Sie sind auf Seite 1von 12

JOURNAL OF MEDICINAL FOOD

J Med Food 17 (12) 2014, 12611272


REVIEW
# Mary Ann Liebert, Inc., and Korean Society of Food Science and Nutrition
DOI: 10.1089/jmf.2014.7000

The Gut Microbiome and the Brain


Leo Galland
Foundation for Integrated Medicine, New York, New York, USA.

ABSTRACT The human gut microbiome impacts human brain health in numerous ways: (1) Structural bacterial compo-
nents such as lipopolysaccharides provide low-grade tonic stimulation of the innate immune system. Excessive stimulation
due to bacterial dysbiosis, small intestinal bacterial overgrowth, or increased intestinal permeability may produce systemic
and/or central nervous system inflammation. (2) Bacterial proteins may cross-react with human antigens to stimulate dys-
functional responses of the adaptive immune system. (3) Bacterial enzymes may produce neurotoxic metabolites such as D-
lactic acid and ammonia. Even beneficial metabolites such as short-chain fatty acids may exert neurotoxicity. (4) Gut microbes
can produce hormones and neurotransmitters that are identical to those produced by humans. Bacterial receptors for these
hormones influence microbial growth and virulence. (5) Gut bacteria directly stimulate afferent neurons of the enteric nervous
system to send signals to the brain via the vagus nerve. Through these varied mechanisms, gut microbes shape the architecture
of sleep and stress reactivity of the hypothalamic-pituitary-adrenal axis. They influence memory, mood, and cognition and are
clinically and therapeutically relevant to a range of disorders, including alcoholism, chronic fatigue syndrome, fibromyalgia,
and restless legs syndrome. Their role in multiple sclerosis and the neurologic manifestations of celiac disease is being
studied. Nutritional tools for altering the gut microbiome therapeutically include changes in diet, probiotics, and prebiotics.

KEY WORDS: D-lactic acid endotoxin microbial endocrinology microbiome prebiotics probiotics short-chain
fatty acids trimethylamine oxide (TMAO)

INTRODUCTION A study of germ-free (GF) mice found that the vast ma-
jority of chemicals circulating in blood are dependent on the
T he most surprising revelation of the Human Genome
Project is the small size of the human gene poolabout
26,000 functioning units1compared with the genomes
microbiome for their synthesis, although many are subse-
quently modified by the host.8 These chemicals have a
profound effect on mammalian behavior and neuroendo-
of much simpler organisms. Rice (Oryza sativa), for exam-
crine responses. This review will focus on research done in
ple, has about 46,000 functioning genes that have evolved
humans, but work done with GF rodents signals the evolu-
over 15 million years.2 Researchers call this the genome-
tionary importance of the microbiome in shaping mamma-
complexity conundrum,3 and some speculate that human
lian behavior, with important implications for human health.
physiologic and behavioral complexity may depend on the
The developmental abnormalities found in GF mice are
large number of microbial genes present in the human body.
totally reversible by colonization with intestinal bacteria
The term gut microbiome, in its strictest sense, describes
early in life but not in adulthood, suggesting that the mi-
the composite microbial genome found in the mammalian
crobiome influences brain development.9,10
gastrointestinal tract. The hundred trillion bacteria in the
When compared with conventional mice, GF mice show
body of an adult human contain about 4 million distinct
greater exploratory activity in an open-field activity box,
bacterial genes, with more than 95% of them located in the
suggesting less vigilance and caution.11 Similar behavioral
large intestine.4 Since most of these genes encode for en-
changes are produced in conventional mice by administra-
zymes and structural proteins that influence the functioning
tion of a mixture of nonabsorbed antibiotics for 7 days.12
of mammalian cells, the gut microbiome can be viewed as
Although some researchers have attributed these behavioral
an anaerobic bioreactor programmed to synthesize mole-
changes to diminished anxiety, elevation of striatal norepi-
cules which direct the mammalian immune system,5 modify
nephrine, dopamine, and serotonin turnover in the brains of
the mammalian epigenome,6 and regulate host metabolism.7
GF mice11 and elevated plasma levels of adrenal cortico-
trophic hormone and corticosterone in response to restraint
Manuscript received 9 September 2014. Revision accepted 9 October 2014. stress13 demonstrate heightened stress reactivity in GF mice,
Address correspondence to: Leo Galland, MD, Foundation for Integrated Medicine, New
an effect also seen in GF rats, who are, however, less active
York, NY, 10011, USA, E-mail: Lgallandmd@aol.com. and more cautious than conventional rats.14 It appears that

1261
1262 GALLAND

both timidity, a behavior pattern associated with mice, and mood, increased anxiety, and impaired long-term memory
aggressiveness, a behavior pattern associated with rats, re- for emotional stimuli.27 In addition, visceral pain sensitivity
quire a microbiome for their characteristic expression. Ei- thresholds are reduced and visceral pain (provoked by rectal
ther behavioral deviationthe increased risk taking of GF distension) is rated as more unpleasant after administration
mice or the withdrawal of GF ratscan significantly impair of low-dose LPS.28
survival in the wild, where the need to gather food should be Increased exposure to gut microbiome-derived LPS (en-
balanced against the need to avoid predators. These rodent dotoxemia) may occur in the elderly, in whom it is dimin-
studies suggest that the gut microbiome has strategic evo- ished by yogurt consumption,29 as a consequence of small
lutionary importance by modulating stress responses and intestinal bacterial overgrowth (SIBO),30,31 and secondary
influencing behaviors that impact the survival of species.15 to increased intestinal permeability resulting from extreme
Studies with different strains of laboratory mice indicate physiologic stress,32 ethanol exposure,33 or a fast-food
that there may be specific behavioral effects induced by style Western diet, high in both carbohydrate and saturated
specific microbiota. Balb/C mice, for example, are more fat.34
susceptible to stressors and to the effects of the anxiogenic LeClercq et al.35 have reported increased intestinal per-
neurohormone corticotrophin-releasing factor than are NIH meability, elevated blood LPS and peptidoglycan levels, and
Swiss mice.16 When GF variants of either strain are colo- low-grade systemic inflammation associated with psycho-
nized by gut microbes of the other strain, they begin be- logical symptoms of alcohol dependence in alcohol-dependent
having similar to conventional versions of the strain whose subjects. They tested inflammatory responses of peripheral
microbiome they have received.12 Balb/C mice become less blood mononuclear cells (PBMCs) to gut-derived bacterial
stress reactive, and NIH Swiss mice become more stress products in healthy controls and in chronic alcoholics before
reactive than their conventional counterparts. and during ethanol detoxification. They found activation of
Central nervous system (CNS) effects of the microbiome Toll-like receptors by LPS and peptidoglycans in PBMCs of
may be produced by immunologic, biochemical, or neuro- alcoholics, associated with increased messenger RNA and
endocrine mechanisms.17 plasma levels of IL-8, IL-1b, and IL-18. Levels of IL-8 and
IL-1b were positively correlated with alcohol consumption
IMMUNOLOGIC MECHANISMS and alcohol-craving scores. Using Cr51-EDTA as a probe of
intestinal permeability, they divided their population of
The innate immune system
chronic alcoholics into those with high and normal perme-
Structural components of the microbial cell wall continu- ability.36 The high permeability group had higher scores of
ally stimulate the innate immune system to produce cytokines, depression, anxiety, and alcohol craving than the low per-
creating a basal state of immune activation that begins at the meability group, as well as a distinct pattern of changes in the
intestinal mucosal surface and impacts the entire body.18 gut microbial population, characterized by decreased coloni-
The gut microbiome interacts with the hypothalamic- zation with bacteria known to have anti-inflammatory effects,
pituitary-adrenal (HPA) axis to shape the normal architecture Bifidobacterium species and Faecalibacterium prausnitzkii in
of sleep. Bacterial peptides induce intestinal macrophages particular. Those alcoholics who showed persistence of in-
and T-cells to produce the cytokines interleukin-1beta (IL- testinal hyperpermeability after 3 weeks of ethanol with-
1b) and tumor necrosis factor alpha (TNFa)19; bacterial cell drawal also demonstrated persistence of depression, anxiety,
wall lipopolysaccharides (LPS) induce synthesis of IL-18.20 and alcohol craving. Their theory is that for some alcoholics
The adult human gut is believed to contain about one gram of (probably 3050% of the total), ethanol consumption alters
LPS.15 IL-1b,21 TNFa22, and IL-1823 are inducers of nonrapid the gut microbiome to deplete protective bacteria, increas-
eye movement (nREM) sleep. Cortisol inhibits immune cell ing intestinal permeability and producing systemic inflam-
synthesis of these cytokines. IL-1b and TNFa show a circadian mation provoked by absorption of bacterial peptidoglycans
rhythm in human blood, with peak levels at midnight, when and LPS, which amplifies the psychopathology of ethanol
cortisol is the lowest, and trough levels in the early morning, addiction.
as plasma cortisol surges.24 The cortisol-induced decline in Increased intestinal permeability has also been described
microbiome-stimulated circulating IL-1b may orchestrate the in patients with chronic fatigue syndrome (CFS),37 fi-
normal shift from early sleep, which is predominantly nREM, bromyalgia, and complex regional pain syndrome.38 SIBO
to late sleep, which is dominated by REM.25 by itself can increase intestinal permeability39; SIBO is as-
Although cytokine secretion induced by low-level expo- sociated with fibromyalgia40 and restless legs syndrome
sure of immune cells to bacterial cell wall components (RLS),41 with treatment of SIBO producing clinically sig-
contributes to normal sleep patterns, excessive cytokine nificant improvement in a small group of patients with
levels are associated with disrupted sleep.26 Parenteral ad- RLS.41 The CNS effects of elevated gut-derived LPS or
ministration of LPS to humans in nanogram quantities peptidoglycan exposure might contribute to the pathogene-
(0.4 ng/kg body weight) increases plasma concentration of sis of these disorders.
pro-inflammatory cytokines IL-6 and TNFa and the anti- Researchers at Johns Hopkins University School of
inflammatory cytokines IL-10 and IL-1 receptor antagonist, Medicine found evidence of increased gut bacterial translo-
along with salivary and plasma cortisol and plasma nor- cation in schizophrenic patients, unrelated to antipsychotic
epinephrine. These changes are accompanied by depressed treatment. Presence of the translocation marker soluble CD14
tripled the risk of schizophrenia and was positively associated
with C-reactive protein (CRP) but not with LPS-binding
protein (LBP), suggesting that gut bacterial components other
than LPS may be stimulating monocyte activation and in-
flammation in schizophrenics.42
In summary, the gut microbiome stimulates a chronic
state of low-level activation of the innate immune system in
humans, which is influenced by the circadian pattern of
adrenal cortical function. Altered exposure to structural
components of the microbiome, which may occur because
of increased intestinal permeability or SIBO, may disrupt
normal neuroendocrine regulation and has been associated
with several disorders linked to abnormal CNS function.

Adaptive immunity
The adaptive immune system responds to specific microbes
with antibodies or antigen-specific cellular immune responses
and can produce CNS dysfunction through auto-immune re-
actions caused by molecular mimicry between bacterial and
self proteins. Although this is an area of ongoing investiga-
tion, there is presently little evidence for a link between the
gut microbiome, the adaptive immune system, specific auto-
immunity, and disorders of the CNS in humans.43,44 However,
in a laboratory model of multiple sclerosis, mice sensitized to
the autoantigen, myelin oligodendrocyte glycoprotein, only
developed experimental autoimmune encephalitis in the
presence of commensal bacteria.45
Celiac disease (CD) is a notable exception, although the
mechanism is indirect. Alterations in the gut microbiome
may play a primary role in the pathogenesis of CD,46 a
gluten-sensitive disease in which the adaptive immune
system damages not only the gut but also the brain. The most
common CNS manifestations of CD are ataxia (with or
without myoclonus), headache, and cognitive dysfunction.
Gastrointestinal symptoms are often absent in neurologic
CD, as are the usual marker of intestinal CD, transgluta-
minase (TG) antibodies. The autoimmune target in neuro-
logic gluten sensitivity is TG6 rather than TG2, which is the
target for autoantibodies measured in commercial tests.47
Most studies, but not all,48 have found significant differ-
ences between healthy children and children with CD in the
duodenal49,50 and oral51 microbial populations. Some of these
differences are the result of inflammation and disappear dur-
ing a gluten-free diet, but reduced levels of Bifidobacterium
species, a replicable finding, do not become normal with a
gluten-free diet.52,53 Infants at high risk of developing CD
because of family history and personal genotype show a
reduction in Bifidobacteria before the onset of illness.54
Bifidobacteria protect human intestinal cells from the toxic
effects of gliadin peptides, the inflammatory triggers of
CD, by altering their structure.55 They also induce an anti-
inflammatory response in stimulated human mononuclear
cells in tissue culture.56 Destruction of protective Bifido-
bacteria can explain the association between incident CD
and previous antibiotic exposure.57 Loss of Bifidobacteria
may play a pathogenetic role in CD and contribute to its
rising prevalence. Administration of Bifidobacterium long-
(FOS) favor acetate over lactate as an end-product of car-
bohydrate metabolism. Horses who had barley added to their
diets experienced a change in fecal flora characterized by
increased concentrations of lactic acid bacteria belonging to
the genera Lactobacillus and Streptococcus, associated with
an increase in D-lactate concentration in the stool. These
changes were prevented by administration of FOS.72

Ammonia
Ammonia is a well-known neurotoxin, produced in the
intestinal tract from urea by the action of bacterial ureases.
the stools of autistic children,105,106 may be responsible for impact of stress responses on infection, independent of the
increased propionate production.107 Autism is associated with effect of stress on host immunity.
early weaning from breast milk to infant formula.108 Com- The interbacterial communication system known as
pared with breast milk, infant formula feeding increases fecal quorum sensing utilizes hormone-like compounds referred
concentration of propionate and butyrate.109 to as inducers to regulate bacterial gene expression. En-
Williams et al. examined ileal biopsies of autistic children terohemorrhagic Escherichia coli (EHEC) serotype
with gastrointestinal (GI) complaints and found a deficit of O157:H7 is responsible for outbreaks of bloody diarrhea.
genes encoding disaccharidases and hexose transport en- Sperandio et al. showed that exogenous epinephrine is an
zymes, indicating impairment of the primary pathway for inducer of the 0157:H7 virulence factor.120 EHEC growing
carbohydrate digestion and absorption in enterocytes,110 a in a stressed host may be more virulent than in a non-
finding which suggests that bacterial dysbiosis results from an stressed host.
underlying impairment of digestion and absorption. In a In addition to specific effects on potential pathogens, host
subsequent report, they observed the presence of a unique stress responses may provoke widespread changes in gut
genus of aerobic gram-negative rods, Sutterella, in ileal bi- microbial composition. Bailey et al. stressed mice with a
opsies of autistic children with GI complaints but no children process called Social Disruption (SDR), which significantly
with GI complaints who were not autistic.111 Western im- alters bacterial community structure in the cecum, espe-
munoblots revealed plasma IgG or IgM antibody reactivity to cially when the microbiota are assessed immediately after
the species Sutterella wadsworthensis in the majority of exposure to the social stressor. SDR reduces the relative
children with positive biopsies. S. wadsworthensis is a gas- abundance of bacteria from the genus Bacteroides, while
trointestinal pathogen that may be mistaken for Campylo- increasing the relative abundance of bacteria from the genus
bacter jejuni and may also be found in the stool of healthy Clostridium. It also increases circulating levels of inflam-
individuals.112 Following the report by Williams et al., Wang matory cytokines, IL-6 in particular, which are significantly
et al. confirmed an association between abundance of Sut- correlated with stressor-induced changes in microbiome
terella and the presence of autism. They studied stool spec- composition. Pretreatment of mice with antibiotics alters the
imens, not ileal biopsies, so they were able to examine the changes in community structure and attenuates the cytokine
relationship between Sutterella and GI complaints. There was response after SDR.121
none. Levels of Sutterella were related to autism only.113 A study of college students undergoing the stress of final
A role for the gut microbiome and its metabolites in examinations found a decrease in the relative concentration
ASD is one of the leading areas in autism research these of lactic acid bacteria in feces after the examination122
days,114,115 but the findings do not yet permit a single co- (speciation was not performed). Since lactic acid bacteria
herent theory on which to base therapeutic decisions. A have immunomodulating effects123,124 and may influence
recent report in the New England Journal of Medicine de- the broader composition of the gut microbiome,125 it seems
scribes structural brain abnormalities in autistic children that likely that humans respond to psychosocial stress with re-
began during prenatal brain development,116 indicating that sponses that are comparable to, if distinct from, the reactions
the roots of autism may be found in utero. Perhaps greater of laboratory animals.
focus should be placed on the maternal gestational micro- Since gut microbes modify stress responses in laboratory
biome. Immune activation of pregnant mice (maternal im- animals, several human clinical trials have been conducted
mune activation [MIA]) can create behavioral changes similar using probiotics to study their impact on stress reactivity and
to ASD in their offspring.117 Administration of a single pro- mood. In the most frequently cited study, healthy French
biotic, Bacteroides fragilis, corrects excessive gut perme- adults were administered a combination of Lactobacillus
ability, alters gut microbial composition, and ameliorates helveticus R0052 and B. longum R0175 (PF) for 30 days in a
defects in communication and stereotypic, anxiety-like, and double-blind, placebo-controlled, randomized parallel group
sensorimotor behaviors in the MIA model.

NEUROENDOCRINE MECHANISMS
Bacteria can synthesize and respond to hormones and
neurotransmitters. Lactobacillus species produce acetyl-
choline and gamma-amino butyrate (GABA); Bifido-
bacterium species produce GABA; Escherichia produce
norepinephrine, serotonin and dopamine; Streptococcus and
Enterococcus produce serotonin; and Bacillus species pro-
duce norepinephrine and dopamine.17 These organisms are
responsive to human hormones and neurotransmitters,118
which impact their growth and virulence. Lyte119 has re-
viewed research indicating that growth of Escherichia coli
and other Proteobacteria is greatly enhanced by physio-
logic concentrations of norepinephrine, explaining a direct
THE GUT MICROBIOME AND THE BRAIN 1267

microbiome (the microbial metabolome) is at the in food-associated biofilms produce unique signal-
cutting edge of microbiome research.151 Although ing molecules and may represent a new dimension
inter-bacterial inhibition has been understood for a in the relationship between food, microbes, and
long time, inter-bacterial growth synergy may be as human health.160
important. Propionibacterium freudenreichii, a bac-
terium found in Swiss cheese, produces substances
CONCLUSION
that enhance growth of Bifidobacteria.152 Adminis-
tration of this bifidogenic substance to patients with Experimental studies with human volunteers and with
ulcerative colitis produced an increase in fecal buty- small mammals demonstrate effects of commensal intestinal
rate associated with clinical improvement.153 bacteria on behavior and brain function that are contextually
(2) Most studies indicate that both health and decreased meaningful and which appear to be biologically significant.
adiposity are associated with increased diversity of Gut bacteria influence reactivity of the HPA axis and the
the gut microflora. Dietary restriction increases di- induction and maintenance of nREM sleep. They may in-
versity; dietary excess tends to reduce it.154 fluence mood, pain sensitivity and normal brain develop-
(3) Extreme dietary changes, such as adoption of a ke- ment.
togenic diet, produce immediate profound changes in Clinical studies have demonstrated distinct pathological
the human gut microbiome. Less dramatic interven- CNS effects of commensal gut bacteria in hepatic cirrhosis
tions produce mild to moderate changes that vary and short bowel syndrome and have led researchers to
from person to person and tend to be less than inter- speculate on possible adverse effects of gut microbes in al-
individual variability.155 cohol dependence, CFS, fibromyalgia, RLS, ASD, schizo-
(4) A normal microbiome increases nutrient bioavail- phrenia, mood disorders, and degenerative or autoimmune
ability. In order to maintain their health, GF mice neurologic disease. Adverse effects have been attributed to
should be fed a diet of higher nutrient quantity and alterations in bacterial community structure (dysbiosis),
diversity than conventional mice.156 The effect of diet SIBO, and increased intestinal permeability.
change on the microbiome is not likely to be unidi- Several mechanisms, none mutually exclusive, may en-
rectional. An altered microbiome may change the able commensal gut bacteria to influence function or dys-
effect of food on the host. function in the CNS: (1) stimulation of host immune
(5) Alterations of the microbiome may alter the physio- responses leading to diverse patterns of systemic cytokine
logic effect of nutrients. A critical example of this activation; (2) synthesis of absorbable neuroactive metab-
phenomenon is revealed in the work of Hazen and olites, including neurotransmitters; and (3) alterations in
colleagues at The Cleveland Clinic, who found that neuronal circuitry by direct microbial effects on the ENS,
higher plasma levels of the vascular toxin trimethyla- with CNS transmission through vagal and other routes. The
mine-N-oxide (TMAO) conferred an increased risk of only mechanisms with a high level of proof in humans are
major cardiovascular events during a 3-year follow- the neurotoxic effects of ammonia in HE and of D-lactic
up.157 They also demonstrated that plasma TMAO is the acid in short bowel syndrome.
product of gut microbial metabolism of dietary choline CNS and neuroendocrine activity, stress responses in
to trimethylamine (TMA), followed by hepatic oxida- particular, may, in turn, influence the composition of the gut
tion of TMA to TMAO. The same team demonstrated microbiome by differentially altering the growth of bacterial
that the gut microbiome of human vegetarians produces species and the production of bacterial virulence factors.
significantly less TMA than the gut microbiome of Enterobacteriaceae, a family that includes most of the
omnivores when fed L-carnitine, another substrate for aerobic Gram-negative pathogens, is especially well tuned
TMA synthesis.158 In this way, an essential dietary nu- to exploiting host stress responses for enhancing bacterial
trient (choline) is converted to a vasculopathic sub- growth and virulence.
stance by the action of a microbiome whose Dietary patterns also modify microbiome composition
composition is determined by a previous dietary pattern. and function, in complex ways that vary among individuals
(6) Diet involves more than nutrients. Polyphenols are and cultures and are the subject of intense ongoing research.
bioactive non-nutrient plant compounds whose bio- Prebiotics, probiotics, and fermented foods such as yogurt
availability and physiologic effects greatly depend on may influence the impact of the gut microbiome on the CNS
their transformation by components of the gut mi- and have shown significant effects on brain function in a
crobiota. Polyphenols, in turn, alter microbial growth number of experimental trials and clinical studies. Along
patterns. The polyphenol composition of an individ- with diet, these functional food components may offer fu-
uals diet may be more important than macronutrient ture opportunities for altering the microbiome to enhance
composition for determining growth effects on gut cognitive or emotive function and prevent or treat neuro-
microbes.159 logic disorders.
(7) Colonic bacteria have been found in biofilms formed
around food particles. These organize gut microbes
AUTHOR DISCLOSURE STATEMENT
into distinct communities that behave differently
from their planktonic counterparts. Bacteria living No competing financial interests exist.
THE GUT MICROBIOME AND THE BRAIN 1267

microbiome (the microbial metabolome) is at the in food-associated biofilms produce unique signal-
cutting edge of microbiome research.151 Although ing molecules and may represent a new dimension
inter-bacterial inhibition has been understood for a in the relationship between food, microbes, and
long time, inter-bacterial growth synergy may be as human health.160
important. Propionibacterium freudenreichii, a bac-
terium found in Swiss cheese, produces substances
CONCLUSION
that enhance growth of Bifidobacteria.152 Adminis-
tration of this bifidogenic substance to patients with Experimental studies with human volunteers and with
ulcerative colitis produced an increase in fecal buty- small mammals demonstrate effects of commensal intestinal
rate associated with clinical improvement.153 bacteria on behavior and brain function that are contextually
(2) Most studies indicate that both health and decreased meaningful and which appear to be biologically significant.
adiposity are associated with increased diversity of Gut bacteria influence reactivity of the HPA axis and the
the gut microflora. Dietary restriction increases di- induction and maintenance of nREM sleep. They may in-
versity; dietary excess tends to reduce it.154 fluence mood, pain sensitivity and normal brain develop-
(3) Extreme dietary changes, such as adoption of a ke- ment.
togenic diet, produce immediate profound changes in Clinical studies have demonstrated distinct pathological
the human gut microbiome. Less dramatic interven- CNS effects of commensal gut bacteria in hepatic cirrhosis
tions produce mild to moderate changes that vary and short bowel syndrome and have led researchers to
from person to person and tend to be less than inter- speculate on possible adverse effects of gut microbes in al-
individual variability.155 cohol dependence, CFS, fibromyalgia, RLS, ASD, schizo-
(4) A normal microbiome increases nutrient bioavail- phrenia, mood disorders, and degenerative or autoimmune
ability. In order to maintain their health, GF mice neurologic disease. Adverse effects have been attributed to
should be fed a diet of higher nutrient quantity and alterations in bacterial community structure (dysbiosis),
diversity than conventional mice.156 The effect of diet SIBO, and increased intestinal permeability.
change on the microbiome is not likely to be unidi- Several mechanisms, none mutually exclusive, may en-
rectional. An altered microbiome may change the able commensal gut bacteria to influence function or dys-
effect of food on the host. function in the CNS: (1) stimulation of host immune
(5) Alterations of the microbiome may alter the physio- responses leading to diverse patterns of systemic cytokine
logic effect of nutrients. A critical example of this activation; (2) synthesis of absorbable neuroactive metab-
phenomenon is revealed in the work of Hazen and olites, including neurotransmitters; and (3) alterations in
colleagues at The Cleveland Clinic, who found that neuronal circuitry by direct microbial effects on the ENS,
higher plasma levels of the vascular toxin trimethyla- with CNS transmission through vagal and other routes. The
mine-N-oxide (TMAO) conferred an increased risk of only mechanisms with a high level of proof in humans are
major cardiovascular events during a 3-year follow- the neurotoxic effects of ammonia in HE and of D-lactic
up.157 They also demonstrated that plasma TMAO is the acid in short bowel syndrome.
product of gut microbial metabolism of dietary choline CNS and neuroendocrine activity, stress responses in
to trimethylamine (TMA), followed by hepatic oxida- particular, may, in turn, influence the composition of the gut
tion of TMA to TMAO. The same team demonstrated microbiome by differentially altering the growth of bacterial
that the gut microbiome of human vegetarians produces species and the production of bacterial virulence factors.
significantly less TMA than the gut microbiome of Enterobacteriaceae, a family that includes most of the
omnivores when fed L-carnitine, another substrate for aerobic Gram-negative pathogens, is especially well tuned
TMA synthesis.158 In this way, an essential dietary nu- to exploiting host stress responses for enhancing bacterial
trient (choline) is converted to a vasculopathic sub- growth and virulence.
stance by the action of a microbiome whose Dietary patterns also modify microbiome composition
composition is determined by a previous dietary pattern. and function, in complex ways that vary among individuals
(6) Diet involves more than nutrients. Polyphenols are and cultures and are the subject of intense ongoing research.
bioactive non-nutrient plant compounds whose bio- Prebiotics, probiotics, and fermented foods such as yogurt
availability and physiologic effects greatly depend on may influence the impact of the gut microbiome on the CNS
their transformation by components of the gut mi- and have shown significant effects on brain function in a
crobiota. Polyphenols, in turn, alter microbial growth number of experimental trials and clinical studies. Along
patterns. The polyphenol composition of an individ- with diet, these functional food components may offer fu-
uals diet may be more important than macronutrient ture opportunities for altering the microbiome to enhance
composition for determining growth effects on gut cognitive or emotive function and prevent or treat neuro-
microbes.159 logic disorders.
(7) Colonic bacteria have been found in biofilms formed
around food particles. These organize gut microbes
AUTHOR DISCLOSURE STATEMENT
into distinct communities that behave differently
from their planktonic counterparts. Bacteria living No competing financial interests exist.
REFERENCES 19. Heumann D, Barras C, Severin A, Glauser MP, Tomasz A:
Gram-positive cell walls stimulate synthesis of tumor necrosis
1. Venter JC, Adams MD, Myers EW, et al.: The sequence of the factor alpha and interleukin-6 by human monocytes. Infect
human genome. Science 2001;291:13041351. Immun 1994;62:27152721.
2. Jacquemin J, Ammiraju JS, Haberer G, et al.: Fifteen million 20. Ulevitch RJ, Tobias PS: Receptor-dependent mechanisms of
years of evolution in the Oryza genus shows extensive gene cell stimulation by bacterial endotoxin. Annu Rev Immunol
family expansion. Mol Plant 2014;7:642656. 1995;13:437457.
3. Surjyadipta B, Lukiw WJ, Alzheimers disease and the micro- 21. Alam MN, McGinty D, Bashir T, et al.: Interleukin-1beta
biome. Front Cell Neurosci 2013;7:153160. modulates state-dependent discharge activity of preoptic area
4. Qin J, Li R, Raes J, Arumugam M, et al.: A human gut mi- and bqs!d for%br@in neurons: role in{lEap--$;4.5(regulavigo.)]TJ/F31Tf27
crobial gene catalogue established by metagenomic sequencing.
Nature 2010;464:5965.
5. Hooper LV: Bacterial contributions to mammalian gut devel-
opment. Trends Microbiol 2004;12:129134.
6. Li M, Wang B, Zhang M, et al.: Symbiotic gut microbes mod-
ulate human metabolic phenotypes. PNAS 2008;105:2117
2122.
7. Jacobsen UP, Nielsen HB, Hildebrand F, et al.: The chemical
interactome space between the human host and the genetically
defined gut metabotypes. ISME J 2013;7:730742.
8. Wikoff WR, Anfora AT, Liu J, et al.: Metabolomics analysis
reveals large effects of gut microflora on mammalian blood
metabolites. Proc Natl Acad Sci USA 2009;106:36983703.
9. Neufeld KM, Kang N, Bienenstock J, Foster JA: Reduced
anxiety-like behavior and central neurochemical change in
germ-free mice. Neurogastroenterol Motil 2011;23:255264,
e119.
10. Diamond B, Huerta PT, Tracey K, Volpe BT: It takes guts to
grow a brain: increasing evidence of the important role of the
intestinal microflora in neuro- and immune-modulatory func-
tions during development and adulthood. Bioessays 2011;33:
588591.
11. Diaz Heijtz R, Wang S, Anuar F, et al.: Normal gut microbiota
modulates brain development and behavior. Proc Natl Acad Sci
USA 2011;108:30473052.
12. Bercik P, Park AJ, Sinclair D, et al.: The anxiolytic effect of
Bifidobacterium longum NCC3001 involves vagal pathways for
gut-brain communication. Neurogastroenterol Motil 2011;23:
11321139
13. Sudo N, Chida Y, Aiba Y, et al.: Postnatal microbial coloni-
zation programs the hypothalamic-pituitary-adrenal system for
stress response in mice. J Physiol 2004;558(Pt 1):263275.
14. Crumeyrolle-Arias M, Jaglin M, Bruneau A, et al.: Absence
of the gut microbiota enhances anxiety-like behavior and neu-
roendocrine response to acute stress in rats. Psychoneur-
oendocrinology 2014;42:207217.
15. Bested AC, Logan AC, Selhub EM: Intestinal microbiota,
probiotics and mental health: from Metchnikoff to modern ad-
vances: Part IIcontemporary contextual research. Gut Pathog
2013;5:3.
16. Conti LH, Costello DG, Martin LA, et al.: Mouse strain dif-
ferences in the behavioral effects of corticotropin-releasing
factor (CRF) and the CRF antagonist alpha-helical CRF9-41.
Pharmacol Biochem Behav 1994;48:497503.
17. Cryan JF, Dinan TG: Mind-altering microorganisms: the impact
of the gut microbiota on brain and behaviour. Nat Rev Neurosci
2012;13:701712.
18. Duerkop BA, Vaishnava S, Hooper LV: Immune responses to
the microbiota at the intestinal mucosal surface. Immunity 2009;
31:368376.
108. Tanoue Y, Oda S: Weaning time of children with infantile
autism. J Autism Dev Disord 1989;19:425434.
109. Macia L, Viltart O, Verwaerde C, et al.: Genes involved in
obesity: adipocytes, brain and microflora. Genes Nutr 2006;1:
189212.
110. Williams BL, Hornig M, Buie T, et al.: Impaired carbohydrate
digestion and transport and mucosal dysbiosis in the intestines
of children with autism and gastrointestinal disturbances. PLoS
One 2011;6:e24585.
111. Williams BL, Hornig M, Parekh T, Lipkin WI: Application of
novel PCR-based methods for detection, quantitation, and
phylogenetic characterization of Sutterella species in intestinal
biopsy samples from children with autism and gastrointestinal
disturbances. MBio 2012;3:pii: e00261-11.
112. Engberg J, On SL, Harrington CS, Gerner-Smidt P: Prevalence
of Campylobacter, Arcobacter, Helicobacter, and Sutterella
spp. in human fecal samples as estimated by a reevaluation of
isolation methods for Campylobacters. J Clin Microbiol
2000;38:286291.
113. Wang L, Christophersen CT, Sorich MJ, et al.: Increased
abundance of Sutterella spp. and Ruminococcus torques in feces
of children with autism spectrum disorder. Mol Autism 2013;4:
42.
1270 GALLAND

horses subjected to a sudden change in diet. J Anim Sci 2008; 89. Tan J, McKenzie C, Potamitis M, et al.: The role of short-chain
86:316323. fatty acids in health and disease. Adv Immunol 2014;121:
73. Skowronska M, Albrecht J: Alterations of blood brain barrier 91119.
function in hyperammonemia: an overview. Neurotox Res 90. Harrison IF, Dexter DT: Epigenetic targeting of histone dea-
2012;21:236244. cetylase: therapeutic potential in Parkinsons disease? Phar-
74. Kawaguchi T, Taniguchi E, Sata M: Effects of oral branched- macol Ther 2013;140:3452.
chain amino acids on hepatic encephalopathy and outcome in 91. Mahgoub M, Monteggia LM: Epigenetics and psychiatry.
patients with liver cirrhosis. Nutr Clin Pract 2013;28:580588. Neurotherapeutics 2013;10:734741.
75. Irimia R, Stanciu C, Cojocariu C, et al.: Oral glutamine chal- 92. Haberland M, Montgomery RL, Olson EN: The many roles of
lenge improves the performance of psychometric tests for the histone deacetylases in development and physiology: implica-
diagnosis of minimal hepatic encephalopathy in patients with tions for disease and therapy. Nat Rev Genet 2009;8:3242.
liver cirrhosis. J Gastrointestin Liver Dis 2013;22:277281. 93. Konsoula Z, Barile FA: Epigenetic histone acetylation and
76. Montgomery JY, Bajaj JS: Advances in the evaluation and deacetylation mechanisms in experimental models of neurode-
management of minimal hepatic encephalopathy. Curr Gas- generative disorders. J Pharmacol Toxicol Methods 2012;66:
troenterol Rep 2011;13:2633. 215220.
77. Quero Guillen JC, Groeneweg M, Jimenez Saenz M, et al.: Is it 94. Graff J, Tsai LH: The potential of HDAC inhibitors as cognitive
a medical error if we do not screen cirrhotic patients for mini- enhancers. Annu Rev Pharmacol Toxicol 2013;53:311330.
mal hepatic encephalopathy? Rev Esp Enferm Dig 2002;94: 95. Wauson EM, Lorente-Rodrguez A, Cobb MH: Minireview:
544557. nutrient sensing by G protein-coupled receptors. Mol En-
78. Bajaj JS, Ridlon JM, Hylemon PB, et al.: Linkage of gut mi- docrinol 2013;27:11881197.
crobiome with cognition in hepatic encephalopathy. Am J 96. Heng BC, Aubel D, Fussenegger M: An overview of the diverse
Physiol Gastrointest Liver Physiol 2012;302:G168G175. roles of G-protein coupled receptors (GPCRs) in the patho-
79. Bajaj JS, Hylemon PB, Ridlon JM, et al.: Colonic mucosal physiology of various human diseases. Biotechnol Adv 2013;
microbiome differs from stool microbiome in cirrhosis and 31:16761694.
hepatic encephalopathy and is linked to cognition and inflam- 97. Tazoe H, Otomo Y, Kaji I, et al.: Roles of short-chain fatty
mation. Am J Physiol Gastrointest Liver Physiol 2012;303: acids receptors, GPR41 and GPR43 on colonic functions. J
G675G685. Physiol Pharmacol 2008;59 Suppl 2:251262.
80. Zhang Z, Zhai H, Geng J, et al.: Large-scale survey of gut 98. Kimura I, Inoue D, Maeda T, et al.: Short-chain fatty acids and
microbiota associated with MHE Via 16S rRNA-based pyr- ketones directly regulate sympathetic nervous system via G
osequencing. Am J Gastroenterol 2013;108:16011611. protein-coupled receptor 41 (GPR41). Proc Natl Acad Sci USA
81. Sharma BC, Sharma P, Lunia MK, et al.: A randomized, dou- 2011;108:80308035.
ble-blind, controlled trial comparing rifaximin plus lactulose 99. Puertollano E, Kolida S, Yaqoob P: Biological significance of
with lactulose alone in treatment of overt hepatic encephalop- short-chain fatty acid metabolism by the intestinal microbiome.
athy. Am J Gastroenterol 2013;108:14581463. Curr Opin Clin Nutr Metab Care 2014;17:139144.
82. Malaguarnera M, Greco F, Barone G, et al.: Bifidobacterium 100. MacFabe D. Autism: metabolism, mitochondria, and the mi-
longum with fructo-oligosaccharide (FOS) treatment in minimal crobiome. Global Adv Health Med 2013;2:5266.
hepatic encephalopathy: a randomized, double-blind, placebo- 101. Wang L, Christophersen CT, Sorich MJ, et al.: Elevated fecal
controlled study. Dig Dis Sci 2007;52:32593265. short chain fatty acid and ammonia concentrations in children
83. Liu Q, Duan ZP, Ha DK, et al.: Synbiotic modulation of gut with autism spectrum disorder. Dig Dis Sci 2012;57:20962102.
flora: effect on minimal hepatic encephalopathy in patients with 102. Wang L, et al.: Gut bacterial and fermentation profiles are al-
cirrhosis. Hepatology 2004;39:14411449. tered in children with autism. J Gastroenterol Hepatol 2010;
84. Macfarlane GT, Macfarlane S: Fermentation in the human large 25(Suppl. 3):A116A119.
intestine: its physiologic consequences and the potential con- 103. Gondalia SV, Palombo EA, Knowles SR, et al.: Molecular
tribution of prebiotics. J Clin Gastroenterol 2011;45 Suppl: characterisation of gastrointestinal microbiota of children with
S120S127. autism (with and without gastrointestinal dysfunction) and their
85. Subaric D, Ackar D, Babic J, Milicevic B: Starch for health. neurotypical siblings. Autism Res 2012;5:419427.
Med Glas (Zenica) 2012;9:1722. 104. Kang DW, Park JG, Ilhan ZE, et al.: Reduced incidence of
86. De Preter V, Geboes KP, Bulteel V, Vandermeulen G: Kinetics Prevotella and other fermenters in intestinal microflora of au-
of butyrate metabolism in the normal colon and in ulcerative tistic children. PLoS One 2013;8:e68322.
colitis: the effects of substrate concentration and carnitine on 105. Parracho HM, Bingham MO, Gibson GR, McCartney AL.
the b-oxidation pathway. Aliment Pharmacol Ther 2011;34: Differences between the gut microflora of children with autistic
526532. spectrum disorders and that of healthy children. J Med Micro-
87. Segain JP, Raingeard de la Bletiere D, Bourreille A, et al.: biol 2005;54:987991.
Butyrate inhibits inflammatory responses through NFkappaB 106. Song Y, Liu C, Finegold SM. Real-time PCR quantitation of
inhibition: implications for Crohns disease. Gut 2000;47: clostridia in feces of autistic children. Appl Environ Microbiol
397403. 2004;70:64596465.
88. Al-Lahham SH, Peppelenbosch MP, Roelofsen H, et al.: 107. Wang L, Christophersen CT, Sorich MJ, et al.: Low relative
Biological effects of propionic acid in humans; metabolism, abundances of the mucolytic bacterium Akkermansia mucini-
potential applications and underlying mechanisms. Biochim phila and Bifidobacterium spp. in feces of children with autism.
Biophys Acta 2010;1801:11751183. Appl Environ Microbiol 2011;77:67186721.
108. Tanoue Y, Oda S: Weaning time of children with infantile
autism. J Autism Dev Disord 1989;19:425434.
109. Macia L, Viltart O, Verwaerde C, et al.: Genes involved in
obesity: adipocytes, brain and microflora. Genes Nutr 2006;1:
189212.
110. Williams BL, Hornig M, Buie T, et al.: Impaired carbohydrate
digestion and transport and mucosal dysbiosis in the intestines
of children with autism and gastrointestinal disturbances. PLoS
One 2011;6:e24585.
111. Williams BL, Hornig M, Parekh T, Lipkin WI: Application of
novel PCR-based methods for detection, quantitation, and
phylogenetic characterization of Sutterella species in intestinal
biopsy samples from children with autism and gastrointestinal
disturbances. MBio 2012;3:pii: e00261-11.
112. Engberg J, On SL, Harrington CS, Gerner-Smidt P: Prevalence
of Campylobacter, Arcobacter, Helicobacter, and Sutterella
spp. in human fecal samples as estimated by a reevaluation of
isolation methods for Campylobacters. J Clin Microbiol
2000;38:286291.
113. Wang L, Christophersen CT, Sorich MJ, et al.: Increased
abundance of Sutterella spp. and Ruminococcus torques in feces
of children with autism spectrum disorder. Mol Autism 2013;4:
42.
1272 GALLAND

142. Turnbaugh PJ, Gordon JI: The core gut microbiome, energy 151. Dorrestein PC, Mazmanian SK, Knight R: Finding the missing
balance, and obesity. J Physiol 2009;587:41534158. links among metabolites, microbes, and the host. Immunity
143. Rajilic-Stojanovic M, Heilig HG, Molenaar D, Kajander K, 2014;40:824832.
Surakka A, Smidt H, de Vos WM: Development and application 152. Kaneko T, Mori H, Iwata M, Meguro S: Growth stimulator for
of the human intestinal tract chip, a phylogenetic microarray: bifidobacteria produced by Propionibacterium freudenreichii
analysis of universally conserved phylotypes in the abundant and several intestinal bacteria. J Dairy Sci 1994;77:393404.
microbiota of young and elderly adults. Environ Microbiol 153. Suzuki A, Mitsuyama K, Koga H, et al.: Bifidogenic growth
2009;11:17361751. stimulator for the treatment of active ulcerative colitis: a pilot
144. Dethlefsen L, Eckburg PB, Bik EM, Relman DA: Assembly of study. Nutrition 2006;22:7681.
the human intestinal microbiota. Trends Ecol Evol 2006;21: 154. Joyce SA, Gahan CG: The gut microbiota and the metabolic
517523. health of the host. Curr Opin Gastroenterol 2014;30:120127.
145. Booijink CC, El-Aidy S, Rajilic-Stojanovic M, et al.: High 155. Albenberg LG, Wu GD: Diet and the intestinal microbiome:
temporal and inter-individual variation detected in the human associations, functions, and implications for health and disease.
ileal microbiota. Environ Microbiol 2010;12:32133227. Gastroenterology 2014;146:15641572.
146. Galland L: Patient-centered care: antecedents, triggers, and 156. Jeffery IB, OToole PW: Diet-microbiota interactions and their
mediators. Altern Ther Health Med 2006;12:6270. implications for healthy living. Nutrients 2013;5:234252.
147. Tap J, Mondot S, Levenez F, Pelletier E, et al.: Towards the 157. Tang WH, Wang Z, Levison BS, et al.: Intestinal microbial
human intestinal microbiota phylogenetic core. Environ Mi- metabolism of phosphatidylcholine and cardiovascular risk. N
crobiol 2009;11:25742584. Engl J Med 2013;368:15751584.
148. Wu GD, Chen J, Hoffmann C, et al.: Linking long-term dietary 158. Koeth RA, Wang Z, Levison BS et al.: Intestinal microbiota
patterns with gut microbial enterotypes. Science 2011;334: metabolism of L-carnitine, a nutrient in red meat, promotes
105108. atherosclerosis. Nat Med 2013;19:576585.
149. Walker WA: Initial intestinal colonization in the human infant 159. Laparra JM, Sanz Y: Interactions of gut microbiota with func-
and immune homeostasis. Ann Nutr Metab 2013;63 Suppl 2: tional food components and nutraceuticals. Pharmacol Res
815. 2010;61:219225.
150. Mayne AJ, Handy DJ, Preece MA, et al.: Dietary management 160. Van Wey AS, Cookson AL, Roy NC, et al.: Bacterial biofilms
of D-lactic acidosis in short bowel syndrome. Arch Dis Child associated with food particles in the human large bowel. Mol
1990;65:229231. Nutr Food Res 2011;55:969978.

Das könnte Ihnen auch gefallen