Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Cell Rep ; 38(5): 110311, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108531

ABSTRACT

Gut microbial products direct growth, differentiation, and development in animal hosts. However, we lack system-wide understanding of cell-specific responses to the microbiome. We profiled cell transcriptomes from the intestine, and associated tissue, of zebrafish larvae raised in the presence or absence of a microbiome. We uncovered extensive cellular heterogeneity in the conventional zebrafish intestinal epithelium, including previously undescribed cell types with known mammalian homologs. By comparing conventional to germ-free profiles, we mapped microbial impacts on transcriptional activity in each cell population. We revealed intricate degrees of cellular specificity in host responses to the microbiome that included regulatory effects on patterning and on metabolic and immune activity. For example, we showed that the absence of microbes hindered pro-angiogenic signals in the developing vasculature, causing impaired intestinal vascularization. Our work provides a high-resolution atlas of intestinal cellular composition in the developing fish gut and details the effects of the microbiome on each cell type.


Subject(s)
Gastrointestinal Microbiome/physiology , Host Microbial Interactions/physiology , Intestines/blood supply , Microbiota/physiology , Animals , Germ-Free Life/physiology , RNA, Ribosomal, 16S/metabolism , Zebrafish
2.
Nat Commun ; 12(1): 3105, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050144

ABSTRACT

Environmental factors, mucosal permeability and defective immunoregulation drive overactive immunity to a subset of resident intestinal bacteria that mediate multiple inflammatory conditions. GUT-103 and GUT-108, live biotherapeutic products rationally designed to complement missing or underrepresented functions in the dysbiotic microbiome of IBD patients, address upstream targets, rather than targeting a single cytokine to block downstream inflammation responses. GUT-103, composed of 17 strains that synergistically provide protective and sustained engraftment in the IBD inflammatory environment, prevented and treated chronic immune-mediated colitis. Therapeutic application of GUT-108 reversed established colitis in a humanized chronic T cell-mediated mouse model. It decreased pathobionts while expanding resident protective bacteria; produced metabolites promoting mucosal healing and immunoregulatory responses; decreased inflammatory cytokines and Th-1 and Th-17 cells; and induced interleukin-10-producing colonic regulatory cells, and IL-10-independent homeostatic pathways. We propose GUT-108 for treating and preventing relapse for IBD and other inflammatory conditions characterized by unbalanced microbiota and mucosal permeability.


Subject(s)
Bacteria/metabolism , Colitis/microbiology , Colitis/therapy , Cytokines/metabolism , Dysbiosis/microbiology , Gastrointestinal Microbiome , Germ-Free Life , Animals , Bacteria/genetics , Bile Acids and Salts/metabolism , Colitis/immunology , Disease Models, Animal , Dysbiosis/therapy , Feces/microbiology , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Germ-Free Life/immunology , Germ-Free Life/physiology , Homeostasis , Humans , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Gut Microbes ; 13(1): 1866974, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33459114

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized primarily by motor and non-motor gastrointestinal (GI) deficits. GI symptoms' including compromised intestinal barrier function often accompanies altered gut microbiota composition and motor deficits in PD. Therefore, in this study, we set to investigate the role of gut microbiota and epithelial barrier dysfunction on motor symptom generation using a rotenone-induced mouse model of PD. We found that while six weeks of 10 mg/kg of chronic rotenone administration by oral gavage resulted in loss of tyrosine hydroxylase (TH) neurons in both germ-free (GF) and conventionally raised (CR) mice, the decrease in motor strength and coordination was observed only in CR mice. Chronic rotenone treatment did not disrupt intestinal permeability in GF mice but resulted in a significant change in gut microbiota composition and an increase in intestinal permeability in CR mice. These results highlight the potential role of gut microbiota in regulating barrier dysfunction and motor deficits in PD.


Subject(s)
Gastrointestinal Diseases/pathology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Parkinson Disease/pathology , Rotenone/toxicity , Tight Junctions/pathology , Animals , Brain-Gut Axis , Disease Models, Animal , Dysbiosis/microbiology , Dystonic Disorders/congenital , Dystonic Disorders/pathology , Female , Germ-Free Life/physiology , Male , Mice , Tight Junctions/drug effects , Tyrosine 3-Monooxygenase/metabolism
4.
STAR Protoc ; 1(3): 100226, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33377117

ABSTRACT

We present a gnotobiotic system for microbiota reconstitution on Arabidopsis thaliana under contrasting iron availability. This system induces iron starvation in plants by providing an unavailable form, mimicking conditions in alkaline soils. Inoculation of taxonomically diverse bacteria reconstitutes plants with a synthetic microbiota, allowing observation of nutrient-dependent interactions with commensals. Experimental optimization, including media composition and preparation of seedlings and bacteria, is discussed. This system provides a framework that can be adapted to study plant-microbiota interactions in further nutritional contexts. For complete details on the use and execution of this protocol, please refer to Harbort et al. (2020).


Subject(s)
Cell Culture Techniques/methods , Germ-Free Life/physiology , Plant Roots/growth & development , Arabidopsis/growth & development , Arabidopsis/microbiology , Bacteria , Microbiota , Plant Roots/microbiology , Soil , Soil Microbiology , Symbiosis
5.
Eur Neuropsychopharmacol ; 41: 152-159, 2020 12.
Article in English | MEDLINE | ID: mdl-33191074

ABSTRACT

The gut microbiota modulates brain physiology, development, and behavior and has been implicated as a key regulator in several central nervous system disorders. Its effect on the metabolic coupling between neurons and astrocytes has not been studied to date, even though this is an important component of brain energy metabolism and physiology and it is perturbed in neurodegenerative and cognitive disorders. In this study, we have investigated the mRNA expression of 6 genes encoding proteins implicated in the astrocyte-neuron lactate shuttle (Atp1a2, Ldha, Ldhb, Mct1, Gys1, Pfkfb3), in relation to different gut microbiota manipulations, in the mouse brain hippocampus, a region with critical functions in cognition and behavior. We have discovered that Atp1a2 and Pfkfb3, encoding the ATPase, Na+/K+ transporting, alpha 2 sub-unit, respectively and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3, two genes predominantly expressed in astrocytes, were upregulated in the hippocampus after microbial colonization of germ-free mice for 24 h, compared with conventionally raised mice. Pfkfb3 was also upregulated in germ-free mice compared with conventionally raised mice, while an increase in Atp1a2 expression in germ-free mice was confirmed only at the protein level by Western blot. In a separate cohort of mice, Atp1a2 and Pfkfb3 mRNA expression was upregulated in the hippocampus following 6-week dietary supplementation with prebiotics (fructo- and galacto-oligosaccharides) in an animal model of chronic psychosocial stress. To our knowledge, these findings are the first to report an influence of the gut microbiota and prebiotics on mRNA expression of genes implicated in the metabolic coupling between neurons and astrocytes.


Subject(s)
Astrocytes/metabolism , Gastrointestinal Microbiome/physiology , Germ-Free Life/physiology , Hippocampus/metabolism , Lactic Acid/metabolism , Neurons/metabolism , Animals , Energy Metabolism/physiology , Gene Expression , Male , Mice , Mice, Inbred C57BL , Prebiotics/administration & dosage
6.
Hypertension ; 76(6): 1847-1855, 2020 12.
Article in English | MEDLINE | ID: mdl-33070663

ABSTRACT

Commensal gut microbiota are strongly correlated with host hemodynamic homeostasis but only broadly associated with cardiovascular health. This includes a general correspondence of quantitative and qualitative shifts in intestinal microbial communities found in hypertensive rat models and human patients. However, the mechanisms by which gut microbes contribute to the function of organs important for blood pressure (BP) control remain unanswered. To examine the direct effects of microbiota on BP, we conventionalized germ-free (GF) rats with specific pathogen-free rats for a short-term period of 10 days, which served as a model system to observe the dynamic responses when reconstituting the holobiome. The absence of microbiota in GF rats resulted with relative hypotension compared with their conventionalized counterparts, suggesting an obligatory role of microbiota in BP homeostasis. Hypotension observed in GF rats was accompanied by a marked reduction in vascular contractility. Both BP and vascular contractility were restored by the introduction of microbiota to GF rats, indicating that microbiota could impact BP through a vascular-dependent mechanism. This is further supported by the decrease in actin polymerization in arteries from GF rats. Improved vascular contractility in conventionalized GF rats, as indicated through stabilized actin filaments, was associated with an increase in cofilin phosphorylation. These data indicate that the vascular system senses the presence (or lack of) microbiota to maintain vascular tone via actin polymerization. Overall, these results constitute a fundamental discovery of the essential nature of microbiota in BP regulation.


Subject(s)
Blood Pressure/physiology , Gastrointestinal Microbiome/physiology , Germ-Free Life/physiology , Mesenteric Arteries/physiology , Actin Cytoskeleton/metabolism , Animals , Cell Movement/physiology , Cell Proliferation/physiology , Hypotension/physiopathology , Male , Mesenteric Arteries/cytology , Microbiota/physiology , Polymerization , Rats, Sprague-Dawley , Specific Pathogen-Free Organisms/physiology
7.
Sci Rep ; 10(1): 18168, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097776

ABSTRACT

Drosophila melanogaster females undergo a variety of post-mating changes that influence their activity, feeding behavior, metabolism, egg production and gene expression. These changes are induced either by mating itself or by sperm or seminal fluid proteins. In addition, studies have shown that axenic females-those lacking a microbiome-have altered fecundity compared to females with a microbiome, and that the microbiome of the female's mate can influence reproductive success. However, the extent to which post-mating changes in transcript abundance are affected by microbiome state is not well-characterized. Here we investigated fecundity and the post-mating transcript abundance profile of axenic or control females after mating with either axenic or control males. We observed interactions between the female's microbiome and her mating status: transcripts of genes involved in reproduction and genes with neuronal functions were differentially abundant depending on the females' microbiome status, but only in mated females. In addition, immunity genes showed varied responses to either the microbiome, mating, or a combination of those two factors. We further observed that the male's microbiome status influences the fecundity of both control and axenic females, while only influencing the transcriptional profile of axenic females. Our results indicate that the microbiome plays a vital role in the post-mating switch of the female's transcriptome.


Subject(s)
Drosophila melanogaster/physiology , Germ-Free Life/physiology , Microbiota/physiology , Sexual Behavior, Animal/physiology , Transcriptome/immunology , Animals , Axenic Culture , Drosophila melanogaster/microbiology , Female , Fertility/physiology , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Immunity, Innate/genetics , Male , RNA-Seq , Reproduction/physiology
8.
Toxins (Basel) ; 12(9)2020 08 23.
Article in English | MEDLINE | ID: mdl-32842482

ABSTRACT

Salmonella Typhimurium is a Gram-negative bacterium that causes enterocolitis in humans and pigs. Lipopolysaccharide (LPS) is a component of the outer leaflet of Gram-negative bacteria that provokes endotoxin shock. LPS can be synthesized completely or incompletely and creates S (smooth) or R (rough) chemotypes. Toll-like receptors (TLR) 2, 4, and 9 initiate an inflammatory reaction to combat bacterial infections. We associated/challenged one-week-old gnotobiotic piglets with wild-type S. Typhimurium with S chemotype or its isogenic ∆rfa mutants with R chemotype LPS. The wild-type S. Typhimurium induced TLR2 and TLR4 mRNA expression but not TLR9 mRNA expression in the ileum and colon of one-week-old gnotobiotic piglets 24 h after challenge. The TLR2 and TLR4 stimulatory effects of the S. Typhimurium ∆rfa mutants were related to the completeness of their LPS chain. The transcription of IL-12/23 p40, IFN-γ, and IL-6 in the intestine and the intestinal and plasmatic levels of IL-12/23 p40 and IL-6 but not IFN-γ were related to the activation of TLR2 and TLR4 signaling pathways. The avirulent S. Typhimurium ∆rfa mutants are potentially useful for modulation of the TLR2 and TLR4 signaling pathways to protect the immunocompromised gnotobiotic piglets against subsequent infection with the virulent S. Typhimurium.


Subject(s)
Colon/metabolism , Germ-Free Life/physiology , Ileum/metabolism , Salmonella Infections/metabolism , Salmonella typhimurium/isolation & purification , Toll-Like Receptor 4/metabolism , Animals , Colon/microbiology , Ileum/microbiology , Mutation/physiology , Salmonella Infections/genetics , Salmonella Infections/pathology , Salmonella typhimurium/genetics , Swine , Swine, Miniature
9.
Toxins (Basel) ; 12(9)2020 08 26.
Article in English | MEDLINE | ID: mdl-32859011

ABSTRACT

Alterations in microbiota are known to affect kidney disease conditions. We have previously shown that germ-free conditions exacerbated adenine-induced kidney damage in mice; however, the mechanism by which this occurs has not been elucidated. To explore this mechanism, we examined the influence of germ-free conditions on purine metabolism and renal immune responses involved in the kidney damage. Germ-free mice showed higher expression levels of purine-metabolizing enzymes such as xanthine dehydrogenase, which converts adenine to a nephrotoxic byproduct 2,8-dihydroxyadenine (2,8-DHA). The germ-free mice also showed increased urinary excretion of allantoin, indicating enhanced purine metabolism. Metabolome analysis demonstrated marked differences in the purine metabolite levels in the feces of germ-free mice and mice with microbiota. Furthermore, unlike the germ-free condition, antibiotic treatment did not increase the expression of purine-metabolizing enzymes or exacerbate adenine-induced kidney damage. Considering renal immune responses, the germ-free mice displayed an absence of renal IL-17A expression. However, the adenine-induced kidney damage in wild-type mice was comparable to that in IL-17A-deficient mice, suggesting that IL-17A does not play a major role in the disease condition. Our results suggest that the enhanced host purine metabolism in the germ-free mice potentially promotes the conversion of the administered adenine into 2,8-DHA, resulting in exacerbated kidney damage. This further suggests a role of the microbiota in regulating host purine metabolism.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Adenine/toxicity , Germ-Free Life/physiology , Purines/metabolism , Acute Kidney Injury/pathology , Animals , Interleukin-17/deficiency , Interleukin-17/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
10.
Microbiome ; 7(1): 158, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31831058

ABSTRACT

Given the increasing use of gnotobiotic mouse models for deciphering the effects of human microbial communities on host biology, there is a need to develop new methods for characterizing these animals while maintaining their isolation from environmental microbes. We describe a method for performing open-circuit indirect calorimetry on gnotobiotic mice colonized with gut microbial consortia obtained from different human donors. In this illustrative case, cultured collections of gut bacterial strains were obtained from obese and lean co-twins. The approach allows microbial contributions to host energy homeostasis to be characterized.


Subject(s)
Calorimetry, Indirect/methods , Energy Metabolism/physiology , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Obesity/microbiology , Animals , Feces/microbiology , Germ-Free Life/physiology , Metagenome , Mice
11.
Cell ; 179(1): 59-73.e13, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31539500

ABSTRACT

Development of microbiota-directed foods (MDFs) that selectively increase the abundance of beneficial human gut microbes, and their expressed functions, requires knowledge of both the bioactive components of MDFs and the mechanisms underlying microbe-microbe interactions. Here, gnotobiotic mice were colonized with a defined consortium of human-gut-derived bacterial strains and fed different combinations of 34 food-grade fibers added to a representative low-fiber diet consumed in the United States. Bioactive carbohydrates in fiber preparations targeting particular Bacteroides species were identified using community-wide quantitative proteomic analyses of bacterial gene expression coupled with forward genetic screens. Deliberate manipulation of community membership combined with administration of retrievable artificial food particles, consisting of paramagnetic microscopic beads coated with dietary polysaccharides, disclosed the contributions of targeted species to fiber degradation. Our approach, including the use of bead-based biosensors, defines nutrient-harvesting strategies that underlie, as well as alleviate, competition between Bacteroides and control the selectivity of MDF components.


Subject(s)
Bacteroides/genetics , Dietary Fiber/pharmacology , Gastrointestinal Microbiome/drug effects , Germ-Free Life/physiology , Microbial Interactions/drug effects , Polysaccharides/pharmacology , Proteomics/methods , Animals , Diet/methods , Dietary Fiber/metabolism , Feces/microbiology , Gastrointestinal Microbiome/physiology , Gene Expression Regulation, Bacterial/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Polysaccharides/metabolism
12.
ACS Chem Neurosci ; 10(9): 3953-3960, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31415146

ABSTRACT

Microbial colonization of the gastrointestinal tract plays a crucial role in the development of enteric and central nervous system functionality. The serotonergic system has been heavily implicated in microbiota-gut-brain axis signaling, particularly in proof-of-principle studies in germ-free (GF) animals. One aspect of the serotonergic system that has been left unexplored in relation to the microbiota is the unique ability of the serotonin receptor 2C (5-HT2C) to undergo post-transcriptional editing, which has been implicated in decreased receptor functionality. We investigated whether GF mice, with absent microbiota from birth, have altered 5-HT2C receptor expression and editing in the brain, and if colonization of the microbiota is able to restore editing patterns. Next, we investigated whether microbiota depletion later in life using a chronic antibiotic treatment could affect 5-HT2C receptor editing patterns in rats. We found that GF mice have an increased prevalence of the edited 5-HT2C receptor isoforms in the amygdala, hypothalamus, prefrontal cortex, and striatum, which was partially normalized upon colonization post-weaning. However, no alterations were observed in the hypothalamus after microbiota depletion using an antibiotic treatment in adult rats. This suggests that alterations in the microbiome during development, but not later in life, could influence 5-HT2C receptor editing patterns. Overall, these results demonstrate that the microbiota affects 5-HT2C receptor editing in the brain and may inform novel therapeutic strategies in conditions in which 5-HT2C receptor editing is altered, such as depression.


Subject(s)
Brain/metabolism , Gastrointestinal Microbiome/physiology , Gene Editing/methods , Receptor, Serotonin, 5-HT2C/genetics , Receptor, Serotonin, 5-HT2C/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Brain/drug effects , Gastrointestinal Microbiome/drug effects , Germ-Free Life/drug effects , Germ-Free Life/physiology , Male , Mice , Rats , Rats, Sprague-Dawley
13.
J Neurol Sci ; 400: 160-168, 2019 May 15.
Article in English | MEDLINE | ID: mdl-30954660

ABSTRACT

The gut microbiome is composed of an enormous number of microorganisms, generally regarded as commensal bacteria. Resident gut bacteria are an important contributor to health and significant evidence suggests that the presence of healthy and diverse gut microbiota is important for normal cognitive and emotional processing. Here we measured the expression of monoamine neurotransmitter-related genes in the hippocampus of germ-free (GF) mice and specific-pathogen-free (SPF) mice to explore the effect of gut microbiota on hippocampal monoamine functioning. In total, 19 differential expressed genes (Htr7, Htr1f, Htr3b, Drd3, Ddc, Maob, Tdo2, Fos, Creb1, Akt1, Gsk3a, Pik3ca, Pla2g5, Cyp2d22, Grk6, Ephb1, Slc18a1, Nr4a1, Gdnf) that could discriminate between the two groups were identified. Interestingly, GF mice displayed anxiolytic-like behavior compared to SPF mice, which were not reversed by colonization with gut microbiota from SPF mice. Besides, colonization of adolescent GF mice by gut microbiota was not sufficient to reverse the altered gene expression associated with their GF status. Taking these findings together, the absence of commensal microbiota during early life markedly affects hippocampal monoamine gene-regulation, which was associated with anxiolytic behaviors and monoamine neurological signs.


Subject(s)
Anxiety/genetics , Gastrointestinal Microbiome/physiology , Germ-Free Life/physiology , Hippocampus/metabolism , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Animals , Anxiety/etiology , Anxiety/psychology , Biogenic Monoamines/metabolism , Gene Regulatory Networks/physiology , Male , Mice , Mice, Inbred BALB C , Random Allocation
14.
Behav Brain Res ; 359: 686-693, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30261201

ABSTRACT

Rats detect and use odorant molecules as a source of information about their environment. Some of these molecules come from conspecifics, and many arise as by-products from microbial activity. Thus, compared to conventionally housed rats, germ-free rats are raised in an environment with fewer odorants, but this reduction is rarely quantified. Using gas chromatography-mass spectrometry, we found that germ-free rat faeces samples contained half as many volatile molecules than conventional rat faeces (52 vs 109 (±2.4) molecules; P < 0.001) and overall these were only 12% as abundant. We then investigated if odours from female germ-free rats in oestrus would have pro-erectile effects in conventional male rats. For this aim, conventionally housed Brown Norway (BN) rats (n = 16) with sexual experience with either Fischer or BN females, were exposed to four different odour types: faeces from germ-free Fischer rat in oestrus, faeces from conventional rats in oestrus and di-oestrus (either from Fischer or BN), and a control (either 1-hexanol or male rat faeces). The number of penile erections per test as well as the duration of freezing behaviour was significantly higher with the oestrous odours (germ-free and conventional) compared to the control, with intermediate responses to the di-oestrous faeces. The findings indicate that, despite a significantly reduced composition in terms of volatiles compared to conventionally housed rats, the faeces of germ-free rats contain sufficient odorants to evoke sexual responses in conventional male rats. Oestrous odours of rats thus appear not to be of microbial origin.


Subject(s)
Estrus , Germ-Free Life , Olfactory Perception , Sexual Behavior, Animal , Animals , Estrus/physiology , Feces/chemistry , Female , Germ-Free Life/physiology , Male , Odorants , Penile Erection , Random Allocation , Rats , Sexual Behavior, Animal/physiology , Volatile Organic Compounds/analysis
15.
Lab Anim ; 53(3): 271-280, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30580671

ABSTRACT

The world-wide incidence of many immune-mediated and metabolic diseases, including those of the intestines and liver, is steadily increasing. Gut microbiota plays a central role in the pathogenesis of these diseases as it mediates environmental changes to the intestinal immune system. Various environmental factors including diet, food additives and medication also trigger the compositional and functional alterations of microbiota, that is, dysbiosis, and this dysbiosis is closely associated with many chronic inflammatory diseases. However, the causal relationship remains unclear for the majority of these diseases. In this review, we discuss essential epidemiological data, known pathogenetic factors including those of genetic and environmental nature, while mainly focusing on the role of gut microbiota in the development of selected intestinal and liver diseases. Using specific examples, we also briefly describe some of the most widely-used animal models including gnotobiotic models and their contribution to the research of pathogenetic mechanisms of the host-microbiota relationship.


Subject(s)
Dysbiosis/physiopathology , Gastrointestinal Microbiome/physiology , Germ-Free Life/physiology , Intestinal Diseases/physiopathology , Liver Diseases/physiopathology , Animals , Disease Models, Animal , Dysbiosis/microbiology , Intestinal Diseases/microbiology , Liver Diseases/microbiology , Mice
16.
Sci Rep ; 8(1): 14184, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242285

ABSTRACT

The intestinal barrier encompasses structural, permeability and immune aspects of the gut mucosa that, when disrupted, may contribute to chronic inflammation. Although gnotobiotic studies have demonstrated the effects of microbiota on mucosal and systemic immunity, as well as intestinal barrier architecture and innate immune characteristics, its impact on barrier function remains unclear. We compared germ-free and conventional mice, as well as mice colonized with human fecal microbiota that were followed for 21 days post-colonization. Colonic barrier structure was investigated by immunohistochemistry, molecular and electron microscopy techniques. Permeability was assessed in colon tissue by Ussing chambers, and by serum LPS and MDP detection using TLR4- and NOD2-NFκB reporter assays. Microbiota profile was determined by Illumina 16S rRNA gene sequencing. Low dose dextran sodium sulfate was administered to assess microbiota-induced barrier changes on resistance to colonic injury. Permeability to paracellular probes and mucus layer structure resembled that of conventional mice by day 7 post-colonization, coinciding with reduced claudin-1 expression and transient IL-18 production by intestinal epithelial cells. These post-colonization adaptations were associated with decreased systemic bacterial antigen exposure and reduced susceptibility to intestinal injury. In conclusion, commensal colonization promotes physiological barrier structural and functional adaptations that contribute to intestinal homeostasis.


Subject(s)
Colon/microbiology , Colon/physiology , Gastrointestinal Microbiome/physiology , Homeostasis/physiology , Microbiota/physiology , Animals , Colon/drug effects , Dextran Sulfate/pharmacology , Feces , Female , Gastrointestinal Microbiome/drug effects , Germ-Free Life/drug effects , Germ-Free Life/physiology , Homeostasis/drug effects , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/physiopathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiology , Intestines/drug effects , Intestines/microbiology , Intestines/physiology , Male , Mice , Mice, Inbred C57BL , Microbiota/drug effects , Permeability/drug effects , RNA, Ribosomal, 16S/metabolism
17.
Sci Rep ; 8(1): 10646, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30006625

ABSTRACT

In animals, commensal microbes modulate various physiological functions, including behavior. While microbiota exposure is required for normal behavior in mammals, it is not known how widely this dependency is present in other animal species. We proposed the hypothesis that the microbiome has a major influence on the behavior of the vinegar fly (Drosophila melanogaster), a major invertebrate model organism. Several assays were used to test the contribution of the microbiome on some well-characterized behaviors: defensive behavior, sleep, locomotion, and courtship in microbe-bearing, control flies and two generations of germ-free animals. None of the behaviors were largely influenced by the absence of a microbiome, and the small or moderate effects were not generalizable between replicates and/or generations. These results refute the hypothesis, indicating that the Drosophila microbiome does not have a major influence over several behaviors fundamental to the animal's survival and reproduction. The impact of commensal microbes on animal behaviour may not be broadly conserved.


Subject(s)
Behavior, Animal/physiology , Drosophila melanogaster/physiology , Germ-Free Life/physiology , Host Microbial Interactions/physiology , Microbiota/physiology , Animals , Courtship , Drosophila melanogaster/microbiology , Female , Locomotion/physiology , Male , Sleep/physiology , Symbiosis/physiology
18.
Cell Microbiol ; 20(10): e12867, 2018 10.
Article in English | MEDLINE | ID: mdl-29895095

ABSTRACT

Valosin-containing protein (VCP)/p97/Cdc48 is one of the best-characterised type II cytosolic AAA+ ATPases most known for their role in ubiquitin-dependent protein quality control. Here, we provide functional insights into the role of the Leishmania VCP/p97 homologue (LiVCP) in the parasite intracellular development. We demonstrate that although LiVCP is an essential gene, Leishmania infantum promastigotes can grow with less VCP. In contrast, growth of axenic and intracellular amastigotes is dramatically affected upon decreased LiVCP levels in heterozygous and temperature sensitive (ts) LiVCP mutants or the expression of dominant negative mutants known to specifically target the second conserved VCP ATPase domain, a major contributor of the VCP overall ATPase activity. Interestingly, these VCP mutants are also unable to survive heat stress, and a ts VCP mutant is defective in amastigote growth. Consistent with LiVCP's essential function in amastigotes, LiVCP messenger ribonucleic acid undergoes 3'Untranslated Region (UTR)-mediated developmental regulation, resulting in higher VCP expression in amastigotes. Furthermore, we show that parasite mutant lines expressing lower VCP levels or dominant negative VCP forms exhibit high accumulation of polyubiquitinated proteins and increased sensitivity to proteotoxic stress, supporting the ubiquitin-selective chaperone function of LiVCP. Together, these results emphasise the crucial role LiVCP plays under heat stress and during the parasite intracellular development.


Subject(s)
Intracellular Space/parasitology , Leishmania infantum/growth & development , Valosin Containing Protein/metabolism , 3' Untranslated Regions/genetics , Base Sequence/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/genetics , Germ-Free Life/physiology , Heat-Shock Response/physiology , Leishmania infantum/genetics , Molecular Chaperones/metabolism , Protein Domains/genetics , RNA, Messenger/genetics , Ubiquitin/metabolism , Ubiquitination , Valosin Containing Protein/genetics
19.
PLoS One ; 13(5): e0196510, 2018.
Article in English | MEDLINE | ID: mdl-29763437

ABSTRACT

Accumulating studies have defined a role for the intestinal microbiota in modulation of host behavior. Research using gnotobiotic mice emphasizes that early microbial colonization with a complex microbiota (conventionalization) can rescue some of the behavioral abnormalities observed in mice that grow to adulthood completely devoid of bacteria (germ-free mice). However, the human infant and adult microbiomes vary greatly, and effects of the neonatal microbiome on neurodevelopment are currently not well understood. Microbe-mediated modulation of neural circuit patterning in the brain during neurodevelopment may have significant long-term implications that we are only beginning to appreciate. Modulation of the host central nervous system by the early-life microbiota is predicted to have pervasive and lasting effects on brain function and behavior. We sought to replicate this early microbe-host interaction by colonizing gnotobiotic mice at the neonatal stage with a simplified model of the human infant gut microbiota. This model consortium consisted of four "infant-type" Bifidobacterium species known to be commensal members of the human infant microbiota present in high abundance during postnatal development. Germ-free mice and mice neonatally-colonized with a complex, conventional murine microbiota were used for comparison. Motor and non-motor behaviors of the mice were tested at 6-7 weeks of age, and colonization patterns were characterized by 16S ribosomal RNA gene sequencing. Adult germ-free mice were observed to have abnormal memory, sociability, anxiety-like behaviors, and motor performance. Conventionalization at the neonatal stage rescued these behavioral abnormalities, and mice colonized with Bifidobacterium spp. also exhibited important behavioral differences relative to the germ-free controls. The ability of Bifidobacterium spp. to improve the recognition memory of both male and female germ-free mice was a prominent finding. Together, these data demonstrate that the early-life gut microbiome, and human "infant-type" Bifidobacterium species, affect adult behavior in a strongly sex-dependent manner, and can selectively recapitulate the results observed when mice are colonized with a complex microbiota.


Subject(s)
Behavior, Animal/physiology , Bifidobacterium/physiology , Gastrointestinal Microbiome/physiology , Germ-Free Life/physiology , Animals , Animals, Newborn , Anxiety/microbiology , Bifidobacterium/genetics , Bifidobacterium/growth & development , Female , Gastrointestinal Microbiome/genetics , Humans , Infant , Male , Memory/physiology , Mice , Models, Animal , Motor Skills/physiology , Social Behavior , Specific Pathogen-Free Organisms/physiology
20.
JCI Insight ; 3(5)2018 03 08.
Article in English | MEDLINE | ID: mdl-29515039

ABSTRACT

Current obesity interventions suffer from lack of durable effects and undesirable complications. Fumagillin, an inhibitor of methionine aminopeptidase-2, causes weight loss by reducing food intake, but with effects on weight that are superior to pair-feeding. Here, we show that feeding of rats on a high-fat diet supplemented with fumagillin (HF/FG) suppresses the aggressive feeding observed in pair-fed controls (HF/PF) and alters expression of circadian genes relative to the HF/PF group. Multiple indices of reduced energy expenditure are observed in HF/FG but not HF/PF rats. HF/FG rats also exhibit changes in gut hormones linked to food intake, increased energy harvest by gut microbiota, and caloric spilling in the urine. Studies in gnotobiotic mice reveal that effects of fumagillin on energy expenditure but not feeding behavior may be mediated by the gut microbiota. In sum, fumagillin engages weight loss-inducing behavioral and physiologic circuits distinct from those activated by simple caloric restriction.


Subject(s)
Bacteria/isolation & purification , Cyclohexanes/administration & dosage , Energy Metabolism/drug effects , Fatty Acids, Unsaturated/administration & dosage , Gastrointestinal Microbiome/drug effects , Obesity/drug therapy , Aminopeptidases/antagonists & inhibitors , Animals , Bacteria/drug effects , Bacteria/metabolism , Behavior, Animal/drug effects , Body Weight/drug effects , Diet, High-Fat/adverse effects , Disease Models, Animal , Feces/microbiology , Feeding Behavior/drug effects , Gastrointestinal Microbiome/physiology , Germ-Free Life/drug effects , Germ-Free Life/physiology , Glycoproteins/antagonists & inhibitors , Humans , Male , Methionyl Aminopeptidases , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Rats , Rats, Wistar , Sesquiterpenes/administration & dosage , Treatment Outcome , Weight Loss/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...