ArticlePDF Available

The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders

Authors:
  • VitaImmun Medical Centre Poznań, Poland

Abstract

Depressive episodes are associated not only with changes in neurotransmission in the central nervous system, but also may lead to structural changes in the brain through neuroendocrine, inflammatory, and immunological mechanisms. The aim of this article is to present a new hypothesis connecting the inflammatory theory of depression with IgG food hypersensitivity and leaky gut syndrome. This new potential pathway that may mediate the pathogenesis of depression implies the existence of subsequent developmental stages. Overproduction of zonulin triggered, for example, by gliadin through activation of the epidermal growth factor receptor and protease-activated receptor causes loosening of the tight junction barrier and an increase in permeability of the gut wall ('leaky gut'). This results in a process allowing larger molecules that would normally stay in the gut to cross into the bloodstream and in the induction of IgG-dependent food sensitivity. This condition causes an increased immune response and consequently induces the release of proinflammatory cytokines, which in turn may lead to the development of depressive symptoms. It seems advisable to assess the intestinal permeability using as a marker, for example, zonulin and specific IgG concentrations against selected nutritional components in patients with depression. In the case of increased IgG concentrations, the implementation of an elimination-rotation diet may prove to be an effective method of reducing inflammation. This new paradigm in the pathogenesis of depressive disorders linking leaky gut, IgG-dependent food sensitivity, inflammation, and depression is promising, but still needs further studies to confirm this theory.
The role of IgG hypersensitivity in the
pathogenesis and therapy of
depressive disorders
Hanna Karakuła-Juchnowicz1, Patrycja Szachta2, Aneta Opolska3,
Justyna Morylowska-Topolska1, Mirosława Gałęcka2,
Dariusz Juchnowicz4,PawełKrukow1, Lasik Zofia2
1
Department of Clinical Neuropsychiatry Medical University, Lublin, Poland,
2
Institute for Microecology,
Poznań, Poland,
3
Department of Dietetics Higher School of Social Sciences, Lublin, Poland,
4
Department of Psychology University of Pedagogy, Bialystok, Poland
Depressive episodes are associated not only with changes in neurotransmission in the central nervous
system, but also may lead to structural changes in the brain through neuroendocrine, inflammatory, and
immunological mechanisms. The aim of this article is to present a new hypothesis connecting the
inflammatory theory of depression with IgG food hypersensitivity and leaky gut syndrome. This new
potential pathway that may mediate the pathogenesis of depression implies the existence of subsequent
developmental stages. Overproduction of zonulin triggered, for example, by gliadin through activation of
the epidermal growth factor receptor and protease-activated receptor causes loosening of the tight
junction barrier and an increase in permeability of the gut wall (leaky gut). This results in a process
allowing larger molecules that would normally stay in the gut to cross into the bloodstream and in the
induction of IgG-dependent food sensitivity. This condition causes an increased immune response and
consequently induces the release of proinflammatory cytokines, which in turn may lead to the
development of depressive symptoms. It seems advisable to assess the intestinal permeability using as a
marker, for example, zonulin and specific IgG concentrations against selected nutritional components in
patients with depression. In the case of increased IgG concentrations, the implementation of an
eliminationrotation diet may prove to be an effective method of reducing inflammation. This new
paradigm in the pathogenesis of depressive disorders linking leaky gut, IgG-dependent food sensitivity,
inflammation, and depression is promising, but still needs further studies to confirm this theory.
Keywords: Depression, Leaky gut, IgG hypersensitivity, Zonulin, Inflammatory theory of depression, Gluten sensitivity
Introduction
Depression is a heterogeneous psychiatric disorder
with multifactorial aetiology and therefore needs
improved integration models, based on behavioural
studies, sociology, and neuroscience to better reflect
both the complexity and variety of mood disorders.
1
Among the factors deserving special attention are bio-
logical ones, including psychoneuroendocrinology and
psychoimmunology, posing a bridge between strictly
biological and psychological approaches.
2
More and
more evidence indicates that depressive episodes are
associated not only with changes in neurotransmission
in the central nervous system (CNS), but also may
lead to structural changes in the brain through
neuroendocrine, inflammatory, and immunological
mechanisms.
35
Different factors potentially con-
nected with systemic inflammation in depression are
taken into consideration; these include psychosocial
stressors, poor diet, physical inactivity, obesity,
smoking, altered gut permeability, atopy, dental
caries, sleep, and Vitamin D deficiency.
6
The aim of this article is to present a new hypothesis
connecting the inflammatory theory of depression
with IgG food hypersensitivity and leaky gut
syndrome (LGS).
Inflammatory theory of depression
Among many theories of depression, the cytokine
(macrophage) theory of depression, first demonstrated
in 1991 by Robert Smith,
7
has aroused much interest
among researchers. It is assumed that changes in
behaviour, typical of depression, are the result of the
Correspondence to: Hanna Karakuła-Jachnowicz, Department of Clinical
Neuropsychiatry, Medical University of Lublin, ul. Głuska 1, 20-439
Lublin, Poland. Email: karakula.hanna@gmail.com, hanna.karakula@
umlub.pl
©W.S.Maney&SonLtd2014
MORE OpenChoice articles are open access and distributed under the terms of the Creative Commons Attribution Non-Commercial License 3.0
DOI 10.1179/1476830514Y.0000000158 Nutritional Neuroscience 2014 VOL. 0NO. 01
interaction of proinflammatory cytokines produced in
the peripheral and/or CNS with the neuroendocrine
system. This leads to activation of the hypothalamic-
pituitary-adrenal (HPA) axis and elimination of
tryptophan, a serotonin precursor (as a result of
activation of indole 2,3-dioxygenase, an enzyme that
converts tryptophan into kynurenine).
8
Cytokines are a large group of more than 100 regu-
latory proteins, proinflammatory and anti-inflamma-
tory mediators, which can be considered immune
regulating hormones that regulate growth, prolifer-
ation, and cell activity.
9
A wide range of biological
activities of cytokines known so far includes pyrogenic
activity, hyperalgesic activity, the effect on energy
balance in the body by changing the appetite level
and metabolism, modulation of the autonomic
nervous system activity, the effect on the functioning
and structure of the cardiovascular system, mood low-
ering effects, increased drowsiness, and the regulation
of hormones, and other cytokines.
10
Cytokines are
also signalling molecules involved in a diverse set of
physiological roles with extensive cross-talk.
11,12
The increase in the proinflammatory cytokine con-
centration and their effects on the CNS contribute to
the development of neuropsychological and somatic
depressive symptoms.
13
Many studies conducted so far have shown elevated
levels of proinflammatory cytokines in the serum of
patients with a major depressive episode. In these
studies, multiple cytokines such as tumour necrosis
factor (TNF-α), interferon-γ, interleukin IL-1β, IL-2,
IL-4, IL-6, IL-8, and IL-10 were taken into account.
Two recent meta-analyses confirmed the importance
of higher interleukin-1, -6, and TNF-αlevels in the
serum of patients with depression.
14,15
Elevated levels
of these cytokines in the cerebrospinal fluid of depress-
ive patients were also shown in numerous studies.
16,17
The importance of elevated C-reactive protein is high-
lighted
18,19
in the absence of clearly consistent findings
with regard to other cytokines.
15
An interesting
phenomenon, confirming the link between the inflam-
matory process and depression symptoms, is the
co-occurrence of depression with inflammatory diseases
such as asthma, chronic obstructive pulmonary disease,
diabetes, allergy, and rheumatoid arthritis.
20
Immunostimulatory treatment using interferon-
alpha (IFN-alpha) in hepatitis C or cancer (mela-
noma, leukaemia) shows that this treatment is
associated with much higher rates of depression as
compared to those of the general population.
21
Among patients receiving IFN-alpha, the percentage
of depressed individuals is nearly 45%.
22
Further
evidence on the important role of proinflammatory
cytokines in the pathogenesis of depression is provided
by a study demonstrating that the concentration of
inflammatory cytokines correlates positively with the
severity of depressive symptoms,
23
while antidepress-
ive treatment and clinical improvement leads to
reduction of proinflammatory cytokine concentration
in patients with depression.
10
A meta-analysis of 22
studies evaluating the relationship between the efficacy
of antidepressant medication in the treatment of
depression and levels of inflammatory markers
showed that the use of antidepressive drugs (especially
serotonin-specific reuptake inhibitors) was associated
with decreased levels of IL-1βand IL-6.
24
It has been established that Proinflammatory cyto-
kines may contribute to the development and pro-
gression of depression through the following pathways:
1. Pathological activation of the immune response,
including the acute-phase reaction as well as changes
occurring early in response to tissue damage: this reac-
tion is manifested, among other things, with a sharp
increase in the production of many proteins, including
acute phase proteins: the C-reactive protein, alpha-1
acid glycoprotein, and α-chymotrypsin, together with
changes in their structure.
25,26
2. Changes in neurotransmitter systems: inflammatory
cytokines can cross the bloodbrain barrier, using
both the space with increased permeability and the
active transport principle. The migration of cytokines
to the CNS can trigger various psychopathological
changes,
27
i.a., due to the influence of changes in the
synthesis, reuptake, and metabolism of neurotransmit-
ters involved in the regulation of mood such as dopa-
mine, serotonin, or glutamate.
2830
It turns out that
cytokines can cause a decrease in the availability of
serotonin, with an increase in concentrations of neuro-
toxic tryptophan metabolites via 2,3-dioxygenase
indoleamine activation. Substances produced during
the catabolism of tryptophan (called TRYCATs)
may adversely affect the behaviour processes. For
example, kynurenine elicits anxiety and depressive
behaviour.
31,32
Toxic metabolites of tryptophan are also produced
under the influence of tryptophan 2,3-dioxygenase,
activated by cortisol,
33
whose concentration is often
increased in depression.
34
Dopamine is also a neuro-
transmitter whose concentration and availability is
reduced under the influence of cytokines in selective
areas of the brain.
35
Another process, also disrupted in depression, is
associated with the capacity of cytokines to increase
glutamate release, which consequently leads to an
increase in glutamate transmission, and finally results
in enhanced generation of free radicals.
An important consequence of the increased glutama-
tergic transduction is also reduced production of the
nerve growth factor (brain derived neurotrophic
factor (BDNF)). Increased glutamate neurotrans-
mission and BDNF reduced levels lead to changes in
neuronal plasticity.
36
3. The effect on the HPA axis and release of cortico-
tropin-releasing hormone (CRH) and adrenocortico-
tropic hormone (ACTH): proinflammatory cytokines
Karakuła-Juchnowicz et al. The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders
Nutritional Neuroscience 2014 VOL. 0NO. 02
intensify noradrenergic neurotransmission and
activate the HPA axis.
37
HPA hyperactivity has been
proposed as the neurobiological basis of major
depression.
38
It is well documented that patients with
major depressive disorder have elevated plasma corti-
sol levels as well as decreased sensitivity to external
dexamethasone and CRH.
39
4. The processes described above, expressed in immuno-
logical and glandular malfunctions and neurotrans-
mitters dysregulation, can lead to brain cell loss and
reduction in neurogenesis. According to the latest
views on the pathogenesis of depression, stress
factors can cause atrophy of hippocampal cells (as a
result of hypercortisolism caused by HPA axis hyper-
activity) and impairment of neurogenesis in predis-
posed subjects.
40
Although there is a growing body of evidence sup-
porting the importance of inflammation and
immune activation in at least part of the population
of depressive patients,
3,15,18,41
the role of inflam-
mation and inflammatory processes similar to those
described in cases of schizophrenia
42
requires further
investigation in depression as do potential issues
governing the integrity of the intestinal barrier via
sulphonation,
43
tight-junction (TJ) modulators,
44
the
contributing role of pathogenic agents,
42
and IgG
food hypersensitivity.
45
Leaky gut and IgG hypersensitivity
There is an increasing number of reports on the role
of the gastrointestinal tract in the pathogenesis of
depression.
46
One of the factors leading to a systemic
inflammatory response is increased intestinal per-
meability, also called leaky gut syndrome (LGS).
LGS is a dysfunction of the intestinal barrier,
resulting from damaged connections between entero-
cytes.
47
These connections intercellular tight junc-
tions (TJs) are protein complexes, with junctions
between enterocytes that are the structural basis for
the epithelium barrier.
48
They play two fundamental
roles, serving as: (1) a gate that regulates the passage
of ions, water, and other molecules through the para-
cellular route; and (2) a fence that blocks the lateral
diffusion within the plane of the membrane of lipids
and proteins, thereby maintaining the polarized distri-
bution of lipids and proteins between the apical and
basolateral plasma membrane domains.
49
TJs also
control the balance between the bodys immune toler-
ance and response to antigens. Proteins that make up
TJs zonulin, occludin, and claudins, modulate the
permeability of the described connections.
50,51
We
already know much about the structure of TJs, but
relatively little is known about their physiological
and pathophysiological modulation.
Zonulin is a precursor of acute phase protein-hapto-
globin 2. This protein controls paracellular
permeability through the epidermal growth factor
receptor
52
and protease-activated receptor (PAR)-
2.
44,53
The main role of zonulin is to regulate the flow
of molecules from the intestinal lumen by loosening
TJs.
44,54
It has been shown that overproduction of zonulin
can be triggered by exposure to bacteria,
53,55,56
drugs,
53
stress,
57
or foods.
53
For example, the dietary
protein gliadin, which is a class of proteins present in
wheat and several other cereals within the grass
genus Triticum, binds to the CXCR3 receptor,
leading to MyD88-dependent zonulin release and
increased intestinal permeability.
53,56,58
This, in turn,
may result in higher serum concentrations of the proin-
flammatory cytokines, free radicals, and others.
59
The
bowel dysfunction could, through a TJ opening,
induce immune activation which could contribute to
mitochondrial dysfunction and finally result in oxi-
dative stress.
It remains unclear whether gliadin-induced zonulin
activation is a specific reaction, that is whether
gliadin-dependent activation of the zonulin system
requires interaction of gliadin with a specific entero-
cyte receptor(s), or is a consequence of an unspecific
response.
60
A possible gliadin mechanism of action
may lead to a zonulin-mediated increase in actin poly-
merisation and intestinal permeability. Enterocytes
exposed to gliadin physiologically react by secreting
zonulin into the intestinal lumen. While in normal
intestinal tissues this secretion is self-limited in time,
the zonulin system in gut tissues of subjects with
coeliac disease (CD) is chronically upregulated,
leading to a sustained increase in intestinal
permeability to macromolecules, including gliadin,
from the lumen to the lamina propria, and ultimately
to intestinal permeability, immune, and autoimmune
disorders.
60
Increasing evidence points to possible con-
nections between autoimmune diseases, such as
coeliac disease or type 1 diabetes, and an earlier
increase in intestinal permeability, where, apart from
a genetic predisposition and environmental factors,
impairment of the intestinal barrier function must
occur.
61,62
The hypothesis about the dysregulation of
the zonulin release system, which triggers the disease
mechanism, immunological impairment of the gut
wall with increased intestinal permeability combined
with exposure to non-specific antigens, e.g. dietary or
bacterial factors may result in chronic inflammation
or autoimmune disease in genetically predisposed
subjects.
53,6163
Zonulin impact on intestinal permeability depends
on time of administration and dosage. These results
were confirmed independently in vivo in intestinal per-
meability assay, wherein zonulin induced a significant,
reversible increase in the gastric mucosal permeability
of the duodenum and small intestine.
44
Zonulin can
Karakuła-Juchnowicz et al. The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders
Nutritional Neuroscience 2014 VOL. 0NO. 03
be used as a biomarker of impaired gut barrier func-
tion for several autoimmune, neurodegenerative, and
tumour diseases and can be a potential therapeutic
target for the treatment of these devastating
conditions.
44
However, it should be stressed that other factors, for
instance commensal bacteria
64
or glycated and lipoxi-
dated proteins and peptides,
65
may also disturb the gut
immune homeostasis, leading to LGS, chronic
immunological overactivation, and low-grade inflam-
mation. Advanced glycation end products constitute
a group of heterogeneous compounds, whose high
concentrations in the body may be linked to activation
of a specific receptor called RAGE and substantially
exaggerate and prolong an inflammatory condition.
65
Production of similar compounds is facilitated by
foods undergoing high-temperature processing and
may adversely affect bowel receptors or exert an
impact on systemic inflammation.
65,66
The other proteins that play an important role
in regulating the intestinal barrier by modulating the
permeabilityof tight junctions are tight junction-associ-
ated MARVEL proteins (with occludin being the best
studied member of this protein family), junctional
adhesion molecules, and more than 20 members of
the claudin family.
67,68
It is likely that claudins interact
with many proteins.
69,70
Better understanding of these
interactions may provide insight into the entire cycle
of TJ regulation.
71
For example, the results of some
studies
72,73
demonstrate increased intestinal claudin
expression in response to milk protein components.
Occludin in turn was first identified as an integral mem-
brane protein connected with TJs in chickens
74
and in
mammals.
75
These proteins interact with the actin
cytoskeleton via TJ adaptor proteins like zonula occlu-
dens, which are membrane-associated guanylate kinase
inverted, and cingulin.
67
The factors aggravating TJs functioning are, i.a.,
psychological stress (increasing levels of CRH), proin-
flammatory cytokines, bacterial dysbiosis, nuclear
transcription factor NFKB (involved in the cellular
response to stimulistress, cytokines, free radicals,
antigens), oxidative stress, and others.
45,59
In their
study on mice, Riba et al.
76
found that stress from
maternal separation induces irritable bowel-like syn-
drome connected with increased paracellular intestinal
permeability and visceral hypersensitivity in adult
offspring, but also causes a specific IgG response to
soluble food antigens.
Selective permeability due to loss of intestinal
barrier is observed, i.a., in coeliac disease, inflamma-
tory bowel disease, obesity, atopic dermatitis, food
hypersensitivity, diabetes, sarcoidosis, neoplastic dis-
eases, cystic fibrosis, and also in autism.
5,77,78
There is a growing interest in the role of microbiota
in the maintenance of proper TJ functioning, the
braingut axis, and in the development of psychiatric
disorders.
79
The effect that intestinal bacteria has on
CNS and, consequently, psychiatric disorders are mul-
tidirectional, and is based primarily not only on cyto-
kine levels modulation, tryptophan metabolism, but
also on intestinal permeability.
45,80
Autochthonic bac-
teria are an important component in reducing levels of
proinflammatory cytokines and in maintaining intesti-
nal barrier continuity. This is the reason why bacterial
dysbiosis can lead to TJ unsealing. Increased intestinal
permeability allows bacterial lipopolysaccharides to
penetrate into the blood. In depression, significantly
elevated levels of IgM and IgA antibodies against
gram-negative enterobacteria lipopolysaccharides
were found.
5
This observation is very important
because metabolites of certain bacteria not only
adversely affect the functioning of the CNS but also
penetrate into the blood. A perfect example is the
study of Naseribafrouei et al.
81
They showed that the
Oscillibacter type strain, found in depression patients,
has a homolog of neurotransmitter GABA valeric
acid as its main metabolic end product.
81
This mech-
anism may contribute to the psychopathology of
depression.
The type of diet can also contribute to LGS develop-
ment, either by mechanical TJ damage or by having a
negative effect on microbiota balance. A study by
Drago et al.
56
shows that gliadin containing foods
may lead to increased gut permeability. Gliadin acti-
vates zonulin signalling irrespective of the genetic
expression of autoimmunity, which leads to increased
intestinal permeability to macromolecules.
56
Taken together, many factors contribute to LGS,
which is the reason why intestinal bacteria (autochtho-
nous microflora; microbiota) and incompletely
digested nutrients move from the intestinal lumen
into the blood. This condition leads to the activation
of the immune system,
82
which may initiate
production of specific IgG antibodies against nutri-
ents, and consequently, the development of food
hypersensitivity, which is delayed and IgG-depen-
dent.
83
Inflammation, emerging as a consequence of
this process, is chronically sustained by repeated con-
sumption of allergenic foods.
46,8486
The role of
specific IgG antibodies has been confirmed in coeliac
patients where IgG-dependent delayed reaction to
gluten occurs.
87,88
The delayed nature of the reaction is a considerable
diagnostic obstacle that makes it impossible for the
patient to identify the factor causing the allergy. This
results from the characteristics of IgG-dependent
responses. While IgE antibodies are responsible
for acute, immediately appearing allergic reactions,
IgG-dependent reactions take much longer to
develop.
89,90
These antibodies play a significant role
in shaping the bodys normal immune response.
Karakuła-Juchnowicz et al. The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders
Nutritional Neuroscience 2014 VOL. 0NO. 04
Specific IgG-food-antigen complexes activate the
complementary system and phagocytic migration,
degrading the immune complex.
91
The immune
system can be activated where the immune complex
is formed at the binding site of the receptor, e.g. in
the choroid plexus CNS, renal glomerular basement
membrane, blood vessel walls, etc. Production of
proinflammatory cytokines (IL-1, IL-6, and TNF-α),
proteolytic enzymes, and free radicals damaging
the surrounding tissue is observed. Sustained inflam-
mation can be an initiating factor in the development
of chronic diseases.
9193
As mentioned above, intesti-
nal barrier discontinuity resulting in elevated levels
of proinflammatory cytokines has been reported in
patients with depression.
It is worth mentioning that type II hypersensitivity
reaction (cytostaticcytotoxic reaction) does not lead
to the development of a chronic inflammation state
and, consequently, is not involved in depression devel-
opment and/or its maintenance. In this reaction,
IgM/IgG antibodies bind to the antigens and join
complement fractions. As a result, cytolysis of the
effector cell is observed.
Gluten sensitivity and depression
Only recently has coeliac disease been separated from
gluten sensitivity (non-coeliac gluten sensitivity,
NCGS) and gluten allergic reactions (IgE-mediated).
94
According to the consensus document developed in
2012, a spectrum of gluten-related disorders includes
three main forms of gluten reactions: allergic (e.g.
food allergy), autoimmune (e.g. coeliac disease, der-
matitis herpetiformis, and gluten ataxia), and possibly
immune-mediated (e.g. NCGS).
95,96
CD is a chronic immune-mediated enteropathy trig-
gered by gluten ingestion in subjects who have genetic
compatibility of the HLA DQ2 or DQ8 haplotype.
97
This disorder affects one percent of the general
population and is characterized by villous atrophy,
crypt hyperplasia, and increased intraepithelial
lymphocytes.
94,98
Classic CD manifestations (but only in 50 per cent
of patients) are severe diarrhoea and consequent
weight loss with failure to thrive due to severe intesti-
nal malabsorption. All the other cases are clinically
atypical, associated, for example, with anaemia, osteo-
porosis, musculoskeletal and neurological disorders,
endocrinopathies, or skin diseases.
99
NCGS is a relatively new term for conditions in
which symptoms are triggered by gluten ingestion, in
the absence of coeliac-specific antibodies and of classi-
cal coeliac villous atrophy, with a variable presence of
first generation anti-gliadin antibodies.
100
The Human Leukocyte Antigen (HLA) classifi-
cation is not particularly useful in the NCGS diagnosis
process (only 50% of positive DQ2/DQ8 outcomes)
and, moreover, anti-gliadin antibodies in the first gen-
eration of the IgG class tend to be positive in subjects
with gluten sensitivity.
96,101,102
Unfortunately, despite the fact that NCGS occurs
six times more frequently than CD, the majority of
research has not separated these two disorders.
103
Although there is some evidence connecting CD
with neurologic and psychiatric symptoms,
94
there
have been very few studies so far examining connec-
tions between NCGS and mental disorders. In the
mainstream of this trend, there is Fasanos group
research, focused on possible links between NCGS
and schizophrenia, identifying a biomarker and a sub-
sequent diagnostic tool for the condition of gluten sen-
sitivity and the role of the timing of gluten
introduction in CD pathogenesis in infants.
44
Biesiekierski
104
confirmed the existence of NCGS in
patients with irritable bowel syndrome in a random-
ized, double blind, placebo-controlled trial. The role
of intestinal barrier dysfunction was confirmed in
patients suffering from autism.
105
There is still a lack of studies on the importance of
leaky gut and IgG food allergy in the pathogenesis
of depression. Gluten sensitivity or intolerance has
been mentioned only in a few reports so far. For
example, Carta et al.
106
found that major depressive
disorder, dysthymic disorder, and adjustment dis-
orders were more common in a group of coeliac
patients as compared to the controls. Ludvigsson
et al.
107
achieved similar results with regard to
depression, but prevalence of CD in patients diag-
nosed with bipolar disorder was similar to the con-
trols. Ruuskanen et al.
108
in their research found that
the elderly population with gluten sensitivity was
more than twice as likely to have depression as com-
pared to the elderly sample without gluten sensitivity.
Carr,
95
in turn, described a case of an 11-year-old
girl who had been on a gluten-free diet since early
childhood due to health issues associated with wheat
consumption. At the age of 10 she had to consume a
wheat-containing diet for a week. After this short
period, her mood dropped suddenly, and she also
claimed that she had wanted to kill herself. Her
parents immediately changed her diet back to a strictly
gluten-free diet and after several days her mental state
improved significantly. In the pilot study, Peters
et al.
109
evidenced that even short-term gluten
exposure in patients with NCGS can cause symptoms
similar to depression.
The role of IgG-based elimination diet in the
therapy of depression
There is some evidence confirming the fact that the
quality of diet has an influence on leaky gut syndrome,
immune functioning and systemic inflammation in
depressive patients.
6
For example, whole grain foods
Karakuła-Juchnowicz et al. The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders
Nutritional Neuroscience 2014 VOL. 0NO. 05
include fibre and beta glucans that can probably
modulate immune functioning.
110
Other dietary immunomodulating factors of con-
siderable current interest to mental health researchers
include prebiotics and probiotics.
111
The effectiveness
of prebiotics in promoting the growth of strains of ben-
eficial bacteria, e.g. bifidobacteria in the gut, has been
demonstrated by some studies. Namely, it has been
argued that the use of prebiotics leads to a favourable
change in intestinal microbiology, reduces intestinal
inflammation, and alleviates symptoms, e.g. atopic
eczema, irritable bowel syndrome.
112,113
There are
more and more reports about the positive effect of
such strains of bacteria as Streptococcus thermophilus
or Lactobacillus acidophilus, and even probiotic
Escherichia coli Nissle 1917, which protect the intesti-
nal barrier against harmful factors by reducing the
symptoms of leaky gut.
114,115
Various potential immunomodulating factors
improving tightness of the intestinal barrier also
include amino acid L-glutamine, which has been con-
firmed by some studies demonstrating its positive
effect on intestinal enterocytes as well as its power to
reduce leaky gut.
115,116
Present in curry powder, curcumin is another com-
pound with a beneficial immunomodulating effect,
potentially reducing inflammatory condition and oxi-
dative stress related to the activityof tight junctions.
117
Tight junction damage between enterocytes leads
to increased intestinal permeability that causes
absorption of undigested proteins in small intestine
and higher levels of specific IgG antibodies as a
consequence.
86
The essential treatment in this case should be
implementation of an appropriate diet. An elimin-
ation-rotation diet may be a good choice in patients
with IgG food allergy in many diseases (e.g.
migraine
118,119
). This type of diet relies on elimination
of foods identified as causing allergic reactions and
rotation in taking a certain type of food (only for
one day and then a three-day interval). Such rotation
reduces the risk of occurrence hypersensitivity to
food products that have been well tolerated before.
120
Most publications connecting intestinal per-
meability with mental disorders refer to studies on
autism spectrum disorders. Demonstration of efficacy
of gluten and casein-free diets in patients suffering
from autism
105,121124
has resulted in attempts to use
this type of therapeutic proceedings in other mental
disorders, including depression. So far, studies on the
connection of a gluten-free diet and depression have
involved depressive subjects only with coeliac disease
(CD). For example, Corvaglia et al.
125
reported
several cases of patients with CD who had been unsuc-
cessfully treated with antidepressants and whose
depressive symptoms improved with a gluten-free
diet. Pynnonen et al.
126
also achieved similar results
in adolescents with CD. It may be assumed that elim-
ination of a factor causing inflammation (gluten) con-
tributes to mental health improvement also in patients
experiencing NCGS and depression. So far there have
been no studies that take into consideration a gluten-
free diet in depressive patients with NCGS.
Conclusion
The presented new hypothesis assumes existence of a
new potential pathway that may mediate the patho-
genesis of depression, which implies the existence of
subsequent developmental stages. Overproduction of
zonulin triggered by, e.g. gliadin
60
through activation
of the epidermal growth factor receptor and pro-
tease-activated receptor (PAR)-2 causes loosening of
the TJ barrier and an increase in permeability of the
gut wall (leaky gut). This results in a process allowing
larger molecules that would normally stay in the gut to
cross into the bloodstream and in induction of IgG-
dependent food sensitivity.
127,128
This condition
causes an increased immune response
93,129
and conse-
quently induces the release of proinflammatory cyto-
kines,
15
which in turn may lead to the development
of depressive symptoms.
130
In view of the foregoing considerations, it seems
advisable to assess the intestinal permeability using
as a marker, e.g. zonulin, occludin, and specific IgG
concentrations against selected nutritional com-
ponents in patients with depression. In the case of
increased IgG concentrations, the implementation of
an eliminationrotation diet may prove to be an effec-
tive method of reducing inflammation. It is necessary
to evaluate the concentration of all subclasses of
specific IgG (IgG 14) using validated research tools,
by a quantitative method.
This new paradigm in the pathogenesis of depressive
disorders linking leaky gut, IgG-dependent food sensi-
tivity, inflammation and depression is promising, but
still further studies are needed to confirm this theory.
Another field of interest, i.e. elimination diets in
depression treatment, requires well-designed clinical
trials to check their utility.
Disclaimer statements
Contributors All authors contributed equally.
Funding None.
Conflicts of interest None.
Ethics approval N/A
References
1 Wittchen HU. The burden of mood disorders. Science 2012;
338:15.
2 Puz
˙yn
´ski S. Choroby afektywne nawracaja˛ce. In: Puz
˙yn
´ski S,
Rybakowski J, Wciórka J, (ed.) Psychiatria tom 2, Psychiatria
Karakuła-Juchnowicz et al. The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders
Nutritional Neuroscience 2014 VOL. 0NO. 06
kliniczna. Wrocław: Elsevier Urban & Partner; 2012.
p. 30575.
3 Anisman H. Inflaming depression. J Psychiatry Neurosci 2011;
36:2915.
4 Elomaa AP, Niskanen L, Herzig KH, Viinamäki H, Hintikka J.
Elevated levels of serum IL-5 are associated with an increased
likelihood of major depressive disorder. BMC Psychiatry
2012;12:2.
5 Maes M, Kubera M, Leunis JC. The gut-brain barrier in major
depression: intestinal mucosal dysfunction with an increased
translocation of LPS from gram negative enterobacteria
(leaky gut) plays a role in the inflammatory pathophysiology
of depression. Neuro Endocrinol Lett 2008;29(1):11724.
6 Berk M, Williams LJ, Jacka FN, ONeil A, Pasco JA, Moylan
S, et al. So depression is an inflammatory disease, but where
does the inflammation come from? BMC Med 2013;11:200.
7 Smith RS. The macrophage theory of depression. Med
Hypotheses 1991;35:298306.
8Rys
´A, Miodek A, Szemraj P, Kocur J. Immunological and
endocrine aspects of pathogenesis of depression. Adv
Psychiatry Neurol 2007;16(4):3357.
9Goła˛b J, Jakóbisiak M, Lasek W. Immunology. Warszawa:
PWN; 2004. p. 198247
10 Ufnal M, Wolynczyk-Gmaj D. The brain and cytokines the
mutual origin of depression, obesity and cardiovascular dis-
eases? Adv Hyg Exp Med 2011;65:22835.
11 Refojo D, Liberman AC, Holsboer F, Arzt E. Transcription
factor-mediated molecular mechanisms involved in the func-
tional cross-talk between cytokines and glucocorticoids.
Immunol Cell Biol 2001;79(4):38594.
12 Liberman AC, Druker J, Perone MJ, Arzt E. Glucocorticoids in
the regulation of transcription factorsthat control cytokine syn-
thesis. Cytokine Growth Factor Rev 2007;18(12):4556.
13 Dantzer R, Wollman E, Vitkovic L, Yirmiya R. Cytokines and
depression: fortuitous or causative association? Mol. Psychiatry
1999;4:32832.
14 Howren MB, Lamkin DM, Suls J. Associations of depression
with C-reactive protein, IL-1, and IL-6: a meta-analysis.
Psychosom Med 2009;71:17186.
15 Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim
EK, et al. A meta-analysis of cytokines in major depression.
Biol Psychiatry 2010;67:44657.
16 Levine J, Barak Y, Chengappa KN, Rapoport A, Rebey M,
Barak V. Cerebrospinal cytokine levels in patients with acute
depression. Neuropsychobiology 1999;40(4):1716.
17 Lindqvist D, Janelidze S, Hagell P, Erhardt S, Samuelsson M,
Minthon L, et al. Interleukin-6 is elevated in the cerebrospinal
fluid of suicide attempters and relatedto symptom severity. Biol
Psychiatry 2009;66(3):28792.
18 Danner M, Kasl SV, Abramson JL, Vaccarino V. Association
between depression and elevated C-reactive protein.
Psychosom Med 2003;65(3):34756.
19 Pasco JA, Nicholson GC, Williams LJ, Jacka FN, Henry MJ,
Kotowicz MA, et al. Association of high-sensitivity C-reactive
protein with de novo major depression. Br J Psychiatry 2010;
197(5):3727.
20 Katon W, Lin EH, Kroenke K. The association of depression
and anxiety with medical symptom burden in patients with
chronic medical illness. Gen Hosp Psychiatry 2007;29:14755.
21 Loftis JM, Patterson AL, Wilhelm CJ, McNett H, Morasco BJ,
Huckans M. Vulnerability to somatic symptoms of depression
during interferon-alpha therapy for hepatitis C: a 16-week
prospective study. J Psychosom Res 2013;74(1):5763.
22 Asnis GM, De La Garza R. Interferon-induced depression in
chronic hepatitis C: a review of its prevalence, risk factors,
biology, and treatment approaches. J Clin Gastroenterol 2006;
40(4):32235.
23 Leonard BE, Myint A. Changes in the immune system in
depression and dementia: causal or coincidental effects?
Dialogues Clin Neurosci 2006;8:16374.
24 Hannestad J, DellaGioia N, Bloch M. The effect of anti-
depressant medication treatment on serum levels of inflamma-
tory cytokines: a meta-analysis. Neuropsychopharmacology
2011;36(12):24529.
25 Anisman H, Merali Z, Poulter MO, Hayley S. Cytokines as a
precipitant of depressive illness: animal and human studies.
Curr Pharm Des 2005;11(8):96372.
26 Lim W, Hong S, Nelesen R, Dimsdale JF. The association of
obesity, cytokine levels, and depressive symptoms with diverse
measures of fatigue in healthy subjects. Arch Intern Med
2005;165(8):9105.
27 Schaefer M, Schmidt F, Neumer R, Scholler G, Schwarz M.
Interferon-alpha, cytokines and possible implications for
mood disorders. Bipolar Disord 2002;4(Suppl 1):1113.
28 Khairova RA, Machado-Vieira R, Du J, Manji HK: A potential
role for pro-inflammatory cytokines in regulating synaptic plas-
ticity in major depressive disorder. Int J Neuropsychopharmacol
2009;12:56178.
29 Miller AH. Mechanisms of cytokine induced behavioral
changes: psychoneuroimmunology at the translational inter-
face. Norman Cousins Lecture. Brain Behav Immun 2009;23:
14958.
30 Raison CL, Borisov AS, Majer M, Drake DF, Pagnoni G,
Woolwine BJ. Activation of central nervous system inflamma-
tory pathways by interferon-alpha: relationship to monoamines
and depression. Biol Psychiatry 2009;65(4):296303.
31 Hartai Z, Klivenyi P, Janaky T, Penke B, Dux L, Vecsei L.
Kynurenine metabolism in multiple sclerosis. Acta Neurol
Scand 2005;112(2):936.
32 Oxenkrug GF. Metabolic syndrome, age-associated neuroendo-
crine disorders, and dysregulation of tryptophan-kynurenine
metabolism. Ann NY Acad Sci 2010;1199:114.
33 Maes M, Leonard BE, Myint AM, Kubera M, Verkerk R.
The new 5-HThypothesis of depression: cell-mediated
immune activation induces indoleamine 2,3-dioxygenase,
which leads to lower plasma tryptophan and an increased
synthesis of detrimental tryptophan catabolites (TRYCATs),
both of which contribute to the onset of depression. Prog
Neuropsychopharmacol Biol Psychiatry 2011;35:70221.
34 Dienes KA, Hazel NA, Hammen CL. Cortisol secretion in
depressed, and at-risk adults. Psychoneuroendocrinology
2013;38(6):92740.
35 Kitagami T, Yamada K, Miura H, Hashimoto R, Nabeshima
T, Ohta T. Mechanism of systemically injected interferon-
alpha impeding monoamine biosynthesis in rats: role of nitric
oxide as a signal crossing the blood-brain barrier. Brain Res
2003;978(12):10414.
36 Martin JL, Finsterwald C. Cooperation between BDNF and
glutamate in the regulation of synaptic transmission and neur-
onal development. Commun Integr Biol 2011;4(1):146.
37 Anisman H. Cascading effects of stressors and inflammatory
immune system activation: implications for major depressive
disorder. J Psychiatry Neurosci 2009;34:420.
38 Dinan TG. Glucocorticoids and genesis of depressive illness a
psychological approach. Br J Psychiatry 1994;164:36571.
39 Hasler G, Drevets WC, Manji HK, Charney DS. Discovering
endophenotypes for major depression. Neuropsychopharma-
cology 2004;29(10):176581.
40 Remlinger-MolendaA, Wójciak P, Michalak M, Rybakowski J.
Activity of selected cytokines in bipolar patients during manic
and depressive episodes. Psychiatr Pol 2012;46(4):599611.
41 Raison LR, Miller AH. Is depression an inflammatory dis-
order? Curr Psychiatry Rep 2011;13:46775.
42 Severance EG, Alaedini A, Yang S, Halling M, Gressitt KL,
Stallings CR, et al. Gastrointestinal inflammation and associ-
ated immune activation in schizophrenia. Schizophr Res 2012;
138(1):4853.
43 Bowling FG, Heussler HS, McWhinney A, Dawson PA.
Plasma and urinary sulfate determination in a cohort with
autism. Biochem Genet 2013;51(12):14753.
44 Fasano A. Leaky gut and autoimmune diseases. Clin. Rev.
Allergy Immunol 2012;42:718.
45 Rudzki L, Frank M, Szulc A, Gałe˛cka M, Szachta P, Barwinek
D. From gut to depression the role of intestinal barrier discon-
tinuity and activation of the immune system in the depression
inflammatory hypothesis. Neuropsychiatr Neuropsychology
2012;7(2):7684.
46 Liu Z, Li N, Neu J. Tight junctions, leaky intestines, and pedi-
atric diseases. Acta Paediatr 2005;94(4):38693.
47 Odenwald MA, Turner JR. Intestinal permeability defects: is it
time to treat? Clin Gastroenterol Hepatol 2013;11(9):107583.
48 Jiang Y, Guo C, Zhang D, Zhang J, Wang X, Geng C. The
altered tight junctions: an important gateway of bacterial trans-
location in cachexia patients with advanced gastric cancer. J
Interferon Cytokine Res 2014;34(7):51825.
49 Cereijido M, Valdés J, Shoshani L, Contreras RG. Role of tight
junctions in establishing and maintaining cell polarity. Annu
Rev Physiol 1998;60:16177.
Karakuła-Juchnowicz et al. The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders
Nutritional Neuroscience 2014 VOL. 0NO. 07
50 Kojima T, Kokai Y, Chiba H, Yamamoto M, Mochizuki Y,
Sawada N. Cx32 but not Cx26 is associated with tight junctions
in primary cultures of rat hepatocytes. Exp Cell Res 2001;263:
193201.
51 Singh D, Solan JL, Taffet SM, Javier R, Lampe PD. Connexin
43 interacts with zona occludens-1 and -2 proteins in a cell cycle
stage-specific manner. J Biol Chem 2005;280:3041621.
52 Tripathi A, Lammers KM, Goldblum S, Shea-Donohue T,
Netzel-Arnett S, Buzza MS, et al. Identification of human
zonulin, a physiological modulator of tight junctions, as pre-
haptoglobin-2. Proc Natl Acad Sci USA 2009;106(39):
16799804.
53 Fasano A. Zonulin and its regulation of intestinal barrier func-
tion: the biological door to inflammation, autoimmunity, and
cancer. Physiol. Rev 2011;91:15175.
54 Pabijasz D. Ocena przepuszczalnos
´ci jelitowej na podstawie
ste˛z
˙enia zonuliny u dzieci z nieswoistymi zapaleniami jelit.
Poste˛py Nauk Medycznych 2013;5:34650.
55 El Asmar R, Panigrahi P, Bamford P, Berti I, Not T, Coppa GV.
Host-dependent zonulin secretion causes the impairment of the
small intestine barrier function after bacterial exposure.
Gastroenterology 2002;123(5):160715.
56 Drago S, El Asmar R, Di Pierro M, Grazia ClementeM, Tripathi
A, Sapone A, et al. Gliadin, zonulin and gut permeability: effects
on celiac and non-celiac intestinal mucosa and intestinal cell
lines. Scand J Gastroenterol 2006;41(4):40819.
57 Hart A, Kamm MA. Review article: mechanisms of initiation
and perpetuation of gut inflammation by stress. Aliment
Pharmacol Ther 2002;16(12):201728.
58 Lammers KM, Lu R, Brownley J, Lu B, Gerard C, Thomas K,
et al. Gliadin induces an increase in intestinal permeability and
zonulin release by binding to the chemokine receptor CXCR3.
Gastroenterology 2008;135(1):194204.
59 Kiliaan AJ, Saunders PR, Bijlsma PB, Berin MC, Taminiau
JA, Groot JA, et al. Stress stimulates transepithelial macromol-
ecular uptake in rat jejunum. Am J Physiol 1998;275(5 Pt1):
G103744.
60 Clemente MG, De Virgiliis S, Kang JS, Macatagney R, Musu
MP, Di Pierro MR, et al. Early effects of gliadin on enterocyte
intracellular signalling involved in intestinal barrier function.
Gut 2003;52:21823.
61 Sapone A, de Magistris L, Pietzak M, Clemente MG, Tripathi
A, Cucca F, et al. Zonulin upregulation is associated with
increased gut permeability in subjects with type 1 diabetes
and their relatives. Diabetes 2006;55:14439.
62 Visser J, Rozing J, Sapone A, Lammers K, Fasano A. Tight
junctions, intestinal permeability, and autoimmunity: celiac
disease and type 1 diabetes paradigms. Ann NY Acad Sci
2009;1165:195205.
63 Drago S, Congia M, Fasano A. Early effects of gliadin on
enterocyte intracellular signaling involved in intestinal barrier
function. Gut 2003;52:21823.
64 MacDonald TT, Monteleone G. Immunity, inflammation, and
allergy in the gut. Science 2005;307(5717):19205.
65 Bengmark S. Advanced glycation and lipoxidation end pro-
ducts amplifiers of inflammation: the role of food. J
Parenter Enteral Nutr 2007;31:43040.
66 Uribarri J, Cai W, Sandu O, Peppa M, Goldberg T, Vlassara H.
Diet-derived advanced glycation end products are major contri-
butors to the bodys AGE pool and induce inflammation in
healthy subjects. Ann NY Acad Sci 2005;1043:4616.
67 Shen L, Weber CR, Raleigh DR, Yu D, Turner JR. Tight junc-
tion pore and leak pathways: a dynamic duo. Annu Rev Physiol
2012;73:283309.
68 Betanzos A, Javier-Reyna R, García-Rivera G, Bañuelos C,
González-Mariscal L, Schnoor M, et al. The EhCPADH112
complex of Entamoeba histolytica interacts with tight junction
proteins occludin and claudin-1 to produce epithelial damage.
PLoS One 2013;8(6):e65100.
69 Kotler BM, Kerstetter JE, Insogna KL. Claudins, dietary milk
proteins, and intestinal barrier regulation. Nutr Rev 2013;71(1):
605.
70 Günzel D, Fromm M. Claudins and other tight junction
proteins. Compr Physiol 2012;2(3):181952.
71 Van Itallie CM, Anderson JM. Claudin interactions in and out
of the tight junction. Tissue Barriers 2013;1(3):e25247.
72 Ozawa T, Miyata M, Nishimura M, Ando T, Ouyang Y, Ohba
T, et al. Transforming growth factor-beta activity in commer-
cially available pasteurized cow milk provides protection
against inflammation in mice. J Nutr 2009;139:6975.
73 Sprong RC, Schonewille AJ, van der Meer R. Dietary cheese
whey protein protects rats against mild dextran sulfate
sodium-induced colitis: role of mucin and microbiota. J Dairy
Sci 2010;93:136471.
74 Furuse M, Hirase T, Itoh M, Nagafuchi A, Yonemura S,
Tsukita S, et al. Occludin: a novel integral membrane protein
localizing at tight junctions. J Cell Biol 1993;123(6 Pt 2):
177788.
75 Saitou M, Ando-Akatsuka Y, Itoh M, Furuse M, Inazawa J,
Fujimoto K, et al. Mammalian occludin in epithelial cells:
its expression and subcellular distribution. Eur J Cell Biol
1997;73(3):22231.
76 Riba A, Cartier Ch, Bacquie V, Lencina C, Mallet V, Harkat
Ch. Early maternal separation in mice triggered a specific
IgG response against soluble food antigens. Gastroenterology
2013;144(5):S717.
77 Bjarnason I, MacPherson A, Hollander D. Intestinal per-
meability: an overview. Gastroenterology 1995;108(5):156681.
78 Teixeira TF, Collado MC, Ferreira CL, Bressan J, Peluzio ,
Mdo C. Potential mechanisms for the emerging link between
obesity and increased intestinal permeability. Nutr Res 2012;
32(9):63747.
79 Alonso C, Vicario M, Pigrau M, Lobo B, Santos J. Intestinal
barrier function and the brain-gut axis. In: Lyte M, Cryan JF,
editors. Microbial endocrinology: the microbiota-gut-brain
axis in health and disease. New York: Springer; 2014. pp.
73113.
80 Moloney RD, Desbonnet L, Clarke G, Dinan TG, Cryan JF.
The microbiome: stress, health and disease. Mamm Genome
2014;25(12):4974.
81 Naseribafrouei A, Hestad K, Avershina E, Sekelja M,
Linløkken A, Wilson R, et al. Correlation between the human
fecal microbiota and depression. Neurogastroenterol Motil
2014;26(8):115562.
82 Drisko J, Bischoff B, Hall M, McCallum R. Treating irritable
bowel syndrome with a food elimination diet followed by
food challenge and probiotics. J Am Coll Nutr 2006;25(6):
51422.
83 Johansson SG, Bieber T, Dahl R, Friedmann PS, Lanier BQ,
Lockey RF, et al. Revised nomenclature for allergy for global
use: report of the nomenclature review committee of the
world allergy organization, October 2003. J Allergy Clin
Immunol 2004;113(5):8326.
84 Atkinson W, Sheldon TA, Shaath N, Whorwell PJ. Food
elimination based on IgG antibodies in irritable bowel syn-
drome: a randomised controlled trial. Gut 2004;53(10):145964.
85 Bentz S, Hausmann M, Piberger H, Kellermeier S, Paul S, Held
L, et al. Clinical relevance of IgG antibodies against ood
antigens in Crohns disease: a double-blind cross-over diet inter-
vention study. Digestion 2010;81(4):25264.
86 Frank M, Ignys
´I, Gałe˛cka M, Szachta P. IgG-dependent food
allergy and its role in selected diseases. Pediatr Pol 2012;88(3):
2527.
87 OFarrelly C, Kelly J, Hekkens W. Alpha gliadin antibody
levels: a serological test for coeliac disease. Br Med J (Clin
Res Ed) 1983;286:200710.
88 Beyazit Y, Sayilir A, Tas A, Kekilli M. IgA and IgG anti-
body testing for coeliac disease. Eur J Intern Med 2010;
21(5):4678.
89 Czarnobilska E, Obtułowicz K, Wsołek K. Type IV of hyper-
sensitivity and its subtypes. Przegl Lek 2007;64(78):5068.
90 Zuo XL, Li YQ, Li WJ, Guo YT, Lu XF, Li JM, et al.
Alterations of food antigen-specific serum immunoglobulins
G and E antibodies in patients with irritable bowel syn-
drome and functional dyspepsia. Clin Exp Allergy 2007;
37:82330.
91 Owen J, Punt J, Stranford S. Kuby Immunology. 7th ed.
New York: WH Freeman and Co.; 2013.
92 Chavez AM, Menconi MJ, Hodin RA. Cytokine-induced
intestinal epithelial hyperpermeability: role of nitric oxide.
Crit Care Med 1999;27(10):224651.
93 Ye D, Ma I, Ma TY. Molecular mechanism of tumor necrosis
factor modulation. Am J Physiol Gastrointest Liver Physiol
2006;290:G496G504.
94 Jackson JR, Eaton WW, Cascella NG, Fasano A, Kelly DL.
Neurologic and psychiatric manifestations of celiac disease
and gluten sensitivity. Psychiatr Q 2012;83:91102.
95 Carr AC. Depressed mood associated with gluten sensitivity
resolution of symptoms with a gluten-free diet. N Z Med J
2012;125(1366):812.
Karakuła-Juchnowicz et al. The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders
Nutritional Neuroscience 2014 VOL. 0NO. 08
96 Sapone A, Bai JC, Ciacci C, Dolinsek J, Green PH,
Hadjivassiliou M, et al. Spectrum of gluten-related disorders:
consensus on new nomenclature and classification. BMC Med
2012;10:13.
97 Leonard MM, Vasagar B. US perspective on gluten-related dis-
eases. Clin Exp Gastroenterol 2014;7:2537.
98 Fasano A, Catassi C. Current approaches to diagnosis and
treatment of celiac disease: an evolving spectrum.
Gastroenterology 2001;120:63651.
99 Schuppan D, Zimmer KP. The diagnosis and treatment of
celiac disease. Dtsch Arztebl Int 2013;110(49):83546.
100 Catassi C, Bai JC, Bonaz B, Bouma G, Calabrò A, Carroccio
A, et al. Non-Celiac gluten sensitivity: the new frontier of
gluten related disorders. Nutrients 2013;5(10):383953.
101 Sapone A. Gluten sensitivity: definition and diagnostic process.
Forum 2011;3:46.
102 Catassi C. A new pathology is born: gluten sensitivity. Forum
2011;3:13.
103 Hadjivassiliou M, Grunewald RA, Davies-Jones GA. Gluten
sensitivity as a neurological illness. J Neurol Neurosurg
Psychiatry 2002;72:5603.
104 Biesiekierski JR, Newnham ED, Irving PM, Barrett JS, Haines
M, Doecke JD, et al. Gluten causes gastrointestinal symptoms
in subjects without celiac disease: a double-blind randomized
placebo-controlled trial. Am J Gastroenterol 2011;106(3):
50814.
105 De Magistris L, Familiari V, Pascotto A, Sapone A, Frolli A,
Iardino P, et al. Alterations of the intestinal barrier in patients
with autism spectrum disorders and in their first-degree rela-
tives. J Pediatr Gastroenterol Nutr 2010;51(4):41824.
106 Carta MG, Hardoy MC, Boi MF, Mariotti S, Carpiniello B,
Usai P. association between panic disorder, major depressive
disorder and celiac disease: a possible role of thyroid autoim-
munity. J Psychosom Res 2002;53:78993.
107 Ludvigsson JF, Reutfors J, Osby U, Ekbom A, Montgomery
SM. Coeliac disease and risk of mood disorders a general
population-based cohort study. J Affect Disord 2007;99:11726.
108 Ruuskanen A, Kaukinen K, Collin P, Huhtala H, Valve R,
Maki M, et al. Positive serum antigliadin antibodies without
celiac disease in the elderly population: does it matter? Scand
J Gastroenterol 2010;45:1197202.
109 Peters SL, Biesiekierski JR, Yelland GW, Muir JG, Gibson PR.
Randomised clinical trial: gluten may cause depression in sub-
jects with non-coeliac gluten sensitivity an exploratory clinical
study. Aliment Pharmacol Ther 2014;39(10):110412.
110 Volman JJ, Ramakers JD, Plat J. Dietary modulation of
immune function by beta-glucans. Physiol Behav 2008;94:
27684.
111 Bested AC, Logan AC, Selhub EM. Intestinal microbiota,
probiotics and mental health: from Metchnikoff to modern
advances: part II-contemporary contextual research. Gut
Pathog 2013;5(3):114.
112 Cani PD, Delzenne NM. Interplay between obesity and associ-
ated metabolic disorders: new insights into the gut microbiota.
Curr Opin Pharmacol 2009;9:73743.
113 Roberfroid M, Gibson GR, Hoyles L, McCartney AL, Rastall
R, Rowland I, et al. Prebiotic effects: metabolic and health
benefits. Br J Nutr 2010;104(Suppl 2):S163.
114 Ukena SN, Singh A, Dringenberg U, Engelhardt R, Seidler U,
Hansen W, et al. Probiotic Escherichia coli Nissle 1917 inhibits
leaky gut by enhancing mucosal integrity. PLoS One 2007;2:
e1308.
115 Rapin JR, Wiernsperger N. Possible links between intestinal
permeablity and food processing: a potential therapeutic
niche for glutamine. Clinics (Sao Paulo) 2010;65(6):63543.
116 Ulluwishewa D, Anderson RC, McNabb WC, Moughan PJ,
Wells JM, Roy NC. Regulation of tight junction permeability
by intestinal bacteria and dietary components. J Nutr 2011;
141(5):76976.
117 Kosin
´ska A, Andlauer W. Modulation of tight junction integ-
rity by food components. Food Res Int 2013;54(1):95160.
118 Alpay K, Ertas M, Orhan EK, Üstay DK, Lieners C, Baykan
B. Diet restriction in migraine, based on IgG against foods: a
clinical double-blind, randomised, cross-over trial.
Cephalalgia 2010;30(7):82937.
119 Aydinlar EI, Dikmen PY, Tiftikci A, Saruc M, Aksu M,
Gunsoy HG, et al. IgG-based elimination diet in migraine
plus irritable bowel syndrome. Headache 2013;53(3):51425.
120 Szachta P, Frank M, Gałecka M, Ignys
´I. Gastrointestinal tract
disorders and nutritional therapy in children with autism spec-
trum disorders. Pediatr Pol 2014;89:26976.
121 Jyonouchi H, Geng L, Ruby A, Zimmerman-Bier B.
Dysregulated innate immune responses in young children with
autism spectrum disorders: their relationship to gastrointestinal
symptoms and dietary intervention. Neuropsychobiology 2005;
51(2):7785.
122 Knivsberg AM, Reichelt KL, Hoien T, Nodland M. A random-
ised, controlled study of dietary intervention in autistic
syndromes. Nutr Neurosci 2002;5(4):25161.
123 Millward C, Ferriter M, Calver S, Connell-Jones G. (2008).
Gluten- and casein-free diets for autistic spectrum disorder.
Cochrane Database Syst Rev. 2004;(2):CD003498.
124 Elder JH, Shankar M, Shuster J, Theriaque D, Burns S, Sherrill
L. The gluten-free, casein-free diet in autism: results of a pre-
liminary double blind clinical trial. J Autism Dev Disord
2006;36(3):41320.
125 Corvaglia L, Catamo R, Pepe G, Lazzari R, Corvaglia E.
Depression in adult untreated celiac subjects: diagnosis by the
pediatrician. Am J Gastroenterol 1999;94(3):83943.
126 Pynnönen PA, Isometsa ET, Verkasalo MA, Kähkönen SA,
Sipilä I, Savilahti E, et al. Gluten-free diet may alleviate
depressive and behavioural symptoms in adolescents with
coeliac disease: a prospective follow-up case-series study.
BMC Psychiatry 2005;5:14.
127 Kleinman RE, Walker AW. Antigen processing and uptake
from the intestinal tract. Clin Rev Allergy 1984;2:2537.
128 Saavedra-Delgado AM, Metcalfe DD. Interactions between
food antigens and the immune system in the pathogenesis of
gastrointestinal diseases. Ann Allergy 1985;55(5):694702.
129 Al-Sadi RM, Ma TY. IL-1beta causes an increase in intestinal
epithelial tight junction permeability. J Immunol 2007;178:
46419.
130 Lucas M, Chocano-Bedoya P, Shulze MB, Mirzaei F, OReilly
ÉJ, Okereke OI, et al. Inflammatory dietary pattern and risk of
depression among women. Brain Behav Immun 2014;36:
4653.
Karakuła-Juchnowicz et al. The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders
Nutritional Neuroscience 2014 VOL. 0NO. 09
... 4 Quindi, quello dell'intolleranza alimentare appare come un problema significativo per gli uomini moderni e può essere la causa di molti problemi di salute, come carenza cronica di ferro, rinite cronica, obesità, sindrome del colon irritabile, eczema, ipertensione, diabete di tipo II, emicrania, stanchezza cronica, sindrome da malassorbimento, ipotiroidismo, depressione e asma. 1,[7][8][9][10][11][12][13][14][15][16] Tuttavia, è ancora difficile da riconoscere. ...
... 4 Therefore, food intolerance appears to be a significant problem of modern humans and can be the cause of many health problems, such as chronic iron deficiency, chronic rhinitis, obesity, irritable bowel syndrome, eczema, hypertension, diabetes type II, migraine, chronic fatigue, malabsorption syndrome, hypothyroidism, depression, asthma. 1,[7][8][9][10][11][12][13][14][15][16] Yet it is still difficult to be recognized. ...
... 9.6 to 16.7%, P=0.0008) without significant difference Discussione L'effetto positivo della dieta di eliminazione in base agli anticorpi IgG è stato precedentemente osservato in emicrania, sindrome del colon irritabile, disturbi depressivi e altre condizioni. 1,[7][8][9][10][11][12][13][14][15][16] Tuttavia, sembra che questo sia il primo studio ad aver esaminato gli effetti di tale dieta basata sugli anticorpi IgG sulla prestazione sportiva e i sintomi GI negli atleti professionisti. La disfunzione gastrointestinale viene segnalata come comune (15-30%) negli atleti di endurance 30 e può essere collegata all'intolleranza alimentare. ...
... There are already findings in the literature demonstrating the relationship between the gut and the brain [13,14], especially in the context of subclinical inflammation caused by food hypersensitivity [15]. However, the literature still seems to be very scarce when it comes to studies that demonstrate an association between food and the quality of energy generation represented in brain electrical activity, especially in individuals with food incompatibility (food intolerance, allergy, or hypersensitivity). ...
... To consider the criterion of subclinical inflammation, from the total leukocytes above 6000, this study considered several findings [13][14][15]21,37] who showed consensus regarding values, on average, above 6000 mm 3 of total leukocytes as a strong indicator of subclinical inflammation. ...
Article
Background: There are findings demonstrating the association between the functional neurometry-FN exam and blood biomarkers. Objective: The association between FN-exam, biomarkers indicating subclinical inflammation and amplitude of Delta (2-4 Hz) and Theta (4-8 Hz) in ADHD was evaluated. Method: FN-assessments, blood tests and quantitative EEG (QEEG) were performed. The groups were: control = 32 and ADHD = 61. FN investigated: anxiety control-AC [Expected value ≥ 75%], Baroreflex index-BRI [≥ 90%], hemodynamics-HD [≤ 10%] and physiological response-PR [31.5º to 32.5º]. The biomarkers were: total leukocytes-TL, ferritin, C-reactive protein-CRP, fibrinogen, 25-hydroxy-vitamin-D3, IgE, expressed in frequencies and percentages, analyzed by the chi-square, and by the Mann-Whitney QEEG. All results were expressed as Median (Minimum-Maximum), p ≤ 0.05*. Results: The ADHD presented: AC = 37.12 (9.00-75.18), BRI = 87.28 (67.33-93.66), HD = 17.66 (12.11-32.50) and PR = 26.10 (21.00-29.00) with the highest frequencies of the biomarkers: TL > 6000 [44/61, (81.19%)], eosinophils > 7: [54/61, (88.52%)], neutrophils > 55: [40/61, (65.57%)], 25-hydroxyvitamin-D3 < 40 [47/61, (77.04%)] and IgE > 200 [20/61, (32.78%)], compared to controls: AC = 80.44 (68.00-85.00); BRI = 92.00 (88.00-95.00); HD = 9.00 (08.00-10.00); PR = 32.00 (31.00-32.50); TL > 6000 [12/32, (37.50%)], eosinophils > 7: [7/32, (21.87%)], neutrophils > 55: [9/32, (28.12%)], 25-hydroxyvitamin-D3 < 40 [11/32, (34.37%)] and IgE > 200 [3/32, (9.37%)]. There was an increase in the amplitudes of delta (F7 = 19u, F3 = 16.90u, Fp1 = 20.20u, Fz = 18.50u, Fp2 = 20.10u, F4 = 16.80u and F8 = 18.80u) and theta (F7 = 27.7u, F3 = 29.3u, Fp1 = 26.5u, Fz = 29, 7u, Fp2 = 23.7u, F4 = 29.7u and F8 = 26.7u) in the ADHD, compared to the respective controls (F7 = 10.90u, F3 = 11.10u, Fp1 = 10.30u, Fz = 9.25u, Fp2 = 13u, F4 = 9.95u and F8 = 10.10u) and (F7 = 22.65u, F3 = 23.45u, Fp1 = 23.55u, Fz = 24.5u, Fp2 = 22, 1u, F4 = 23.55u and F8 = 22u). Conclusion: The functional neurometry exam indicated low anxiety control and functional oxygen in patients with ADHD due to adrenal insufficiency and/or liver overload, low baroreflex index, hemodynamics with reduced blood flow velocity and/or low temperature in the proximal annular phalanx, indicating possible food incompatibility. Subclinical inflammation caused by allergenic and inflammatory foods was also observed and, finally, high amplitudes of delta and theta waves were observed in patients with ADHD, which seems to indicate a consequence of subclinical inflammation and low functional oxygen at the mitochondrial level of the brain. Thus, the NF test, blood biomarkers indicative of subclinical inflammation, and high results of delta and theta brain wave amplitudes, when associated with the clinic, can help in the diagnosis and treatment of ADHD. Abstract Citation: Murilo Toledo Calafange., et al.
... Other leaky gut biomarkers, such as interleukin (IL)− 6, C reactive protein (CRP), zonulin (a tight-junction modulator), LPS binding protein, and intestinal fatty acid binding protein (which is released upon gut mucosal damage) have also been associated with depression in independent studies [51][52][53]. These events, together with the notion that an increased enterobacterial translocation triggers the activation of oxidative and nitrosative pathways [50,54], lead to the establishment of an immune-inflammatory status typically found in depression [55,56]. Indeed, it should be kept in mind that the interplay between bacterial species and the gut mucosal immune cells is crucial to maintain the correct balance within the mucosal immune system [57]. ...
Article
Full-text available
Depression is the most prevalent mental disorder in the world associated with huge socioeconomic consequences. While depressive-related symptoms are well known, the molecular mechanisms underlying disease pathophysiology and progression remain largely unknown. The gut microbiota (GM) is emerging as a key regulator of the central nervous system homeostasis by exerting fundamental immune and metabolic functions. In turn, the brain influences the intestinal microbial composition through neuroendocrine signals, within the so-called gut microbiota-brain axis. The balance of this bidirectional crosstalk is important to ensure neurogenesis, preserve the integrity of the blood-brain barrier and avoid neuroinflammation. Conversely, dysbiosis and gut permeability negatively affect brain development, behavior, and cognition. Furthermore, although not fully defined yet, changes in the GM composition in depressed patients are reported to influence the pharmacokinetics of common antidepressants by affecting their absorption, metabolism, and activity. Similarly, neuropsychiatric drugs may shape in turn the GM with an impact on the efficacy and toxicity of the pharmacological intervention itself. Consequently, strategies aimed at re-establishing the correct homeostatic gut balance (i.e., prebiotics, probiotics, fecal microbiota transplantation, and dietary interventions) represent an innovative approach to improve the pharmacotherapy of depression. Among these, probiotics and the Mediterranean diet, alone or in combination with the standard of care, hold promise for clinical application. Therefore, the disclosure of the intricate network between GM and depression will give precious insights for innovative diagnostic and therapeutic approaches towards depression, with profound implications for drug development and clinical practice.
... At present, the effects of food-specific IgG on human health remain controversial. Numerous studies have suggested that food-specific IgG is involved in the development and progression of specific diseases, such as inflammatory bowel disease [1], irritable bowel syndrome [2], migraine [3,4] and mental disease [5][6][7]. The symptoms of these disease can be relieved by food-specific IgG-based diet recommendations [8][9][10][11][12][13]. ...
Article
Full-text available
Background Although the association of food-specific IgG with the development and progression of specific diseases was shown by many studies, it is also present in the population without clinical symptoms. However, the association between food-specific IgG and physical examination outcomes in healthy people has not been studied yet. Methods An asymptomatic physical examination cohort (APEC) was selected according to the inclusion and exclusion criteria, the physical examination data were compared between IgG positive and IgG negative groups, and their odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using multivariable logistic regression. Results The data of 28,292 subjects were included in the analysis. The overall IgG positive rate was up to 52.30%, mostly with mild to moderate IgG positivity. The multivariable Logistic regression showed the prevalence of hypertriglyceridemia, abnormal fasting blood glucose and overweight was lower in the IgG (+) positive group (OR 0.87, 95% CI 0.83–0.92; OR 0.93, 95% CI 0.87–0.99; OR 0.92, 95% CI 0.87–0.96) but there was a higher prevalence of thyroid disease (OR 1.09, 95% CI 1.04–1.15). Conclusion Food-specific IgG positivity was widespread in the APEC and was associated with lower prevalence of hypertriglyceridemia, abnormal fasting blood glucose and overweight. The underlying physiological mechanism merits further study.
... At present, the role of food-specific IgG in the health of human body remains controversial. Numerous studies have suggested that food-specific IgG is involved in the development and progression of some diseases, such as inflammatory bowel disease [1][2][3][4], irritable bowel syndrome [5], migraine [6,7]and mental disease [8][9][10][11], of which the symptoms can be relieved by food-specific IgG-based diet recommendations [12][13][14][15][16][17]. IgG antibody can form an immune complex with allergens in foods and thus induce the body's mild inflammatory reactions [18] that are manifested as various system symptoms and diseases. ...
Preprint
Full-text available
Background: Although food-specific IgG is associated with the development and progression of some diseases as shown by many studies, it is also present in the population without clinical symptoms. However, the damage of the immune response it evokes to the health of such population has not been studied yet. Methods: The asymptomatic physical examination population (APEP) was selected according to the inclusion and exclusion criteria, and the physical examination data were collected. The subjects were divided into IgG-positive group (IgG-po group) and IgG-negative group (IgG-neg group). The hematologic and imaging examination results were compared between the two groups, and their odds ratios (OR) and 95% confidence interval (95%CI) were calculated using binary logistic regression to determine the relationship between food-specific IgG and different health outcomes. Results: The data of 28,292 subjects were included in the analysis of the study. The overall IgG positive rate was up to 52.3%, with mild to moderate IgG positive predominantly. Compared with IgG negative group, IgG positive group was associated with a decrease in the risks of hypertriglyceridemia, abnormal fasting blood glucose and overweight(OR=0.87,95%:CI:0.83-0.92; OR=0.93,95%CI:0.87-0.99; OR=0.92,95%CI:0.87-0.96) and with an increase in the incidence rate of thyroid nodule (OR=1.09,95%CI:1.04-1.15). Conclusion: Food-specific IgG is generally present in APEP and has an unique distribution profile, which plays a positive role in maintaining normal body weight and metabolic indicators(triglyceride(TG) and fasting blood glucose(FBG)), without influencing the nutritional status. The physiological mechanism is worth further study.
... [17] According to a study conducted in 2017, IgG food hypersensitivity may lead to systemic inflammation and may be a triggering factor for the development of MDD. [18] The undigested food compounds, which will normally be broken down in the intestine, and enters the bloodstream and the epitopes merge with food IgG antibodies to form immune complexes. This triggers the immune inflammatory cascade. ...
Article
Full-text available
Food allergies are the hypersensitivity reactions of our body against food by means of immune system. They should not be confused with reactions known as “food intolerance” where an allergen and hypersensitivity reaction are not involved. Food allergy can affect many organs and systems in the body. Itching, burning, redness, urticaria (hives), nasal obstruction, sneezing, watery eyes and anaphylaxis are the symptoms of general reaction. Contrary to popular belief, food allergy is a problem not only for children and adolescents but also for adults. The question of whether these reactions affecting many organs and systems is a direct cause or predisposition factor for psychiatric disorders has been the subject of many studies for many years. Major depressive disorder, bipolar personality disorder, panic attack, anxiety and social phobia are just a few of these diseases. Food allergy reduces the quality of life of the individual. Therefore the nutrients and allergic reactions they cause during the diagnosis and treatment of these diseases are of great importance.
... While there is overall insufficient evidence to recommend specific diets as treatment for mental disorders, some individuals may benefit from self-experimenting with short-term elimination diets followed by reintroduction of specific foods to assess their perceived impact on psychiatric and other symptoms. 69,70 As diet, microbiome composition, and potential dietary sensitivities are highly variable between individuals and thus more difficult to empirically study in group analyses, this case-by-case approach to treating psychiatric symptoms with dietary modifications may be viewed as a form of personalized medicine. ...
Article
Full-text available
Background: Converging and accumulating evidence for the cross-communication among the nervous, immune, and endocrine systems, a field of study known as psychoneuroimmunology, implicates immunological dysfunction as a shared and common mechanism of both mental and physical illness. For example, psychiatric disorders like schizophrenia, bipolar disorder, major depression, and anxiety disorders have higher prevalence rates across a spectrum of autoimmune conditions compared to the general population. Additionally, subclinical immunological abnormalities are observed in a variety of psychiatric conditions, with chronic inflammation most extensively studied in the pathophysiology of depression. These observations blur the historical distinctions between mental and physical illness, yet clinical practice remains fragmented and primarily focused on differentially treating individual symptoms. Proposed thesis: Therapeutically targeting inflammation offers translational opportunities for integrating mental and physical healthcare, a key niche of the interdisciplinary field of health psychology. Conclusion: Utilizing a psychoneuroimmunological lens, health psychologists and clinicians can reconceptualize healthcare through integrative treatment approaches and advocacy for comprehensive policy-level reform at both the individual-level of care as well as community-wide prevention approaches.
Article
Full-text available
Most adverse reactions to food are patient self-reported and not based on validated tests but nevertheless lead to dietary restrictions, with patients believing that these restrictions will improve their symptoms and quality of life. We aimed to clarify the myths and reality of common food intolerances, giving clinicians a guide on diagnosing and treating these cases. We performed a narrative review of the latest evidence on the widespread food intolerances reported by our patients, giving indications on the clinical presentations, possible tests, and dietary suggestions, and underlining the myths and reality. While lactose intolerance and hereditary fructose intolerance are based on well-defined mechanisms and have validated diagnostic tests, non-coeliac gluten sensitivity and fermentable oligosaccharide, disaccharide, monosaccharide, and polyol (FODMAP) intolerance are mainly based on patients’ reports. Others, like non-hereditary fructose, sorbitol, and histamine intolerance, still need more evidence and often cause unnecessary dietary restrictions. Finally, the main outcome of the present review is that the medical community should work to reduce the spread of unvalidated tests, the leading cause of the problematic management of our patients.
Article
Full-text available
Adult hippocampal neurogenesis generates functional neurons from neural progenitor cells in the hippocampal dentate gyrus (DG) to complement and repair neurons and neural circuits, thus benefiting the treatment of depression. Increasing evidence has shown that aberrant microglial activity can disrupt the appropriate formation and development of functional properties of neurogenesis, which will play a crucial role in the occurrence and development of depression. However, the mechanisms of the crosstalk between microglia and adult hippocampal neurogenesis in depression are not yet fully understood. Therefore, in this review, we first introduce recent discoveries regarding the roles of microglia and adult hippocampal neurogenesis in the etiology of depression. Then, we systematically discuss the possible mechanisms of how microglia regulate adult hippocampal neurogenesis in depression according to recent studies, which involve toll-like receptors, microglial polarization, fractalkine-C-X3-C motif chemokine receptor 1, hypothalamic-pituitary-adrenal axis, cytokines, brain-derived neurotrophic factor, and the microbiota-gut-brain axis, etc. In addition, we summarize the promising drugs that could improve the adult hippocampal neurogenesis by regulating the microglia. These findings will help us understand the complicated pathological mechanisms of depression and shed light on the development of new treatment strategies for this disease.
Article
Full-text available
Dysfunction of gut barrier is known as “leaky gut” or increased intestinal permeability. Numerous recent scientific evidences showed the association between gut dysfunction and multiple gastrointestinal tract (GI) and non‐GI diseases. Research also demonstrated that food plays a crucial role to cause or remedy gut dysfunction related to diseases. We reviewed recent articles from electronic databases, mainly PubMed. The data were based on animal models, cell models, and human research in vivo and in vitro models. In this comprehensive review, our aim focused on the relationship between dietary factors, intestinal permeability dysfunction, and related diseases. This review synthesizes currently available literature and is discussed in three parts: (a) the mechanism of gut barrier and function, (b) food and dietary supplements that may promote gut health, and food or medication that may alter gut function, and (c) a table that organizes the synthesized information by general mechanisms for diseases related to leaky gut/intestinal permeability and associated dietary influences. With future research, dietary intervention could be a new target for individualized disease prevention and management. This review summarizes current literature, mainly from PubMed, on the mechanisms of gut barrier and function, including food and dietary supplements or medications that promote or alter gut health. It also provides a comprehensive and clear table about basic mechanisms for disease‐related leaky gut/increased intestinal permeability with associated dietary influences.
Article
Full-text available
Intestinal permeability dysfunction might be considered either a consequence or cause of systemic inflammation with release of proinflammatory cytokines. A growing body of evidence indicates a key role of the immune system and cytokines in depression. Cytokines cause tryptophan and 5-HT depletion and elevation of neurotoxic tryptophan catabolites by inducing indoleamine 2,3-dioxygenase (IDO). Another enzyme causing elevation of tryptophan catabolites is tryptophan 2,3-dioxygenase (TDO), which is activated by cortisol, often elevated during depression. Additionally, pro-inflammatory cytokines activate noradrenergic neurotransmission and the HPA axis, and also cause glucocorticoid receptor resistance. There is a growing body of evidence indicating a role of increased intestinal permeability (leaky gut syndrome) in many chronic diseases such as inflammatory bowel diseases, diabetes type I, allergy, asthma, autism, and depression. Numerous investigations indicate that many antidepressant drugs exhibit anti-inflammatory properties by lowering levels of pro-inflammatory cytokines, which might be related to their antidepressant mechanisms of action. A large group of anti-inflammatory substances and antioxidants also exhibit antidepressant potential or potentiate efficacy of antidepressant pharmacotherapy. The aim of the authors of this review is to present a potential relation between increased gut permeability, activation of the inflammatory response system, IgG food allergy (type III hypersensitivity reaction), and depression. Detection of a relationship between IgGdependent allergy and depression may form the basis for improving the therapeutic scheme.
Article
Full-text available
Background Depression is a chronic syndrome with a pathogenesis linked to various genetic, biological, and environmental factors. Several links between gut microbiota and depression have been established in animal models. In humans, however, few correlations have yet been demonstrated. The aim of our work was therefore to identify potential correlations between human fecal microbiota (as a proxy for gut microbiota) and depression.Methods We analyzed fecal samples from 55 people, 37 patients, and 18 non-depressed controls. Our analyses were based on data generated by Illumina deep sequencing of 16S rRNA gene amplicons.Key ResultsWe found several correlations between depression and fecal microbiota. The correlations, however, showed opposite directions even for closely related Operational Taxonomic Units (OTU's), but were still associated with certain higher order phylogroups. The order Bacteroidales showed an overrepresentation (p = 0.05), while the family Lachnospiraceae showed an underrepresentation (p = 0.02) of OTU's associated with depression. At low taxonomic levels, there was one clade consisting of five OTU's within the genus Oscillibacter, and one clade within Alistipes (consisting of four OTU's) that showed a significant association with depression (p = 0.03 and 0.01, respectively).Conclusions & InferencesThe Oscillibacter type strain has valeric acid as its main metabolic end product, a homolog of neurotransmitter GABA, while Alistipes has previously been shown to be associated with induced stress in mice. In conclusion, the taxonomic correlations detected here may therefore correspond to mechanistic models.
Article
IgG-dependent allergy may be one of a reason for the development or maintenance of the gastrointestinal diseases such as irritable bowel syndrome (IBS) or inflammatory bowel disease (IBD). Incorrect IgG-dependent reactions to food, result in formation of immune complex and consequently - chronic inflammation. An elimination diet based on the results of the study of specific IgG antibodies against the food antigens is beneficial in treatment of IBS, IBD and psychiatric disorders.
Article
Objectives. To analyze the relationship between depression and the immune system disorders that mediate an increased activity of the hypothalamic-pituitary-adrenal axis (HPA). Review. In the 1980s the relation between depression and cell-mediated response impairment (i.e. a reduced absolute number of T and B lymphocytes, and diminished number and hypoactivity of NK cells) was described in many studies. In the 1990s reports were published concerning the effect of HPA axis disturbances on the pathogenesis of depression, as well as the association of depressive disorders with an increased level of proinflammatory cytokines in patients in whom the concurrence of depression and an inflammatory process had been excluded. Conclusion. In the 20th century a significant progress was made in the understanding of mechanisms underlying the immune and endocrine system dysfunctions in the pathogenesis of recurrent depressive disorders. A number of stressful stimuli can lead to an increase in the levels of proinflammatory cytokines such as IL-1, IL-6 or TNF-α produced in the peripheral and central nervous system. The increased cytokine concentration in the c.n.s. results in an increased activity of the HPA axis and in consequence, in an increased concentration of corticoliberin and glucocorticosteroids. The latter can evoke and exacerbate the symptoms characteristic of depressive disorders.
Article
The primary function of the human intestine is to absorb nutrients and water. However, equally important is its ability to act as a selective barrier to protect the human system. Intestinal epithelium is formed by a monolayer of epithelial cells. Adjacent cells of the monolayer are sealed together by the formation of tight junctions (TJs)-complex protein systems. The structure of TJ involves transmembrane proteins linked to a cytoplasmic plaque, which is formed by a network of scaffolding and adaptor proteins, signalling components and actin-binding cytoskeleton linkers. TJs regulate paracellular transport of compounds as well as physical barrier function of epithelium, which is linked to pathogenesis of inflammatory bowel disease, ulcerative colitis, Crohn's disease and food allergies. Epithelium is intensively exposed to food components. Various food components can affect the functioning of TJ by modifying expression of TJ protein components and affecting signalling pathways involved in TJ regulation. The targeted usage of food components to modulate TJ permeability is of vital importance for enhancing the absorption of poorly permeable drugs or bioactive compounds and, on the other hand, for sealing the junction in order to limit the risk of intestine pathology.
Article
The luminal-mucosal interface of the intestinal tract is the first relevant location where microorganism-derived antigens and all other potentially immunogenic particles face the scrutiny of the powerful mammalian immune system. Upon regular functioning conditions, the intestinal barrier is able to effectively prevent most environmental and external antigens to interact openly with the numerous and versatile elements that compose the mucosal-associated immune system. This evolutionary super system is capable of processing an astonishing amount of antigens and non-immunogenic particles, approximately 100 tons in one individual lifetime, only considering food-derived components. Most important, to develop oral tolerance and proper active immune responses needed to prevent disease and inflammation, this giant immunogenic load has to be managed in a way that physiological inflammatory balance is constantly preserved. Adequate functioning of the intestinal barrier involves local and distant regulatory networks integrating the so-called brain-gut axis. Along this complex axis both brain and gut structures participate in the processing and execution of response signals to external and internal changes coming from the digestive tract, using multidirectional pathways to communicate. Dysfunction of brain-gut axis facilitates malfunctioning of the intestinal barrier, and vice versa, increasing the risk of uncontrolled immunological reactions that may trigger mucosal and brain low-grade inflammation, a putative first step to the initiation of more permanent gut disorders. In this chapter, we describe the structure, function and interactions of intestinal barrier, microbiota and brain-gut axis in both healthy and pathological conditions.