbiota is characterized by an overgrowth of potentially pathogenic bacteria (i.e., gram negative species) and a decrease in autochthonous familiae. Here we summarize the available literature on the risk of gut dysbiosis in liver cirrhosis and its clinical consequences. We therefore described the features of the complex interaction between gut microbiota and cirrhotic host, the so called "gut-liver axis", with a particular attention to the acquired risk of bacterial translocation, systemic inflammation and the relationship with systemic infections in the cirrhotic patient. Such knowledge might help to develop novel and innovative strategies for the prevention and therapy of gut dysbiosis and its complication in liver cirrhosis.
Article
Full-text available
Liver disease is associated with qualitative and quantitative changes in the intestinal microbiota. In cirrhotic patients the alteration in gut microbiota is characterized by an overgrowth of potentially pathogenic bacteria (i.e., gram negative species) and a decrease in autochthonous familiae. Here we summarize the available literature on the risk of gut dysbiosis in liver cirrhosis and its clinical consequences. We therefore described the features of the complex interaction between gut microbiota and cirrhotic host, the so called "gut-liver axis", with a particular attention to the acquired risk of bacterial translocation, systemic inflammation and the relationship with systemic infections in the cirrhotic patient. Such knowledge might help to develop novel and innovative strategies for the prevention and therapy of gut dysbiosis and its complication in liver cirrhosis.
Article
Full-text available
Significance Alcohol-dependent subjects frequently develop emotional symptoms that contribute to the persistence of alcohol drinking. These subjects are also characterized by gastrointestinal disturbances. In this study, we showed that alcohol-dependent subjects with altered intestinal permeability had also altered gut-microbiota composition and activity and remained with high scores of depression, anxiety, and alcohol craving after a short-term detoxification program. These results are consistent with the existence of a gut–brain axis in alcohol dependence, in which the gut microbiota could alter the gut-barrier function and influence behavior in alcohol dependence. Therefore, this study opens a previously unidentified field of research for the treatment and the management of alcohol dependence, targeting the gut microbiota.
Article
Full-text available
Background The microbiota of the mammalian gastrointestinal (GI) tract consists of diverse populations of commensal bacteria that interact with host physiological function. Dysregulating these populations, through exogenous means such as antibiotics or dietary changes, can have adverse consequences on the health of the host. Studies from laboratories such as ours have demonstrated that exposure to psychological stressors disrupts the population profile of intestinal microbiota. To date, such studies have primarily focused on prolonged stressors (repeated across several days) and have assessed fecal bacterial populations. It is not known whether shorter stressors can also impact the microbiota, and whether colonic mucosa-associated populations can also be affected. The mucosa-associated microbiota exist in close proximity to elements of the host immune system and the two are tightly interrelated. Therefore, alterations in these populations should be emphasized. Additionally, stressors can induce differential responses in anxiety-like behavior and corticosterone outputs in variant strains of mice. Thus, whether stressor exposure can have contrasting effects on the colonic microbiota in inbred C57BL/6 mice and outbred CD-1 mice was also examined. Results In the present study, we used high throughput pyrosequencing to assess the effects of a single 2-hour exposure to a social stressor, called social disruption (SDR), on colonic mucosa-associated microbial profiles of C57BL/6 mice. The data indicate that exposure to the stressor significantly changed the community profile and significantly reduced the relative proportions of two genera and one family of highly abundant intestinal bacteria, including the genus Lactobacillus. This finding was confirmed using a quantitative real-time polymerase chain reaction (qPCR) technique. The use of qPCR also identified mouse strain-specific differences in bacterial abundances. L. reuteri, an immunomodulatory species, was decreased in stressor-exposed CD-1 mice, but not C57BL/6 mice. Conclusions These data illustrate that stressor exposure can affect microbial populations, including the lactobacilli, that are closely associated with the colonic mucosa. Because the lactobacilli can have beneficial effects on human health, stressor-induced reductions of their population could have important health implications.
Article
Introduction. Increasing experimental data reveal the importance of the gut bacteria in mediating behavioral changes. However, clinical evidences are lacking. In one hand, alcohol-dependent subjects (AD) are known to present with depression, anxiety and neuropsychological disorders. On the other hand, heavy and chronic alcohol consumption is suggested to alter the gut microbiota composition. The aim …
Article
Background Increased intestinal permeability has been reported in one study of adult asthmatics. Aim To determine whether children with asthma have altered intestinal permeability. Methods Thirty two asthmatic children, and 32 sex and age matched controls were recruited. The dual sugar (lactulose and mannitol) test was used to evaluate intestinal permeability, and the percentage of ingested lactulose (L) and mannitol (M) in the urine, and the L:M ratio were determined. All patients were skin prick tested for common aeroallergens, and specific IgE to some food items was determined. Results The median value of L in asthmatic children (2.29, IQR 0.91–4.07) was significantly higher than that in controls (0.69, IQR 0.45–1.08), and that of M was almost similar. The ratio L:M was significantly higher in asthmatic children (0.20, IQR 0.11–0.40) than in controls (0.06, IQR 0.04–0.09). Intestinal permeability did not correlate with eczema, inhaled steroids, positive skin prick test to aeroallergens, or severity of asthma. Conclusions Intestinal permeability is increased in children with asthma, suggesting that the whole mucosal system may be affected.
Article
Unlabelled: Intestinal barrier dysfunction is an important contributor to alcoholic liver disease (ALD). Translocated microbial products trigger an inflammatory response in the liver and contribute to steatohepatitis. Our aim was to investigate mechanisms of barrier disruption after chronic alcohol feeding. A Lieber-DeCarli model was used to induce intestinal dysbiosis, increased intestinal permeability, and liver disease in mice. Alcohol feeding for 8 weeks induced intestinal inflammation in the jejunum, which is characterized by an increased number of tumor necrosis factor alpha (TNF-α)-producing monocytes and macrophages. These findings were confirmed in duodenal biopsies from patients with chronic alcohol abuse. Intestinal decontamination with nonabsorbable antibiotics restored eubiosis, decreased intestinal inflammation and permeability, and reduced ALD in mice. TNF-receptor I (TNFRI) mutant mice were protected from intestinal barrier dysfunction and ALD. To investigate whether TNFRI on intestinal epithelial cells mediates intestinal barrier dysfunction and ALD, we used TNFRI mutant mice carrying a conditional gain-of-function allele for this receptor. Reactivation of TNFRI on intestinal epithelial cells resulted in increased intestinal permeability and liver disease that is similar to wild-type mice after alcohol feeding, suggesting that enteric TNFRI promotes intestinal barrier dysfunction. Myosin light-chain kinase (MLCK) is a downstream target of TNF-α and was phosphorylated in intestinal epithelial cells after alcohol administration. Using MLCK-deficient mice, we further demonstrate a partial contribution of MLCK to intestinal barrier dysfunction and liver disease after chronic alcohol feeding. Conclusion: Dysbiosis-induced intestinal inflammation and TNFRI signaling in intestinal epithelial cells mediate a disruption of the intestinal barrier. Therefore, intestinal TNFRI is a crucial mediator of ALD.
Article
Background Glucocorticoids (GCs) are steroid hormones used to induce remission in moderate-to-severe inflammatory bowel disease (IBD). A substantial fraction of patients do not respond to GC treatment and require alternate therapies or surgery. At present, non-response can only be assessed empirically by observing continued disease activity. Methods To identify potential biomarkers of GC response, we retrospectively identified and recruited 18 GC-responsive and 18 GC-nonresponsive IBD patients. This sample included 14 patients with ulcerative colitis (UC) and 22 patients with Crohn's disease (CD), all previously treated with steroids. In peripheral blood mononuclear cells from each patient, we performed in vitro assays to measure GC inhibition of three different immune stimulants (phytohemagglutinin [PHA], α-CD3/α-CD28, and lipopolysaccharide [LPS]). Results In both diseases, we found that inhibition of PHA-mediated T cell proliferation was significantly associated with clinical GC response (P = 0.04). Inhibition of proliferation due to direct T cell receptor stimulation using α-CD3/α-CD28 was also significantly associated with clinical GC response in UC patients (P = 0.009), but not in CD patients (P = 0.78). Interestingly, inhibition of LPS-mediated cytokine secretion showed the strongest association with clinical GC response across both diseases (P = 0.005). Conclusions We show that inhibition of LPS stimulation is more strongly associated with clinical GC response in IBD patients than inhibition of PHA and α-CD3/α-CD28-mediated proliferation. These results support an important role of bacterial recognition and innate immunity in the etiology of IBD. This assay could be a powerful predictor of clinical response to GCs.