Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Gut Microbes ; 6(3): 207-13, 2015.
Article in English | MEDLINE | ID: mdl-25835343

ABSTRACT

Through extensive microbial-mammalian co-metabolism, the intestinal microbiota have evolved to exert a marked influence on health and disease via gut-brain-microbiota interactions. In this addendum, we summarize the findings of our recent study on the fecal microbiota and metabolomes of children with pervasive developmental disorder-not otherwise specified (PDD-NOS) or autism (AD) compared with healthy children (HC). Children with PDD-NOS or AD have altered fecal microbiota and metabolomes (including neurotransmitter molecules). We hypothesize that the degree of microbial alteration correlates with the severity of the disease since fecal microbiota and metabolomes alterations were higher in children with PDD-NOS and, especially, AD compared to HC. Our study indicates that the levels of free amino acids (FAA) and volatile organic compounds (VOC) differ in AD subjects compared to children with PDD-NOS, who are more similar to HC. Finally, we propose a new perspective on the implications for the interaction between intestinal microbiota and AD.


Subject(s)
Child Development Disorders, Pervasive/epidemiology , Child Development Disorders, Pervasive/microbiology , Dysbiosis/complications , Gastrointestinal Microbiome , Microbiota , Amino Acids/analysis , Feces/chemistry , Humans , Volatile Organic Compounds/analysis
2.
Brain Behav Immun ; 44: 100-5, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25218900

ABSTRACT

Animal models indicate that maternal infection during pregnancy can result in behavioral abnormalities and neuropathologies in offspring. We examined the association between maternal inpatient diagnosis with infection during pregnancy and risk of ASD in a Swedish nationwide register-based birth cohort born 1984-2007 with follow-up through 2011. In total, the sample consisted of 2,371,403 persons with 24,414 ASD cases. Infection during pregnancy was defined from ICD codes. In the sample, 903 mothers of ASD cases (3.7%) had an inpatient diagnosis of infection during pregnancy. Logistic regression models adjusted for a number of covariates yielded odds ratios indicating approximately a 30% increase in ASD risk associated with any inpatient diagnosis of infection. Timing of infection did not appear to influence risk in the total Swedish population, since elevated risk of ASD was associated with infection in all trimesters. In a subsample analysis, infections were associated with greater risk of ASD with intellectual disability than for ASD without intellectual disability. The present study adds to the growing body of evidence, encompassing both animal and human studies, that supports possible immune-mediated mechanisms underlying the etiology of ASD.


Subject(s)
Child Development Disorders, Pervasive/epidemiology , Child Development Disorders, Pervasive/etiology , Pregnancy Complications, Infectious/epidemiology , Adult , Child Development Disorders, Pervasive/immunology , Child Development Disorders, Pervasive/microbiology , Cohort Studies , Female , Hospitalization , Humans , Male , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/microbiology , Risk Factors , Sweden/epidemiology
3.
PLoS One ; 9(8): e103740, 2014.
Article in English | MEDLINE | ID: mdl-25170769

ABSTRACT

Alterations in gut microbiome composition have an emerging role in health and disease including brain function and behavior. Short chain fatty acids (SCFA) like propionic (PPA), and butyric acid (BA), which are present in diet and are fermentation products of many gastrointestinal bacteria, are showing increasing importance in host health, but also may be environmental contributors in neurodevelopmental disorders including autism spectrum disorders (ASD). Further to this we have shown SCFA administration to rodents over a variety of routes (intracerebroventricular, subcutaneous, intraperitoneal) or developmental time periods can elicit behavioral, electrophysiological, neuropathological and biochemical effects consistent with findings in ASD patients. SCFA are capable of altering host gene expression, partly due to their histone deacetylase inhibitor activity. We have previously shown BA can regulate tyrosine hydroxylase (TH) mRNA levels in a PC12 cell model. Since monoamine concentration is known to be elevated in the brain and blood of ASD patients and in many ASD animal models, we hypothesized that SCFA may directly influence brain monoaminergic pathways. When PC12 cells were transiently transfected with plasmids having a luciferase reporter gene under the control of the TH promoter, PPA was found to induce reporter gene activity over a wide concentration range. CREB transcription factor(s) was necessary for the transcriptional activation of TH gene by PPA. At lower concentrations PPA also caused accumulation of TH mRNA and protein, indicative of increased cell capacity to produce catecholamines. PPA and BA induced broad alterations in gene expression including neurotransmitter systems, neuronal cell adhesion molecules, inflammation, oxidative stress, lipid metabolism and mitochondrial function, all of which have been implicated in ASD. In conclusion, our data are consistent with a molecular mechanism through which gut related environmental signals such as increased levels of SCFA's can epigenetically modulate cell function further supporting their role as environmental contributors to ASD.


Subject(s)
Butyric Acid/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Enterobacteriaceae/physiology , Host-Pathogen Interactions , PC12 Cells/microbiology , Propionates/metabolism , Animals , Child Development Disorders, Pervasive/microbiology , Gene Expression Regulation , Gene Regulatory Networks , PC12 Cells/metabolism , Rats , Synaptic Transmission , Transcriptional Activation , Tyrosine 3-Monooxygenase/genetics
9.
Brain Behav Immun ; 37: 197-206, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24333160

ABSTRACT

Autism spectrum disorder (ASD) is a heterogeneous group of complex neurodevelopmental disorders with evidence of genetic predisposition. Intestinal disturbances are reported in ASD patients and compositional changes in gut microbiota are described. However, the role of microbiota in brain disorders is poorly documented. Here, we used a murine model of ASD to investigate the relation between gut microbiota and autism-like behaviour. Using next generation sequencing technology, microbiota composition was investigated in mice in utero exposed to valproic acid (VPA). Moreover, levels of short chain fatty acids (SCFA) and lactic acid in caecal content were determined. Our data demonstrate a transgenerational impact of in utero VPA exposure on gut microbiota in the offspring. Prenatal VPA exposure affected operational taxonomic units (OTUs) assigned to genera within the main phyla of Bacteroidetes and Firmicutes and the order of Desulfovibrionales, corroborating human ASD studies. In addition, OTUs assigned to genera of Alistipes, Enterorhabdus, Mollicutes and Erysipelotrichalis were especially associated with male VPA-exposed offspring. The microbial differences of VPA in utero-exposed males deviated from those observed in females and was (i) positively associated with increased levels of caecal butyrate as well as ileal neutrophil infiltration and (ii) inversely associated with intestinal levels of serotonin and social behaviour scores. These findings show that autism-like behaviour and its intestinal phenotype is associated with altered microbial colonization and activity in a murine model for ASD, with preponderance in male offspring. These results open new avenues in the scientific trajectory of managing neurodevelopmental disorders by gut microbiome modulation.


Subject(s)
Child Development Disorders, Pervasive/microbiology , Intestine, Large/microbiology , Microbiota/physiology , Acetic Acid/analysis , Animals , Butyric Acid/analysis , Child Development Disorders, Pervasive/metabolism , Disease Models, Animal , Female , Intestine, Large/metabolism , Lactic Acid/analysis , Male , Mice , Mice, Inbred BALB C , Microbiota/drug effects , Sex Factors , Valproic Acid/toxicity
10.
Cell ; 155(7): 1446-8, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24360269

ABSTRACT

Hsaio and colleagues link gut microbes to autism spectrum disorders (ASD) in a mouse model. They show that ASD symptoms are triggered by compositional and structural shifts of microbes and associated metabolites, but symptoms are relieved by a Bacteroides fragilis probiotic. Thus probiotics may provide therapeutic strategies for neurodevelopmental disorders.


Subject(s)
Child Development Disorders, Pervasive/microbiology , Gastrointestinal Tract/microbiology , Animals , Female , Humans
11.
Cell ; 155(7): 1451-63, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24315484

ABSTRACT

Neurodevelopmental disorders, including autism spectrum disorder (ASD), are defined by core behavioral impairments; however, subsets of individuals display a spectrum of gastrointestinal (GI) abnormalities. We demonstrate GI barrier defects and microbiota alterations in the maternal immune activation (MIA) mouse model that is known to display features of ASD. Oral treatment of MIA offspring with the human commensal Bacteroides fragilis corrects gut permeability, alters microbial composition, and ameliorates defects in communicative, stereotypic, anxiety-like and sensorimotor behaviors. MIA offspring display an altered serum metabolomic profile, and B. fragilis modulates levels of several metabolites. Treating naive mice with a metabolite that is increased by MIA and restored by B. fragilis causes certain behavioral abnormalities, suggesting that gut bacterial effects on the host metabolome impact behavior. Taken together, these findings support a gut-microbiome-brain connection in a mouse model of ASD and identify a potential probiotic therapy for GI and particular behavioral symptoms in human neurodevelopmental disorders.


Subject(s)
Child Development Disorders, Pervasive/microbiology , Gastrointestinal Tract/microbiology , Animals , Anxiety/metabolism , Anxiety/microbiology , Bacteroides fragilis , Behavior, Animal , Brain/physiology , Child , Child Development Disorders, Pervasive/metabolism , Disease Models, Animal , Female , Gastrointestinal Tract/metabolism , Humans , Mice , Mice, Inbred C57BL , Microbiota , Probiotics/administration & dosage
12.
PLoS One ; 8(10): e76993, 2013.
Article in English | MEDLINE | ID: mdl-24130822

ABSTRACT

This study aimed at investigating the fecal microbiota and metabolome of children with Pervasive Developmental Disorder Not Otherwise Specified (PDD-NOS) and autism (AD) in comparison to healthy children (HC). Bacterial tag-encoded FLX-titanium amplicon pyrosequencing (bTEFAP) of the 16S rDNA and 16S rRNA analyses were carried out to determine total bacteria (16S rDNA) and metabolically active bacteria (16S rRNA), respectively. The main bacterial phyla (Firmicutes, Bacteroidetes, Fusobacteria and Verrucomicrobia) significantly (P<0.05) changed among the three groups of children. As estimated by rarefaction, Chao and Shannon diversity index, the highest microbial diversity was found in AD children. Based on 16S-rRNA and culture-dependent data, Faecalibacterium and Ruminococcus were present at the highest level in fecal samples of PDD-NOS and HC children. Caloramator, Sarcina and Clostridium genera were the highest in AD children. Compared to HC, the composition of Lachnospiraceae family also differed in PDD-NOS and, especially, AD children. Except for Eubacterium siraeum, the lowest level of Eubacteriaceae was found on fecal samples of AD children. The level of Bacteroidetes genera and some Alistipes and Akkermansia species were almost the highest in PDD-NOS or AD children as well as almost all the identified Sutterellaceae and Enterobacteriaceae were the highest in AD. Compared to HC children, Bifidobacterium species decreased in AD. As shown by Canonical Discriminant Analysis of Principal Coordinates, the levels of free amino acids and volatile organic compounds of fecal samples were markedly affected in PDD-NOS and, especially, AD children. If the gut microbiota differences among AD and PDD-NOS and HC children are one of the concomitant causes or the consequence of autism, they may have implications regarding specific diagnostic test, and/or for treatment and prevention.


Subject(s)
Autistic Disorder/microbiology , Child Development Disorders, Pervasive/microbiology , Feces/microbiology , Metabolome , Microbiota/genetics , Amino Acids/metabolism , Bacteria/genetics , Bacteria/metabolism , Child , Child, Preschool , Female , Humans , Male , Sequence Analysis , Volatile Organic Compounds/metabolism
13.
JAMA Pediatr ; 167(4): 374-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23400224

ABSTRACT

The mammalian gastrointestinal tract harbors a highly diverse microbial population that plays a major role in nutrition, metabolism, protection against pathogens, and development of the immune system. It is estimated that at least 1000 different bacterial species cohabit the human intestinal tract. Most recently, the Human Microbiome Project, using new genomic technologies, has started a catalog of specific microbiome composition and its correlation with health and specific diseases. Herein we provide a brief review of the intestinal microbiome, with a focus on new studies showing that there is an important link between the microbes that inhabit the intestinal tract and the developing brain. With future research, an understanding of this link may help us to treat various neurobehavioral problems such as autism, schizophrenia, and anxiety.


Subject(s)
Brain/physiology , Intestines/microbiology , Intestines/physiology , Metagenome/physiology , Animals , Brain/growth & development , Child , Child Development Disorders, Pervasive/epidemiology , Child Development Disorders, Pervasive/microbiology , Comorbidity , Enteric Nervous System/physiology , Gastrointestinal Diseases/epidemiology , Humans , Infant, Newborn , Infant, Premature/physiology , Intestines/immunology , Metagenome/immunology , Toll-Like Receptors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...