Gut microbiota and central nervous system development

Gut microbiota and central nervous system development

Accepted Manuscript Gut microbiota and central nervous system development Nicola Principi, Susanna Esposito PII: S0163-4453(16)30251-1 DOI: 10.1016...

754KB Sizes 0 Downloads 150 Views

Accepted Manuscript Gut microbiota and central nervous system development Nicola Principi, Susanna Esposito PII:

S0163-4453(16)30251-1

DOI:

10.1016/j.jinf.2016.09.010

Reference:

YJINF 3824

To appear in:

Journal of Infection

Received Date: 5 August 2016 Revised Date:

26 September 2016

Accepted Date: 29 September 2016

Please cite this article as: Principi N, Esposito S, Gut microbiota and central nervous system development, Journal of Infection (2016), doi: 10.1016/j.jinf.2016.09.010. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT 1

GUT MICROBIOTA AND CENTRAL NERVOUS SYSTEM DEVELOPMENT

2

Nicola Principi, Susanna Esposito

4

Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation,

5

Università degli Studi di Milano, Fondazione IRCCS Ca’ Granda Ospedale Maggiore

6

Policlinico, Milan, Italy

7

Short title: Gut microbiota and central nervous system

9

Abstract: 244 words. Text: 5,520 words.

M AN U

10

SC

8

RI PT

3

11 12

Correspondence and requests for reprints should be addressed to:

14

Susanna Esposito,

15

Pediatric Highly Intensive Care Unit,

16

Department of Pathophysiology and Transplantation,

17

Università degli Studi di Milano,

18

Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico,

19

Via Commenda 9,

20

20122 Milano,

21

Italy.

22

Tel.: +39-02-55032498; Fax: +39-02-50320206;

23

E-mail: [email protected]

AC C

EP

TE D

13

1

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT 1

ABSTRACT

3

Objectives: Gut dysbiosis has been associated with several clinically relevant conditions,

4

including alterations of central nervous system (CNS) structure and function development.

5

This review discussed aspects of the relationship between gut microbiota and the CNS

6

during development.

7

Methods: PubMed was used to search for all of the studies published over the last 15

8

years using the key word “microbiota” and “gut” or “intestinal” and “nervous system”. More

9

than 350 articles were found, and only those published in English and providing data on

SC

RI PT

2

aspects related to neurologic diseases were included in the evaluation.

11

Results: The data indicate that the gut microbiota influences CNS development and

12

function and that gut dysbiosis is associated with significant neurological problems.

13

However, most of these data have been collected in experimental animals and cannot be

14

transferred to humans. Moreover, it is not definitively established whether neurologic

15

diseases depend on a generic modification of the gut microbiota or whether a single

16

bacterial phylum or species plays a specific role for any single condition. Furthermore,

17

limited information exists regarding protective bacteria.

18

Conclusions: Both probiotics and prebiotics can have different impacts on CNS according

19

to the microbial species or oligosaccharides that are administered. In humans, particularly

20

in children, several factors may be important in conditioning gut microbiota modifications;

21

unfortunately, most of these factors act simultaneously. More efforts are required to fully

22

define both the array of complex behaviors that are influenced by the gut microbiota at the

23

CNS level and the mechanisms involved.

AC C

EP

TE D

M AN U

10

24 25

Key-words: attention deficit hyperactivity disorder; autism spectrum disorder; central

26

nervous system; gut microbiota; probiotics; prebiotics. 2

ACCEPTED MANUSCRIPT 27 28

INTRODUCTION Several trillions of commensal microbes live in the human gut and are collectively

30

referred to as the gut microbiota. The gut microbiota performs several functions that are

31

considered essential for health and survival. Studies have suggested that the gut

32

microbiota serves as a barrier inhibiting the proliferation of pathogenic organisms.1

33

Moreover, it contributes to the digestion of food and the breakdown of toxins and drugs,1

34

regulates lipid and glucose metabolism,2 plays a fundamental role in the induction,

35

training, and function of the host immune system,3 modulates gene expression,4 and

36

reduces inflammation.5 In addition, 20-40% of the small molecules in the peripheral blood

37

are microbial metabolites, many of which have profound effects on the development and

38

function of the central nervous system (CNS).3, 4

M AN U

SC

RI PT

29

Gut dysbiosis, i.e., a significant modification in the gut microbiota composition, has

40

been associated with several clinically relevant conditions. These conditions include

41

obesity,6 cardiovascular diseases,7 liver diseases,8 kidney diseases,9 type 1 and type 2

42

diabetes,10,

43

neurological disorders have been associated with gut dysbiosis.15 Alterations of brain

44

structure and function development are among the most relevant problems ascribed to

45

modifications of the gut microbial composition because interactions between the CNS and

46

the gut microbiota, which form the so-called gut-brain axis, appear to be already

47

established during fetal life and continue until old age.16 It has been demonstrated that

48

communication between the gastrointestinal tract and the CNS occurs continuously

49

through several routes (including hormonal, immune, and neuronal pathways) that are

50

mostly conditioned by the microbiota composition.17, 18 Consequently, dysbiosis may affect

51

the CNS, particularly during the first years of life when the developing brain is highly

52

vulnerable.16

rheumatoid arthritis,12 cancer,13 and allergic diseases.14 Moreover, several

AC C

EP

11

TE D

39

3

ACCEPTED MANUSCRIPT In this review, several aspects of the relationship between the gut microbiota and the

54

CNS during development are discussed. Particular attention is paid to factors that

55

condition the modification of the gut microbiota and the possibility of managing

56

neurological diseases by modifying the gut microbial composition. PubMed was used to

57

search for all of the studies published over the last 15 years using the key word

58

“microbiota” and “gut” or “intestinal” and “nervous system”. More than 350 articles were

59

found, and only those published in English and providing data on aspects related to

60

neurologic diseases were included in the evaluation.

SC

61

CHARACTERISTICS OF THE GUT MICROBIOTA DURING DEVELOPMENT

M AN U

62

RI PT

53

The gut microbiota’s definitive composition is mainly based on four major phyla

64

covering more than 90% of the total bacterial population (namely Firmicutes,

65

Bacteroidetes, Actinobacteria, and Proteobacteria) and include many additional minor

66

phyla such as Verrucomicrobia and Fusobacteria.18 The Firmicutes phylum is composed of

67

Gram-positive aerobic and anaerobic bacteria. Prominent members are included in the

68

genus

69

Staphylococcus, Escherichia, and Klebsiella. Bacteroidetes are Gram-negative bacteria

70

and include genus Bacteroides and Prevotella. Actinobacteria are Gram-positive bacteria,

71

among which genus Bifidobacterium, Corynebacterium, Propionibacterium, Atopobium are

72

the most frequently detected. The Proteobacteria phylum contains Gram-negative

73

bacteria, most notably the family of Enterobacteriaceae, including Enterobacter species.

74

However, this composition of the gut microbiota is achieved only at the end of the third

75

year of life. At birth and during the first months of life, the proportion of the different phyla

76

can differ markedly from subject to subject due to the influence of several factors that are

77

summarized below.18, 19 Table 1 summarizes the factors influencing gut microbiota during

78

development.

Enterococcus,

Clostridium,

Ruminococcus,

Streptococcus,

AC C

EP

Lactobacillus,

TE D

63

4

ACCEPTED MANUSCRIPT 79 80

Composition of the gut microbiota during fetal life

81

Until recently, it was thought that the fetus and intrauterine environment were sterile

82

and that the gut microbiota would develop only at birth after passage through the vaginal

83

canal or, in cases of cesarean section, after contact with environmental microbes.20,

84

However, recently collected data suggest that maternal-fetal exchange of commensal

85

bacteria may occur before birth via placental colonization. Together with the demonstration

86

of bacteria in amniotic fluid,22 umbilical cord blood,23 meconium,24 and fetal membranes,25

87

research has shown the existence of a unique placental microbiota niche composed of

88

non-pathogenic commensal microbiota from the Firmicutes and Fusobacteria phyla.26

89

Existence of placental microbiota and early colonization of the fetus through the placenta

90

was recently confirmed by Collado et al., who reported that the placenta and the amniotic

91

fluid harbor a distinct microbiota characterized by low richness, low diversity and the

92

predominance of Proteobacteria.27 Moreover, they found that the placenta and meconium

93

microbiota composition were similar, suggesting microbial transfer at the feto-maternal

94

interface.27

TE D

M AN U

SC

RI PT

21

The composition of placental and fetal gut microbiota can significantly vary,

96

according to several maternal conditions. Antibiotic therapy, malnutrition or over-nutrition,

97

obesity, diabetes, and eczema are all factors that can condition a significant modification

98

of the gut microbiota in pregnant woman with a predominance of potentially negative

99

bacteria such as Enterobacteriaceae and Pseudomonadaceae and a poor presence of

100

protective microbiota such as the Lactobacillus genus.28 Similar findings have been

101

reported in studies that have evaluated pregnant women with stress. Animal studies have

102

shown that a single 2-hour exposure to a social stressor can greatly reduce the relative

103

proportions of two genera and one family of highly abundant, probably protective, intestinal

104

bacteria, including the species of the genus Lactobacillus.29

AC C

EP

95

5

ACCEPTED MANUSCRIPT In humans, Zijlmans et al. studied the development of the infant gut microbiota over

106

the first 110 days after birth in a healthy cohort of 56 vaginally born infants.30 They

107

reported that maternal prenatal stress was strongly and persistently associated with the

108

infants' gut microbiota composition. Infants of mothers with high stress during pregnancy

109

had significantly higher relative abundances of Escherichia, Serratia, and Enterobacter

110

and lower relative abundances of lactic acid bacteria (i.e., Lactobacillus, Lactococcus,

111

Aerococcus) and Bifidobacteria. Moreover, a study by Jasarevic et al. showed that

112

maternal stress altered proteins related to vaginal immunity and abundance of species of

113

the genus Lactobacillus, the prominent taxa in the maternal vagina.31 Loss of maternal

114

vaginal Lactobacillus resulted in decreased transmission of this bacterium to the offspring.

M AN U

SC

RI PT

105

115 116

Postnatal period

Colonization after birth is strictly dependent on the duration of gestation, mode of

118

delivery, use of antibiotics and type of feeding. In term infants born to healthy mothers with

119

vaginal delivery, breastfeeding, and no antibiotic use, the gut microbiota is characterized

120

by low diversity and a relative dominance of Proteobacteria and Actinotobacteria.32

121

Bacteria found in the feces of these neonates are similar to those normally detectable in

122

the maternal vagina.33 With time, the gut microbiota becomes diverse with the emergence

123

of Firmicutes and Bacteroidetes. During the first few months, these changes are mainly

124

due to breast milk, which contains great amounts of streptococci and staphylococci.

125

Moreover, it includes many complex oligosaccharides that stimulate the growth of

126

Staphylococci and Bifidobacteria.34, 35 Further differentiation occurs after the introduction of

127

solid foods with enhanced introduction of butyrate producers, including Bacteroides and

128

Clostridium species.36 At the end of the third year, when the final composition of gut

129

microbiota is achieved, members of the Firmicutes and Bacteroidetes phyla are the most

130

common bacteria, followed by Actinobacteria, which is mainly represented by the genus

AC C

EP

TE D

117

6

ACCEPTED MANUSCRIPT 131

Bifidobacterium. Once the final composition is achieved, the gut microbiota tends to

132

remain stable.19 Infants born through cesarean section are not exposed to maternal gut

134

microorganisms and are colonized mainly by microbiota derived by their mother’s skin.37

135

Staphylococcus, Corynebacterium, and Propionibacterium species are the most common

136

colonizers, together with certain bacteria that come from environmental sources such as

137

health-care workers and the hospital environment. Colonization with Clostridium difficile is

138

more common than in vaginally delivered infants, whereas that with Bacteroides and

139

Bifidobaterium species occurs later.37 Modifications of the gut microbiota induced by

140

cesarean section persist for a long time. Longitudinal studies have shown that the delayed

141

and altered colonization pattern in cesarean-section-delivered infants persists at least until

142

the age of one year,38 and even after 7 years of age, minor differences can be detected.39

M AN U

SC

RI PT

133

The gut microbiota of the formula-fed infant differs from that of the breastfed infant

144

because it is characterized by a wider microbiota spectrum. In particular, the counts and

145

incidences of Clostridium (C. paraputrificum, C. perfringens, C. clostridiiforme, C. difficile,

146

and C. tertium) and Streptococcus (S. bovis, S. faecalis, and S. faecium) species, Bacillus

147

subtilis, Bacteroides vulgatus, Veillonella parvula, Lactobacillus acidophilus, Escherichia

148

coli, and Pseudomonas aeruginosa in bottle-fed infants are significantly higher than those

149

in breastfed infants. By contrast, Lactobacillus rhamnosus and Staphylococci prevails in

150

breast-fed infants, whereas Staphylococcus epidermidis is almost absent in samples from

151

the feces of formula-fed infants.40

AC C

EP

TE D

143

152

Premature infants have a markedly different gut microbiota from term infants, with a

153

bacilli-dominated phase, delayed or missed acquisition of Bifidobacteria, and an earlier

154

acquisition of Firmicutes.27,

155

the gut microbiota, with differences that are mainly related to the spectrum of activity of the

41

Furthermore, the use of antibiotics can significantly modify

7

ACCEPTED MANUSCRIPT 156

administered drugs.6 However, an increase in Gram-negative rods with a negative

157

metabolic influence has been frequently reported.33

158

GUT MICROBIOTA AND THE CENTRAL NERVOUS SYSTEM (CNS)

160

Importance of the normal microbiota for normal brain development

RI PT

159

The gut microbiota is essential for normal CNS development. Generally, gut

162

microbiota absence is associated with several CNS developmental problems. Diaz Heiitz

163

et al. reported that compared with conventionally raised mice, germ-free (GF) animals had

164

an increased expression of PSD-95 and synaptophysin in the striate nucleus.42-44 Because

165

these proteins are, respectively, a stimulator of excitatory synapse maturation and a

166

hallmark of synaptic vesicle maturation, these findings seem to indicate that when the

167

conventional microbiota is lacking, synaptic maturation is accelerated. This might affect

168

motor control and anxiety-like behavior in later life, as shown in GF animals.45 Hoban et al.

169

investigated changes in the homeostatic regulation of the neuronal transcription in GF

170

mice within the prefrontal cortex.46 They found a marked, concerted upregulation of genes

171

linked to myelination and myelin plasticity, leading to hypermyelinated axons.

TE D

M AN U

SC

161

Colonization of experimental animals by a conventional microbiota reversed the

173

myelin changes, confirming the necessity of intestinal flora for normal CNS development.

174

Absence of microbiota during early life increases activity-related transcriptional pathways

175

in the amygdala. Stilling et al. exploited unbiased genome-wide transcriptional profiling to

176

determine gene expression in the amygdala of GF and GF mice that were colonized after

177

weaning.47 Using RNA-sequencing and a comprehensive downstream analysis pipeline,

178

these authors studied the amygdala transcriptome and found significant differences at the

179

levels of differential gene expression, exon usage and RNA-editing. Upregulation of

180

several immediate early response genes such as Fos, Fosb, Egr2 or Nr4a1 in association

181

with increased CREB signaling in GF mice was evidenced. In addition, the differential

AC C

EP

172

8

ACCEPTED MANUSCRIPT 182

expression and recoding of several genes implicated in brain physiology processes such

183

as

184

demonstrated.

neurotransmission,

neuronal

plasticity,

metabolism

and

morphology

were

Microbes also control microglia maturation and function. GF mice display global

186

defects in microglia, with altered cell proportions and an immature phenotype.48 Because

187

microglia are formed by immune cells, this leads to impaired innate immune responses. In

188

animals without risk factors for intestinal flora modifications, temporal gut microbiota

189

eradication or limited microbiota complexity severely change the microglia properties.48

190

Moreover, colonization with a complex microbiota partially restores microglia features,

191

highlighting the role of gut microbiota in conditioning normal CNS development.

M AN U

SC

RI PT

185

192 193

Mechanisms by which the gut microbiota can interfere with central nervous system

194

(CNS) development

Interaction between the CNS and the gut microbiota involves the gut microbes, the

196

brain, the endocrine system, the immune system, and the autonomic nervous system

197

(Table 2). Signals from the brain can influence the motor, sensory, and secretory

198

modalities of the gastrointestinal tract, and conversely, visceral messages from the

199

gastrointestinal tract can influence brain function.49 Despite the exact mechanisms by

200

which the gut and brain reciprocally influence structure and function not being precisely

201

defined, several data indicate that the passage of certain bacterial components through

202

the intestinal wall plays a fundamental role in this regard. The most important bacterial

203

components are lipopolysaccharides in the cell wall of some Gram-negative bacteria (such

204

as Enterobacteriaceae and Pseudomonadaceae); the lipopolysaccharides may translocate

205

from the intestinal mucosa to the systemic circulation, where they can affect immune

206

regulation and CNS function.50 Lipopolysaccharides act upon toll-like receptor 4 to activate

207

the systemic inflammation response with the production of pro-inflammatory cytokines

AC C

EP

TE D

195

9

ACCEPTED MANUSCRIPT 208

such as tumor necrosis factor-α (TNF-α), interleukin(IL)-6, and IL-1β.51 These cytokines, in

209

turn, critically affect the CNS.52 By contrast, other bacteria, such as those included in the

210

genus Lactobacillus, seem to exert a protective effect, probably because they secrete

211

chemicals that limit colonization by inflammatory microorganisms.53 Microbial metabolites, mainly gut-derived tryptophan metabolites, can have a critical

213

importance. These metabolites are capable of modulating the immune system by various

214

mechanisms, including the regulation of T cells, notably Th17 lymphocytes.54, 55 Moreover,

215

high levels of tryptophan metabolites increase kynurenine concentration, which can also

216

engage aryl hydrocarbon receptors. This induces the differentiation of regulatory T cells

217

and increases the levels of indoleamine 2,3-dioxygenase, an enzyme that further induces

218

the catabolism of tryptophan and an increase in the amount of circulating metabolites of

219

this amino acid.56 In mice, it has been demonstrated that even when no transfer of the

220

microbiota to the fetus from the mother has occurred, tryptophan metabolites that cross

221

the placenta are active on the aryl hydrocarbon receptors and can significantly influence

222

CNS development of the fetus.57 Finally, the level of systemic tryptophan metabolites

223

influences the concentration of serotonin and γ- aminobutyric acid the CNS and the

224

production of neurotoxic molecules in astrocytes and microglia.58, 59

EP

TE D

M AN U

SC

RI PT

212

A further modulation of CNS function is provided by short chain fatty acids (SCFAs)

226

produced by bacterial fermentation of complex polysaccharides (starches and fibers) in the

227

colon.60 Particularly, Clostridia and Bacteroides are important SCFA producers. Major

228

effects of these compounds are the alteration of mitochondrial function via the citric acid

229

cycle and carnitine metabolism, or the epigenetic modulation of genes controlling brain

230

function.

231

neuroinflammatory, metabolic, and epigenetic changes. MacFabe et al. have shown that

232

when propionic acid or other short-chain fatty acids are injected into the cerebral ventricles

AC C

225

All

these

factors

can

produce

reversible

behavioral,

electrographic,

10

ACCEPTED MANUSCRIPT 233

of rats, the rats show biologic, chemical, and pathologic changes that are characteristic of

234

autism.61

235

It

has

also

been

suggested

that

intestinal

bacteria

can

affect

fetal

neurodevelopment by influencing 5-hydroxytryptamine (5-HT) serum levels.62 Indigenous

237

spore-forming bacteria from the microbiota promote 5-HT biosynthesis from colonic

238

enterochromaffin cells (ECs), which supply 5-HT to the mucosa, lumen, and circulating

239

platelets. Yano et al. identified select fecal metabolites that are increased by spore-forming

240

bacteria and that elevate 5-HT in chromaffin cell cultures, suggesting direct metabolic

241

signaling of gut microbes to ECs.62 Furthermore, elevating luminal concentrations of

242

particular microbial metabolites increases colonic and blood 5-HT in GF mice and,

243

consequently, fetal neuronal cell division and differentiation, physiologically regulated by

244

this compound.63 Depletions of 5-HT during development were reported to have effects on

245

the maturation of cortical neurons64, 65 and to alter barrel cortex development.66. 67

M AN U

SC

RI PT

236

A further mechanism with which gut microbiota can influence CNS is through the

247

endocrine pathway. Prenatal stress is accompanied by increased cortisol levels, mainly via

248

the systemic and peripheral release of corticotropin-releasing factor (CRF) from the

249

amygdala, which is important in the control of emotional and autonomic responses to

250

stress and contains CRF nerve terminals, CRF cell bodies, and CRF receptors.68

251

Hypercortisolemia increases gut permeability and favors bacterial lipopolysaccharide

252

leakage across the gut wall.69 Moreover, it is associated with an increased amygdala

253

volume70 and this with depression.71

AC C

EP

TE D

246

254

Modification of the blood-brain barrier (BBB) can play a role in conditioning

255

alterations in CNS development. The BBB acts as a gatekeeper to control the passage

256

and exchange of molecules and nutrients between the circulatory system and the brain

257

parenchyma. The BBB also ensures homeostasis of the CNS. Braniste et al. reported that

258

GF mice, beginning with intrauterine life, display increased BBB permeability compared 11

ACCEPTED MANUSCRIPT 259

with non-GF mice with a normal gut microbial composition.72 The increased BBB

260

permeability is maintained in GF mice after birth and during adulthood and is associated

261

with reduced expression of the tight junction proteins occludin and claudin-5, which are

262

known to regulate BBB function in endothelial tissues. Finally, the gut microbiota interacts with intestinal cells to stimulate the production of

264

peptides that activate afferent endings of the vagus nerve. The resultant signals are

265

transmitted to the CNS, affecting behavior and efferent neural activity. The same function

266

appears to be exerted by pro-inflammatory cytokines. Consequently, the brain receives

267

information regarding systemic inflammation, contributing to affective symptoms and

268

initiating behavioral responses, including depression and other sickness behaviors.73

269

Vagal signals from the gut are also thought to be responsible for “gut feelings,” which act

270

as signals to the brain that an environment may be threatening or anxiety provoking.74

271

Activated efferent fibers of the vagus, in turn, carry anti-inflammatory signals to the

272

periphery, reducing the production of proinflammatory cytokines.75

TE D

M AN U

SC

RI PT

263

If the gut microbiota plays an essential role in the modulation of brain structure and

274

function, it is clear that when unbalance between “positive” and “negative” bacteria occurs

275

and the latter microbes prevail, inflammation arises, chronic immune disease can occur,

276

and the CNS structure and function may be altered.50 Moreover, when chronic dysbiosis

277

occurs, a systemic inflammatory state tends to persist.

279

AC C

278

EP

273

IMPACT OF DYSBIOSIS ON THE CENTRAL NERVOUS SYSTEM (CNS)

280

The potential clinical effects of dysbiosis on the CNS are significantly more

281

important during the developmental period. Younger individuals can suffer from relevant

282

and permanent modification of CNS structure and function.43 However, even older

283

subjects, despite a complete development of the CNS, can have associated psychiatric

284

problems or so-called sickness behaviors, including fatigue, insomnia, lack of appetite and 12

ACCEPTED MANUSCRIPT 285

depression.76,

77

286

disease78 and Alzheimer’s disease79 have been associated with gut microbiota

287

modifications.

Moreover, also neurodegenerative disorders, such as Parkinson’s

All the conditions that can cause gut dysbiosis have been associated with

289

impairment of the CNS structure and function. However, in some instances, the data

290

collected in humans have been conflicting. Moreover, in cases exhibiting simultaneous

291

conditions, the importance of the various factors is difficult to ascertain.

RI PT

288

Pregnant women with an unhealthy alteration or imbalance in the microbial

293

composition frequently have higher circulating levels of pro-inflammatory cytokines such

294

TNF-α, IL-6, and IL-1β and can produce sons with behavioral problems.6, 80 Infants born to

295

stressed mothers frequently exhibit increased impulsivity, anxiety problems, attention

296

deficit hyperactivity disorder (ADHD) and autism spectrum disorders (ASD).81-85

M AN U

SC

292

Studies employing animal models have revealed that delivery by cesarean section

298

is frequently associated with alterations of the mesolimbic and mesostriatal dopamine

299

pathway, with persistent blunting of stress-induced dopamine release in the right prefrontal

300

cortex.86 Moreover, compared with vaginally delivered animals, those delivered by

301

cesarean section suffer more frequently from behavioral abnormalities that have been

302

associated with ASD.87 However, the importance of the cesarean section in conditioning

303

negative effects on the CNS is debated. A meta-analysis by Curran et al., who reviewed

304

the literature published up to 28 February 2014, concluded that children born through

305

cesarean section had a 23% increased risk of developing ASD, compared with those born

306

by vaginal delivery.88 However, more recently, the same authors reported contrary

307

conclusions in two different studies. In the first study, they enrolled sibling pairs discordant

308

with respect to ASD status and found that subjects born via elective or emergency

309

cesarean section were approximately 20% more likely to be diagnosed as having ASD

310

compared with those with unassisted vaginal delivery.89 However, in the sibling control

AC C

EP

TE D

297

13

ACCEPTED MANUSCRIPT analysis, neither elective nor emergency cesarean section was associated with ASD,

312

implying that this association was likely due to familial confounding factors (genetic and/or

313

environmental) rather than the mode of delivery. In the second study, Curran et al.

314

evaluated a nationally representative UK cohort of 13,141 children and did not find any

315

association between cesarean section and ASD or ADHD.90 However, more favorable for

316

a possible negative impact of cesarean section are the data collected by Adler and Wong-

317

Kee-You.91 They explored whether differences in spatial attention would occur in infants

318

delivered by cesarean or vaginal section. Three-month-old infants performed either a

319

spatial cueing task or a visual expectation task. The cesarean-section-delivered infants'

320

stimulus-driven, reflexive attention was slower than that of the vaginally delivered infants,

321

whereas their cognitively driven, voluntary attention was unaffected. Based on the authors’

322

conclusions, these findings suggest that the types of birth experience influence at least

323

one form of infant attention, and possibly any cognitive process that relies on spatial

324

attention.

TE D

M AN U

SC

RI PT

311

Conflicting results were also reported in studies regarding breastfeeding and

326

cognitive development. In the USA, a national cohort study of 5,475 children of normal

327

birth weight, having ever being breastfed, was associated with a significant higher child

328

intelligence quotient (IQ).92 However, the positive effect of breastfeeding was no longer

329

evident after an adjustment for confounders, mainly maternal intelligence. On the contrary,

330

in UK, even after adjusting for confounders, a significant difference in the mean score of

331

cognitive development between children who were breastfed and children who were never

332

breastfed was evidenced. The differences were greater in children with long-term

333

breastfeeding and in premature infants.93 Similarly, in Asia, Cai et al. demonstrated that

334

higher breastfeeding exposure was associated with better memory at 6 months,94 as

335

demonstrated by increased preferential gazing toward correctly matched items during the

336

early portions of a relational memory task. At 24 months of age, breastfed children were

AC C

EP

325

14

ACCEPTED MANUSCRIPT 337

more likely to display sequential memory during a deferred imitation memory task, and

338

toddlers with more exposure to breastfeeding scored higher in receptive language as well

339

as expressive language. Further data suggesting a positive impact of breastfeeding on infants’ neurological

341

development have been collected by Krol et al.95 These authors examined whether and

342

how the duration of exclusive breastfeeding affected the neural processing of emotional

343

signals by measuring electro-cortical responses to body expressions in 8-month-old

344

infants. The analyses revealed that infants with high-exclusive breastfeeding showed a

345

significantly greater neural sensitivity to happy body expressions than did those with low-

346

exclusive breastfeeding. Moreover, regression analyses revealed that the neural bias

347

toward happiness or fearfulness differed as a function of the duration of exclusive

348

breastfeeding. Specifically, a longer duration was associated with a happy bias, whereas a

349

shorter breastfeeding duration was associated with a fearful bias.

M AN U

SC

RI PT

340

ASD is a neurologic condition for which a strict relationship with gut dysbiosis has

351

been repeatedly hypothesized because, in children with this condition, particularly in those

352

with associated gastrointestinal dysfunctions (i.e., altered bowel habits, chronic abdominal

353

pain, reflux and vomiting), several studies have shown a frequent significant modification

354

of the gut microflora.96, 97 Finegold et al. compared the gut microflora of children exhibiting

355

ASD of various severities with the gut microflora of healthy controls and reported that at

356

the phylum level, Bacteroidetes were found at higher concentrations in the severely

357

autistic group while Firmicutes were more predominant in the control group.98 Smaller but

358

significant differences also occurred in the Actinobacterium and Proteobacterium phyla.

359

Desulfovibrio species and Bacteroides vulgatus were detected more commonly in the

360

stools of severely autistic children than in controls. However, in several cases (those with

361

late-onset ASD), 10-fold higher levels of Clostridium species, included in the Firmicutes

362

phylum, were observed.99

AC C

EP

TE D

350

15

ACCEPTED MANUSCRIPT 363

Recent

studies

have

evidenced

a

possible

relationship

between

some

neurodegenerative disorders that usually occur in adults or in old people such as

365

Parkinson's and Alzheimer’s diseases and gut dysbiosis. Interestingly, it has been reported

366

that in patients with Parkinson's disease the abundance of Prevotellaceae in feces was

367

reduced by 77.6% as compared with controls.100 The relative abundance of

368

Enterobacteriaceae was positively associated with the severity of postural instability and

369

gait difficulty suggesting not only that the intestinal microbiota is altered in Parkinson’s

370

disease but that it is related to motor phenotype.100

SC

RI PT

364

Regarding Alzheimer’s disease, it has been demonstrated that in germ-free mouse

372

models a drastic reduction of cerebral amyloid pathology could be demonstrated when

373

these animals were compared to control Alzheimer’s disease animals with intestinal

374

microbiota. Sequencing bacterial 16S rRNA from fecal samples revealed a remarkable

375

shift in the gut microbiota of conventionally raised Alzheimer’s disease mice as compared

376

to healthy, wild-type mice.101 Colonization of germ-free Alzheimer mice with harvested

377

microbiota from conventionally-raised Alzheimer mice dramatically increased cerebral Aβ

378

pathology. In contrast, colonization with microbiota from control wild-type mice was

379

ineffective in increasing cerebral amyloid level, suggesting a microbial involvement in the

380

development of Alzheimer’s disease pathology.101

382 383

TE D

EP

AC C

381

M AN U

371

THERAPEUTIC MANIPULATION OF THE GUT MICROBIOTA

384

The evidence that gut dysbiosis can be associated with significant alterations of

385

CNS structure and function has led to the supposition that gut microbiota manipulation

386

might be a rational approach to limit neurologic clinical problems.

387

Attempts to normalize the gut microbiota by treating children with ASD and

388

gastrointestinal problems with specific antibiotics active against Clostridium species have 16

ACCEPTED MANUSCRIPT been suggested.102 A significant reduction in these bacteria, which have been

390

demonstrated to have a negative effect on the CNS and were found in higher

391

concentrations in children with ASD, was proposed as an option. Oral vancomycin was

392

considered the best solution because this antibiotic is minimally absorbed by the intestinal

393

tract. Unfortunately, this attempt had only partial success. Administration of the antibiotic to

394

children with regressive-onset ASD was followed by a significant improvement of

395

neurological conditions, but only during the period of drug use.84 Neurological conditions

396

returned to baseline shortly after treatment suspension.

SC

RI PT

389

A second approach was the administration of probiotics and/or prebiotics. Probiotics

398

are live microorganisms that when administered in adequate amounts, confer a health

399

benefit to the host.103 Studies have revealed that many of these bacteria can modify the

400

bacterial gut composition, with a reduction of negative strains and an increase in those

401

considered protective. Moreover, probiotics can reduce the concentration of certain

402

bacterial products that once they have crossed the intestinal wall, cause inflammation,

403

immune system alterations, modulation of gene expression and modification of the CNS

404

structure and function.1044 However, the beneficial effects are divergent and are dependent

405

on the strain. Bifidobacterium breve NCIMB 702258 and Bifidobacterium breve DPC 6330

406

were found to have contrasting effects on the gut microbiota. Compositional sequencing of

407

the gut microbiota showed a tendency for greater proportions of Clostridiaceae (25%,

408

12%, and 18%; p=0.08) and lower proportions of Eubacteriaceae (3%, 12%, and 13%;

409

p=0.06) in mice supplemented with Bifidobacterium breve DPC 6330 than in mice

410

supplemented with the second microorganism and unsupplemented controls.105

AC C

EP

TE D

M AN U

397

411

However, studies in the experimental animals have shown that the administration of

412

selected probiotics to animals may be effective in reducing neurologic signs and symptoms

413

associated with gut dysbiosis. Chronic administration of Bifidobacterium infantis to rats

414

deprived of maternal contact at an early age and having stress-related gastrointestinal and 17

ACCEPTED MANUSCRIPT mood disorders results in normalization of the immune response, reversal of behavioral

416

deficits, and restoration of basal noradrenaline concentrations in the brainstem.106

417

Moreover, Bifidobacterium longum NCC3001 normalizes anxiety-like behavior and

418

hippocampal brain derived neurotrophic factor (BDNF) in mice with infectious colitis. 107 In

419

rats, the administration of Lactobacillus helveticus and Lactobacillus rhamnosus can

420

prevent

421

demonstrated with Lactobacillus farciminis.109 Hsiao et al. demonstrated gastrointestinal

422

barrier defects and microbiota alterations in the maternal immune activation (MIA) mouse

423

model, which is known to display features of ASD.110 Oral treatment of the offspring with

424

the human commensal Bacteroides fragilis corrected the gut permeability, altered the

425

microbial composition, and ameliorated the defects in communicative, stereotypic, anxiety-

426

like and sensorimotor behaviors. MIA offspring displayed an altered serum metabolomic

427

profile, and Bacteroides fragilis modulated the levels of several metabolites. One

428

metabolite of particular interest was 4-ethylphenylsulfate (4EPS). 4EPS is thought to be a

429

uremic toxin, as is p-cresol (4-methylphenol), a chemically related metabolite reported to

430

be a possible urinary biomarker for ASD.111 This study showed a 46-fold increase in 4EPS

431

in the MIA model and normalization upon Bacteroides fragilis treatment. Moreover, the

432

treatment of mice with 4EPS potassium salt was sufficient to induce anxiety (but not

433

autism)-like behaviors similar to that of the MIA offspring.

intestinal

abnormalities.108 The

same

effect

was

AC C

EP

TE D

M AN U

SC

chronic-stress-induced

RI PT

415

434

Unfortunately, studies in humans are scarce and have not always yielded

435

satisfactory results, highlighting the difficulties in extrapolating experimental findings to

436

human physiology. Daily consumption of a fermented milk product with probiotics

437

(Bifidobacterium animalis subspecies lactis, Streptococcus thermophiles, Lactobacillus

438

bulgaricus, and Lactococcus lactis subspecies lactis) for 4 weeks was associated with a

439

significant modification of the activity of CNS regions that control emotion and

440

sensation.112 Moreover, it was shown that probiotic supplementation early in life might 18

ACCEPTED MANUSCRIPT reduce the risk of neuropsychiatric disorder development later in childhood.113 Seventy-five

442

infants who were randomized to receive Lactobacillus rhamnosus GG or placebo during

443

the first 6 months of life were followed for 13 years. At this age, ADHD was diagnosed in

444

6/35 (17.1%) children in the placebo group and none in the probiotic group (p=0.008). The

445

mean (standard deviation) numbers of Bifidobacterium species bacteria in feces during the

446

first 6 months of life were significantly lower in affected children [8.26 (1.24) log cells/g]

447

than in healthy children [9.12 (0.64) log cells/g; p=0.03)]. A positive effect on CNS function

448

was also demonstrated by Romeo et al., who examined the administration of Lactobacillus

449

reuteri or Lactobacillus rhamnosus to preterm infants for 6 weeks or until discharge.114 The

450

neurological structured assessment performed at 1 year of age revealed a significantly

451

lower incidence of neurological abnormalities in children receiving probiotics than in

452

untreated controls. By contrast, the administration of a mixture of Lactobacillus acidophilus

453

and Bifidobacteria infantis to preterm very-low-birth-weight infants until discharge from the

454

hospital (approximately 45 days) did not modify the psychometric parameters measured at

455

3 years corrected age in comparison with control subjects not receiving probiotic

456

supplementation.115

TE D

M AN U

SC

RI PT

441

Very few studies have evaluated the effects of probiotics on ASD clinical features in

458

humans. Parracho et al. investigated the potential of Lactobacillus plantarum WCSF1 to

459

modulate the gut microbiota of autistic subjects through a double-blind, placebo-controlled,

460

crossover-designed feeding study.116 The fecal microbiota, gut function and behavior

461

scores of subjects were examined throughout the 12-week study. Probiotic administration

462

significantly increased the Lactobacilli and Enterococci group and significantly reduced

463

Clostridium cluster XIVa compared with placebo. In addition, probiotic feeding caused

464

significant differences in behavior scores compared to baseline. Another study reported

465

that probiotic administration can induce significant metabolic modifications considered

466

indicative of the disruption of gut dysbiosis but not marginal behavioral changes such as

AC C

EP

457

19

ACCEPTED MANUSCRIPT 467

improvements in the ability to concentrate and carry out orders.1107 Considering the need

468

for further studies able to definitively establish whether probiotics can reduce ASD

469

symptoms, a very well-designed protocol for a large, definitive, randomized controlled trial

470

has recently been proposed.118 Prebiotic oligosaccharides are indigestible nutritional constituents that are purported

472

to have antimicrobial, immunomodulatory and anti-inflammatory properties.119 These

473

effects are attributable to a direct effect on the gut microbiota because they promote the

474

growth of Bifidobacterium species and consequently reduce (through competition)

475

colonization by pathogenic flora. Moreover, they can direct interact with gut cells, assuring

476

the integrity of the intestinal barrier.120 In rodents, in which the absence of gut bacteria is

477

associated with decreased central expression of brain derived neurotropic factor (BDNF)

478

and N-methyl-d-aspartate receptor subunits, oral probiotics increase brain BDNF and

479

impart associated anxiolytic effects.121 Moreover, prebiotic administration normalizes

480

lipopolysaccharide-induced anxiety and the cortical 5-HT2A receptor and IL1-β levels.122

481

Finally, neonatal prebiotic supplementation seems to modify neurotransmission with a

482

long-duration effect persisting until the adult age.123 As with probiotics, prebiotic studies

483

regarding the impact of prebiotic use in humans with neurological problems are few and

484

not conclusive. In adult volunteers, the administration of galactooligosaccharides was

485

associated with a reduction of the waking cortisol response and significant modification of

486

emotional bias, suggesting a potential direct effect on brain function. However, in the same

487

studies, a different prebiotic had no effect, suggesting that as with probiotics, the choice of

488

the right compound is critical to obtain satisfactory modification of the CNS activity.124

AC C

EP

TE D

M AN U

SC

RI PT

471

489

Finally, dietary modification has been considered, and animal studies have been

490

conducted. In most of these instances, certain effects on the gut microbiota were shown,

491

with favorable modifications of CNS function. For example, in rats, ingestion of a diet with

492

a high beef content was associated with the exclusive presence in the gut of certain 20

ACCEPTED MANUSCRIPT 493

bacterial genera, including Proteus, Serratia, Sarcina, and Staphylococcus. These animals

494

displayed increased learning and memory behaviors as well as decreased levels of

495

anxiety-like behavior.125 However, no relevant data have been collected in humans.

496

CONCLUSIONS

RI PT

497

Data seem to indicate that the gut microbiota influences CNS development and

499

function and that gut dysbiosis is associated with significant neurological problems.

500

However, most of these results have been collected in experimental animals and cannot

501

be transferred to humans. The human gut-brain axis fundamentally differs from the rodent

502

axis primarily because of the great expansion of the prefrontal cortex and the frontoinsular

503

regions, which play a major role in human emotional regulation. Moreover, it is not

504

definitively established whether neurologic diseases depend on a generic modification of

505

the gut microbiota or whether a single bacterial phylum or species plays a specific role for

506

any single condition. Furthermore, limited information exists regarding protective bacteria.

507

The evidence that both probiotics and prebiotics can have different impacts according to

508

the microbial species or oligosaccharides that are administered indicates the difficulty of

509

evaluating the relationship between bacteria and the CNS, as well as the best therapeutic

510

approach. In humans, particularly in children, several factors may be important in

511

conditioning gut microbiota modifications; unfortunately, most of these factors act

512

simultaneously and can induce different gut microbiota alterations. More efforts are

513

required to fully define the array of complex behaviors that are influenced by the gut

514

microbiota at the CNS level, as well as the mechanisms involved. This seems particularly

515

important for younger children, who are at greater risk of significant damage to CNS

516

structure and function.

AC C

EP

TE D

M AN U

SC

498

517 518 21

ACCEPTED MANUSCRIPT 519

ACKNOWLEDGMENTS

521

This review was partially supported by a grant from the Italian Ministry of Health

522

(Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico Ricerca Corrente 2016

523

850/01).

RI PT

520

524

AC C

EP

TE D

M AN U

SC

525

22

ACCEPTED MANUSCRIPT 526 527

REFERENCES

528

1)

529

Bioengineered 2016; 7: 11-20.

530

2)

531

microbiota as a target in the pathogenesis of metabolic disorders: a new approach to novel

532

therapeutic agents. Horm Metab Res 2016; 48: 349-358.

533

3)

534

2014; 157:121-141.

535

4)

536

2014; 71: 1045-1054.

537

5)

538

microbiota shapes rheumatic diseases. Nat Rev Rheumatol 2016; 12: 398-411.

539

6)

540

children. Int J Antimicrob Agents 2016; 47:1771-1779.

541

7)

542

-dietary pro- and prebiotics for the management of cardiovascular risk. Proc Nutr Soc

543

2014; 73: 172-185.

544

8)

545

Hepatol 2014; 29: 1139-1148.

546

9)

547

disease. Exp Physiol 2016; 101: 471-477.

548

10)

549

Hagen M, et al. Compromised gut microbiota networks in children with anti-islet cell

550

autoimmunity. Diabetes 2014; 63: 2006-2014.

Linares DM, Ross P, Stanton C. Beneficial microbes: the pharmacy in the gut.

RI PT

Ejtahed HS, Soroush AR, Angoorani P, Larijani B, Hasani-Ranjbar S. Gut

SC

Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell

M AN U

Takahashi K. Influence of bacteria on epigenetic gene control. Cell Mol Life Sci

Van de Wiele T, Van Praet JT, Marzorati M, Drennan MB, Elewaut D. How the

TE D

Principi N, Esposito S. Antibiotic administration and the development of obesity in

EP

Tuohy KM, Fava F, Viola R. The way to a man's heart is through his gut microbiota'-

AC C

Goel A, Gupta M, Aggarwal R. Gut microbiota and liver disease. J Gastroenterol

Wing MR, Patel SS, Ramezani A, Raj DS. Gut microbiome in chronic kidney

Endesfelder D, Zu Castell W, Ardissone A, Davis-Richardson AG, Achenbach P,

23

ACCEPTED MANUSCRIPT 551

11)

Tilg H, Moschen AR. Microbiota and diabetes: an evolving relationship. Gut 2014;

552

63: 1513-1521.

553

12)

554

4244-4249.

555

13)

556

reveals role in colon tumorigenesis. mSphere 2015; 1 pii: e00001-15.

557

14)

558

gut microbiota in the development of allergic diseases. Curr Opin Allergy Clin Immunol

559

2016; Epub May 31.

560

15)

561

dysbiosis to altered brain function and mental illness: mechanisms and pathways. Mol

562

Psychiatry 2016; 21: 738-748.

563

16)

564

acquired in early life regulates physiology, brain and behaviour. Acta Paediatr. 2014; 103:

565

812-819.

566

17)

567

microbiome: implications for infant health and neurocognitive development. Nurs Res

568

2016; 65: 76-88.

569

18)

570

cells: a review of their role in brain-gut communication. Neurogastroenterol Motil 2016; 28:

571

620-630.

572

19)

573

composition of the gut microbiota throughout life, with an emphasis on early life. Microb

574

Ecol Health Dis 2015; 26: 26050.

575

20)

576

infant intestinal microbiota. PLoS Biol 2007; 5: e177.

Taneja V. Arthritis susceptibility and the gut microbiome. FEBS Lett 2014; 588:

RI PT

Zackular JP, Baxter NT, Chen GY, Schloss PD. Manipulation of the gut microbiota

SC

Simonyte Sjödin K, Vidman L, Rydén P, West CE. Emerging evidence of the role of

M AN U

Rogers GB, Keating DJ, Young RL, Wong ML, Licinio J, Wesselingh S. From gut

TE D

Clarke G, O'Mahony SM, Dinan TG, Cryan JF. Priming for health: gut microbiota

EP

Yang I, Corwin EJ, Brennan PA, Jordan S, Murphy JR, Dunlop A. The Infant

AC C

Latorre R, Sternini C, De Giorgio R, Greenwood-Van Meerveld B. Enteroendocrine

Rodríguez JM, Murphy K, Stanton C, Ross RP, Kober OI, Juge N, et al. The

Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO. Development of the human

24

ACCEPTED MANUSCRIPT 577

21)

Biasucci G, Rubini M, Riboni S, Morelli L, Bessi E, Retetangos C. Mode of delivery

578

affects the bacterial community in the newborn gut. Early Hum Dev 2010; 86(Suppl. 1): 13-

579

15.

580

22)

581

the polymerase chain reaction in the amniotic fluid of patients with cervical insufficiency. J

582

Perinat Med 2010; 38: 261-268.

583

23)

584

Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by

585

cesarean section. Curr Microbiol 2005; 51: 270-274.

586

24)

587

meconium from healthy newborns actually sterile? Res Microbiol 2008; 159: 187-193.

588

25)

589

and inflammatory cells in fetal membranes do not always cause preterm labor. Pediatr Res

590

2005; 57: 404-411.

591

26)

592

harbors a unique microbiome. Sci Transl Med 2014; 6: 237-265.

593

27)

594

may be initiated in utero by distinct microbial communities in the placenta and amniotic

595

fluid. Sci Rep 2016; 6: 23129.

596

28)

597

et al. Gut microbiota composition is associated with body weight, weight gain and

598

biochemical parameters in pregnant women. Br J Nutr 2010; 104: 83-92.

599

29)

600

commensal microbiota and host inflammation. Gut Microbes 2014; 5: 390-396.

RI PT

Oh KJ, Lee SE, Jung H, Kim G, Romero R, Yoon BH. Detection of ureaplasmas by

SC

Jiménez E, Fernández L, Marín ML, Martín R, Odriozola JM, Nueno-Palop C, et al.

M AN U

Jimenez E, Marin ML, Martin R, Odriozola JM, Olivares M, Xaus J, et al. Is

TE D

Steel JH, Malatos S, Kennea N, Edwards AD, Miles L, Duggan P, et al. Bacteria

Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta

AC C

EP

Collado MC, Rautava S, Aakko J, Isolauri E, Salminen S. Human gut colonisation

Santacruz A, Collado MC, García-Valdés L, Segura MT, Martín-Lagos JA, Anjos T,

Galley JD, Bailey MT. Impact of stressor exposure on the interplay between

25

ACCEPTED MANUSCRIPT 601

30)

Zijlmans MA, Korpela K, Riksen-Walraven JM, de Vos WM, de Weerth C. Maternal

602

prenatal

603

Psychoneuroendocrinology 2015; 53: 233-245.

604

31)

605

microbiome by maternal stress are associated with metabolic reprogramming of the

606

offspring gut and brain. Endocrinology 2015; 156: 3265-3276.

607

32)

608

ofestablishing the gut microbiota in infancy has consequences for future health. Gut

609

Microbes 2015; 6: 321-325.

610

33)

611

microbiota and disease. Trends Mol Med 2016; 22: 458-478.

612

34)

613

oligosaccharides promote the growth of staphylococci. Appl Environ Microbiol 2012; 78:

614

4763-4770.

615

35)

616

oligosaccharides on microbiota of infants: opportunities for formulas. Annu Rev Food Sci

617

Technol 2011; 2: 331-351.

618

36)

619

Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad

620

Sci U S A 2011; 108 Suppl. 1: 4578-4585.

621

37)

622

al. Delivery mode shapes the acquisition and structure of the initial microbiota across

623

multiple body habitats in newborns. Proc Natl Acad Sci U S A 2010; 107: 11971-11975.

624

38)

625

Dynamics and stabilization of the human gut microbiome during the first year of life. Cell

626

Host Microbe 2015; 17: 690-703.

stress

is

associated

with

the

infant

intestinal

microbiota.

RI PT

Jašarević E, Howerton CL, Howard CD, Bale TL. Alterations in the vaginal

SC

Dogra S, Sakwinska O, Soh SE, Ngom-Bru C, Brück WM, Berger B, et al. Rate

M AN U

Becattini S, Taur Y, Pamer EG. Antibiotic-induced changes in the intestinal

TE D

Hunt KM, Preuss J, Nissan C, Davlin CA, Williams JE, Shafii B, et al. Human milk

EP

Chichlowski M, German JB, Lebrilla CB, Mills DA. The influence of milk

AC C

Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, et al.

Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, et

Backhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, et al.

26

ACCEPTED MANUSCRIPT 627

39)

Salminen S, Gibson GR, McCartney AL, Isolauri E. Influence of mode of delivery on

628

gut microbiota composition in seven year old children. Gut 2004; 53:1388-1389.

629

40)

630

shaping in newborns. Front Cell Infect Microbiol 2012; 2: 94.

631

41)

632

bifidobacterial colonization in preterm infants: a prospective analysis. J Pediatr

633

Gastroenterol Nutr 2007; 44: 577-582.

634

42)

635

influencing gastrointestinal tract and microbiota immune interaction in preterm infants.

636

Pediatr Res 2015; 77: 726-731.

637

43)

638

microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A 2011;

639

108: 3047-3052.

640

44)

641

involvement in maturation of excitatory synapses. Science 2000; 290: 1364-1368.

642

45)

643

and depression. Trends Neurosci 2015; 36: 305-312.

644

46)

645

Regulation of prefrontal cortex myelination by the microbiota. Transl Psychiatry 2016; 6:

646

e774.

647

47)

648

Microbes & neurodevelopment - Absence of microbiota during early life increases activity-

649

related transcriptional pathways in the amygdala. Brain Behav Immun 2015; 50: 209-220.

650

48)

651

Host microbiota constantly control maturation and function of microglia in the CNS. Nat

652

Neurosci 2015; 18: 965-977.

Guaraldi F, Salvatori G. Effect of breast and formula feeding on gut microbiota

RI PT

Butel MJ, Suau A, Campeotto F, Magne F, Aires J, Ferraris L, et al. Conditions of

M AN U

SC

Collado MC, Cernada M, Neu J, Perez-Martinez G, Gormaz M, Vento M. Factors

Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A. Normal gut

TE D

El-Husseini AE, Schnell E, Chetkovich DM, Nicoll RA, Bredt DS. PSD-95

EP

Foster JA, Neufeld KAM. Gut–brain axis: how the microbiome influences anxiety

AC C

Hoban AE, Stilling RM, Ryan FJ, Shanahan F, Dinan TG, Claesson MJ, et al.

Stilling RM, Ryan FJ, Hoban AE, Shanahan F, Clarke G, Claesson MJ, et al.

Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, et al.

27

ACCEPTED MANUSCRIPT 653

49)

O’Mahony SM, Hyland NP, Dinan TG, Cryan JF. Maternal separation as a model of

654

brain-gut axis dysfunction. Psychopharmacology (Berl.) 2011; 214: 71-88.

655

50)

656

2013; 69: 87-113.

657

51)

658

Neuroinflammation 2013; 10: 46.

659

52)

660

and neurodevelopmental windows: implications for brain disorders. Trends Mol Med 2014;

661

20: 509-518.

662

53)

663

depression during pregnancy: systematic review. Obstet Gynecol 2004; 103: 698-709.

664

54)

665

receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity

666

2012; 36: 92-104.

667

55) Veldhoen M, Hirota K, Westendorf AM, Buer J, Dumoutier L, Renauld JC, et al. The

668

aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins.

669

Nature 2008; 453: 106-109.

670

56) Maes M, Rief W. Diagnostic classifications in depression and somatization should

671

include biomarkers, such as disorders in the tryptophan catabolite (TRYCAT) pathway.

672

Psychiatry Res 2012; 196: 243-249.

673

57) Gomez de Agüero M, Ganal-Vonarburg SC, Fuhrer T, Rupp S, Uchimura Y, Li H, et al.

674

The maternal microbiota drives early postnatal innate immune development. Science.

675

2016; 351: 1296-1302.

676

58) Nguyen N, Nakahama T, Le D, Van Son L, Chu H, Kishimoto T. Aryl hydrocarbon

677

receptor and kynurenine: recent advances in autoimmune disease research. Front

678

Immunol 2014; 5: 551.

Bengmark S. Gut microbiota, immune development and function. Pharmacol Res

RI PT

Theoharides TC, Asadi S, Patel A. Focal brain inflammation and autism. J

SC

Borre YE, O'Keeffe GW, Clarke G, Stanton C, Dinan TG, Cryan JF, et al. Microbiota

M AN U

Bennett HA, Einarson A, Taddio A, Koren G, Einarson TR. Prevalence of

AC C

EP

TE D

Qiu J, Heller JJ, Guo X, Chen ZM, Fish K, Fu YX, et al. The aryl hydrocarbon

28

ACCEPTED MANUSCRIPT 679

59) Barrett E, Ross R, O’Toole P, Fitzgerald G, Stanton C. γ-Aminobutyric acid production

680

by culturable bacteria from the human intestine. J Appl Microbiol 2012; 113: 411-417.

681

60) Wong JM, de Souza R, Kendall CW, Emam A, Jenkins DJ. Colonic health:

682

fermentation and short chain fatty acids. J Clin Gastroenterol 2006; 40: 235-243.

683

61)

684

mitochondria, and mind: implications in autism spectrum disorders. Microb Ecol Health Dis

685

2015; 26: 28177.

686

62)

687

from the gut microbiota regulate host serotonin biosynthesis. Cell 2015; 161: 2764-2769.

688

63)

689

mouse molecular genetics. Nat Rev Neurosci 2003; 4: 1002-1012.

690

64)

691

effects of neurotoxins against monoamines. Exp Brain Res 1996; 110: 183-195.

692

65)

693

of serotonin affects cortical development. Eur. J. Neurosci 2007; 26: 331-344.

694

66)

695

the neocortical projections from the raphe nuclei in perinatal rats: investigation of potential

696

organizational mechanisms. J Comp Neurol 1994; 348: 277−290.

697

67)

698

thalamocortical and serotonergic afferents in the rat barrel field cortex. Cereb. Cortex

699

1991; 1: 380-389.

700

68)

701

corticotropin-releasing factor (CRF) content and release in rat amygdala minces. Brain

702

Res. 1995; 675: 297-302

RI PT

MacFabe DF. Enteric short-chain fatty acids: microbial messengers of metabolism,

SC

Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, et al. Indigenous bacteria

M AN U

Gaspar P, Cases O, Maroteaux L. The developmental role of serotonin: news from

Osterheld-Haas MC, Hornung JP. Laminar development of the mouse barrel cortex:

TE D

Vitalis T, Cases O, Passemard S, Callebert J, Parnavelas JG. Embryonic depletion

EP

Bennett-Clarke CA, Hankin MH, Leslie MJ, Chiaia NL, Rhoades RW. Patterning of

AC C

Blue ME, Erzurumlu RS, Jhaveri S. A comparison of pattern formation by

Cratty MS, Ward HE, Johnson EA, Azzaro AJ, Birkle DL. Prenatal stress increases

29

ACCEPTED MANUSCRIPT 703

69)

Rodiño-Janeiro BK, Alonso-Cotoner C, Pigrau M, Lobo B, Vicario M, Santos J. Role

704

of corticotropin-releasing factor in gastrointestinal permeability. J Neurogastroenterol Motil

705

2015; 21: 33-50.

706

70)

707

cortisol over the course of pregnancy and subsequent child amygdala and hippocampus

708

volumes and affective problems. Proc Natl Acad Sci U S A 2012; 109: E1312-E1319.

709

71)

710

Individual differences in amygdala-medial prefrontal anatomy link negative affect, impaired

711

social functioning, and polygenic depression risk. J Neurosci 2012; 32: 18087-18100.

712

72)

713

microbiota influences blood-brain barrier permeability in mice. Sci Transl Med 2014;

714

6:263ra158.

715

73)

716

cytokines in the pathophysiology of major depression. Biological Psychiatry 2009; 65: 732-

717

741.

718

74)

719

Rev Neurosci 2011; 12: 453-466.

720

75)

721

et al. Regulated extracellular choline acetyltransferase activity - The plausible missing link

722

of the distant action of acetylcholine in the cholinergic anti-inflammatory pathway. PLoS

723

One 2013; 8: e65936.

724

76)

725

psychiatric disorders. Adv Exp Med Biol 2014; 817: 319-356.

726

77)

727

explanations. Am J Phys Anthropol 2015; 157: 1-18.

RI PT

Buss C, Davis EP, Shahbaba B, Pruessner JC, Head K, Sandman CA. Maternal

SC

Holmes AJ, Lee PH, Hollinshead MO, Bakst L, Roffman JL, Smoller JW, et al.

M AN U

Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Tóth M, et al. The gut

TE D

Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of

EP

Mayer EA. Gut feelings: the emerging biology of gut–brain communication. Nature

AC C

Vijayaraghavan S, Karami A, Aeinehband S, Behbahani H, Grandien A, Nilsson B,

Rook GA, Raison CL, Lowry CA. Microbiota, immunoregulatory old friends and

Shattuck EC, Muehlenbein MP. Human sickness behavior: ultimate and proximate

30

ACCEPTED MANUSCRIPT 78)

Mulak A, Bonaz B. Brain-gut-microbiota axis in Parkinson's disease. World J

729

Gastroenterol 2015;21: 10609-10620.

730

79) Shoemark DK, Allen SJ. The microbiome and disease: reviewing the links between the

731

oral microbiome, aging, and Alzheimer's disease. J Alzheimers Dis 2015; 43: 725-738.

732

80) Portune KJ, Benítez-Páez A, Del Pulgar EM, Cerrudo V, Sanz Y. Gut microbiota, diet

733

and obesity-related disorders - the good, the bad and the future challenges. Mol Nutr Food

734

Res 2016; Epub Jun 11.

735

81)

736

stress in human and animal studies: a review of recent evidence. Paediatr Perinat

737

Epidemiol 2008; 22: 438-466.

738

82)

739

science network: fetal and neonatal experience on child and adolescent mental health.

740

Antenatal maternal stress and long-term effects on child neurodevelopment: how and

741

why? J Child Psychol Psychiatry 2007; 48: 245-261.

742

83)

743

during pregnancy is associated with developmental outcome in infancy. J Child Psychol

744

Psychiatry 2003; 44: 810-818.

745

84)

746

stress exerts programming influences on the mother and her fetus. Neuroendocrinology

747

2012; 95: 8-21.

748

85)

749

psychological distress during pregnancy in relation to child development at age two. Child

750

Dev 2006; 77: 573-587.

751

86)

752

dopaminergic function in animal models: possible implications for schizophrenia and other

753

disorders. Neurosci Biobehav Rev 2003; 27: 91-101.

RI PT

728

M AN U

SC

Beydoun H, Saftlas AF. Physical and mental health outcomes of prenatal maternal

TE D

Talge NM, Neal C, Glover V. Early stress, translational research and prevention

EP

Huizink AC, Robles de Medina PG, Mulder EJ, Visser GH, Buitelaar JK. Stress

AC C

Sandman CA, Davis EP, Buss C, Glynn LM. Exposure to prenatal psychobiological

DiPietro JA, Novak MF, Costigan KA, Atella LD, Reusing SP. Maternal

Boksa P, El-Khodor BF. Birth insult interacts with stress at adulthood to alter

31

ACCEPTED MANUSCRIPT 754

87)

Borre YE, Golubeva AV, Crispie F, Scott KA, Hyland NP, Stanton C, et al. Mode of

755

delivery at birth and behavioural outcomes: rewiring of the brain-gut-microbiome axis? In:

756

Society for Neuroscience (SFN) Neuroscience 2014, November 15-19, Washington, USA ,

757

(Washington). Poster Presentation.

758

88)

759

Research review: birth by caesarean section and development of autism spectrum

760

disorder and attention-deficit/hyperactivity disorder: a systematic review and meta-

761

analysis. J Child Psychol Psychiatry 2015; 56: 500-508.

762

89)

763

Association between obstetric mode of delivery and autism spectrum disorder: a

764

population-based sibling design study. JAMA Psychiatry 2015; 72: 935-942.

765

90)

766

mode of delivery and childhood behavior and psychological development in a British

767

cohort. J Autism Dev Disord 2016; 46: 603-614.

768

91)

769

versus vaginally delivered infants. Atten Percept Psychophys 2015; 77: 2529-2539.

770

92)

771

prospective study, sibling pairs analysis, and meta-analysis. BMJ 2006; 333: 945-950.

772

93)

773

associated with improved child cognitive development: a population-based cohort study. J

774

Pediatr 2012; 160: 25-32.

775

94)

776

on early neurocognitive development in Asian children. Am J Clin Nutr 2015; 101: 326-

777

336.

SC

RI PT

Curran EA, O'Neill SM, Cryan JF, Kenny LC, Dinan TG, Khashan AS, et al.

M AN U

Curran EA, Dalman C, Kearney PM, Kenny LC, Cryan JF, Dinan TG, et al.

TE D

Curran EA, Cryan JF, Kenny LC, Dinan TG, Kearney PM, Khashan AS. Obstetrical

Adler SA, Wong-Kee-You AM. Differential attentional responding in caesarean

EP

Der G, Batty GD, Deary IJ. Effect of breast feeding on intelligence in children:

AC C

Quigley MA, Hockley C, Carson C, Kelly Y, Renfrew MJ, Sacker A. Breastfeeding is

Cai S, Pang WW, Low YL, Sim LW, Sam SC, Bruntraeger MB. Infant feeding effects

32

ACCEPTED MANUSCRIPT 778

95)

Krol KM, Rajhans P, Missana M, Grossmann T. Duration of exclusive breastfeeding

779

is associated with differences in infants' brain responses to emotional body expressions.

780

Front Behav Neurosci 2015; 8: 459.

781

96)

782

Pediatr 2002; 14: 583-587.

783

97)

784

Relationship of dietary intake to gastrointestinal symptoms in children with autistic

785

spectrum disorders. Biol Psychiatry 2007; 61: 492-497.

786

98)

787

Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe 2010;

788

16: 444-453.

789

99)

790

microflora studies in late-onset autism. Clin Infect Dis 2002; 35 (Suppl. 1): S6-S16.

791

100) Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, Pekkonen E, et al. Gut

792

microbiota are related to Parkinson's disease and clinical phenotype. Mov Disord 2015; 30:

793

350-358.

794

101) Hu X, Wang T, Jin F. Alzheimer's disease and gut microbiota. Sci China Life Sci

795

2016; Epub Aug 26.

796

102) Critchfield JW, van Hemert S, Ash M, Mulder L, Ashwood P. The potential role of

797

probiotics in the management of childhood autism spectrum disorders. Gastroenterol Res

798

Pract 2011; 2011: 161358.

799

103) D'Mello C, Ronaghan N, Zaheer R, Dicay M, Le T, MacNaughton WK, et al.

800

Probiotics improve inflammation-associated sickness behavior by altering communication

801

between the peripheral immune system and the brain. J Neurosci 2015; 35: 10821-10830.

802

104) Wall R, Marques TM, O'Sullivan O, Ross RP, Shanahan F, Quigley EM. Contrasting

803

effects of Bifidobacterium breve NCIMB 702258 and Bifidobacterium breve DPC 6330 on

RI PT

Horvath K, Perman JA. Autistic disorder and gastrointestinal disease. Curr Opin

SC

Levy SE, Souders MC, Ittenbach RF, Giarelli E, Mulberg AE, Pinto-Martin JA.

M AN U

Finegold SM, Dowd SE, Gontcharova V, Liu C, Henley KE, Wolcott RD, et al.

AC C

EP

TE D

Finegold SM, Molitoris D, Song Y, Liu C, Vaisanen ML, Bolte E. Gastrointestinal

33

ACCEPTED MANUSCRIPT the composition of murine brain fatty acids and gut microbiota. Am J Clin Nutr 2012; 95:

805

1278-1287.

806

105) Desbonnet L, Garrett L, Clarke G, Kiely B, Cryan JF, Dinan TG. Effects of the

807

probiotic Bifidobacterium infantis in the maternal separation model of depression.

808

Neuroscience 2010; 170: 1179-1188.

809

106) Bercik P, Park AJ, Sinclair D, Khoshdel A, Lu J, Huang X, Deng Y, et al. The

810

anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain

811

communication. Neurogastroenterol Motil 2011; 23: 1132-1139.

812

107) Bercik P, Park AJ, Sinclair D, Khoshdel A, Lu J, Huang X, et al. The anxiolytic effect

813

of

814

communication. Neurogastroenterol Motil 2011; 23: 1132-1139.

815

108) Zareie M, Johnson-Henry K, Jury J, Yang PC, Ngan BY, McKay DM, et al.

816

Probiotics prevent bacterial translocation and improve intestinal barrier function in rats

817

following chronic psychological stress. Gut 2006; 55: 1553-1560.

818

109) Ait-Belgnaoui A, Durand H, Cartier C, Chaumaz G, Eutamene H, Ferrier L, et al.

819

Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to

820

an acute psychological stress in rats. Psychoneuroendocrinology 2012; 37: 1885-1895.

821

110) Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T, et al. Microbiota

822

modulate behavioral and physiological abnormalities associated with neurodevelopmental

823

disorders. Cell 2013; 155: 1451-1463.

824

111) Persico AM, Napolioni V. Urinary p-cresol in autism spectrum disorder. Neurotoxicol

825

Teratol 2013; 36: 82-90.

826

112) Tillisch K, Labus J, Kilpatrick L, Jiang Z, Stains J, Ebrat B, et al. Consumption of

827

fermented milk product with probiotic modulates brain activity.Gastroenterology. 2013;

828

144: 1394-1401.

involves

SC

NCC3001

vagal

M AN U

longum

pathways

for

gut-brain

AC C

EP

TE D

Bifidobacterium

RI PT

804

34

ACCEPTED MANUSCRIPT 113) Pärtty A, Kalliomäki M, Wacklin P, Salminen S, Isolauri E. A possible link between

830

early probiotic intervention and the risk of neuropsychiatric disorders later in childhood: a

831

randomized trial. Pediatr Res 2015; 77: 823-828.

832

114) Romeo MG, Romeo DM, Trovato L, Oliveri S, Palermo F, Cota F, et al. Role of

833

probiotics in the prevention of the enteric colonization by Candida in preterm newborns:

834

incidence of late-onset sepsis and neurological outcome. J Perinatol 2011; 31: 63-69.

835

115) Chou IC, Kuo HT, Chang JS, Wu SF, Chiu HY, Su BH, et al. Lack of effects of oral

836

probiotics on growth and neurodevelopmental outcomes in preterm very low birth weight

837

infants. J Pediatr 2010; 156: 393-396.

838

116) Parracho HMRT, Gibson GR, Knott F, Bosscher D, Kleerebezem M, McCartney AL.

839

A double blind, placebo-controlled, crossover-designed probiotic feeding study in children

840

diagnosed with autistic spectrum disorders. Intern J Prob Preb 2010; 5: 69-74.

841

117) Kałużna-Czaplińska J, Błaszczyk S. The level of arabinitol in autistic children after

842

probiotic therapy. Nutrition 2012; 28: 124-126.

843

118) Santocchi E, Guiducci L, Fulceri F, Billeci L, Buzzigoli E, Apicella F, et al. Gut to

844

brain interaction in Autism Spectrum Disorders: a randomized controlled trial on the role of

845

probiotics on clinical, biochemical and neurophysiological parameters. BMC Psychiatry

846

2016; 16: 183.

847

119) Reid G. Probiotics: definition, scope and mechanisms of action. Best Pract Res

848

Clin Gastroenterol 2016; 30: 17-25.

849

120) Akbari P, Fink-Gremmels J, Willems RH, Difilippo E, Schols HA, Schoterman MH, et

850

al. Characterizing microbiota-independent effects of oligosaccharides on intestinal

851

epithelial cells: insight into the role of structure and size: structure-activity relationships of

852

non-digestible oligosaccharides. Eur J Nutr 2016; Epub Jun 13.

AC C

EP

TE D

M AN U

SC

RI PT

829

35

ACCEPTED MANUSCRIPT 121) Savignac HM, Corona G, Mills H, Chen L, Spencer JP, Tzortzis G, Burnet PW.

854

Prebiotic feeding elevates central brain derived neurotrophic factor, N-methyl-D-aspartate

855

receptor subunits and D-serine. Neurochem Int 2013; 63: 756-764.

856

122) Savignac HM, Couch Y, Stratford M, Bannerman DM, Tzortzis G, Anthony DC, et al.

857

Prebiotic administration normalizes lipopolysaccharide (LPS)-induced anxiety and cortical

858

5-HT2A receptor and IL1-β levels in male mice. Brain Behav Immun 2016; 52: 120-131.

859

123) Williams S, Chen L, Savignac HM, Tzortzis G, Anthony DC, Burnet PW. Neonatal

860

prebiotic (BGOS) supplementation increases the levels of synaptophysin, GluN2A-

861

subunits and BDNF proteins in the adult rat hippocampus. Synapse 2016; 70: 121-124.

862

124) Schmidt K, Cowen PJ, Harmer CJ, Tzortzis G, Errington S, Burnet PW. Prebiotic

863

intake reduces the waking cortisol response and alters emotional bias in healthy

864

volunteers. Psychopharmacology (Berl) 2015; 232: 1793-1801.

865

125) Li W, Dowd SE, Scurlock B, Acosta-Martinez V, Lyte M. Memory and learning

866

behavior in mice is temporally associated with diet-induced alterations in gut bacteria.

867

Physiol Behav 2009; 96: 557-567.

SC

M AN U

TE D

EP AC C

868

RI PT

853

36

ACCEPTED MANUSCRIPT 869 870 871 872

Table 1. Factors influencing gut microbiota during development. Factor Antibiotic therapy, malnutrition or overnutrition, obesity, diabetes, eczema, stress during pregnancy Duration of gestation, mode of delivery, use of antibiotics, type of feeding

RI PT

Development phase Fetal life

Postnatal period 873

SC

874 875

M AN U

876 877 878 879

883 884 885 886 887 888

EP

882

AC C

881

TE D

880

889 890 891 892 37

ACCEPTED MANUSCRIPT 893 894

Table 2. Mechanisms of interaction between gut microbiota and central nervous system. Mechanism Passage of lipopolysaccharides in the cell wall of some Gram-negative bacteria from the intestinal mucosa to the systemic circulation Short chain fatty acid production by bacteria fermentation 5-hydroxytryptamine biosynthesis from gut bacteria Stress-related increase in the cortisol level

Result Production cytokines

of

pro-inflammatory

RI PT

895 896 897

Alteration of mitochondrial function

M AN U

SC

Increase in neuronal cell division and differentiation Increased gut permeability and increased volume of the amygdala Modification of the blood-brain barrier Reduced expression of the tight junction proteins occludin and claudin5 Production of peptides that activate afferent Influence on behavior and neural endings of the vagus nerve activity 898

AC C

EP

TE D

899

38

ACCEPTED MANUSCRIPT HIGHLIGHTS •

Gut dysbiosis has been associated with alterations of central nervous system (CNS).



The effects of dysbiosis on the CNS are significantly more important during the

RI PT

developmental period. •

Both probiotics and prebiotics can have different impacts on CNS.



In humans, several factors may be important in conditioning gut microbiota

EP

TE D

microbiota at the CNS level.

M AN U

Efforts are required to clarify the mechanisms that are influenced by the gut

AC C



SC

modifications.