Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Neurol Res ; 41(12): 1104-1112, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31587617

ABSTRACT

Objectives: Lipid metabolism is closely associated with many important biological functions. Here, we conducted this study to explore the effects of gut microbiota on the lipid metabolism in the prefrontal cortex of mice. Methods: Germ-free (GF) mice, specific pathogen-free (SPF) and colonized GF (CGF) mice were used in this study. The open field test (OFT), forced swimming test (FST) and novelty suppressed feeding test (NSFT) were conducted to assess the changes in general behavioral activity. The liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) was used to obtain the lipid metabolites. Both one-way analysis of variance (one-way ANOVA) and orthogonal partial least-squares discriminant analysis (OPLS-DA) were used to obtain the key differential lipid metabolites. Results: The behavioral tests showed that compared to SPF mice, GF mice had more center distance, more center time, less immobility time and less latency to familiar food. Meanwhile, 142 key differential lipid metabolites between SPF mice and GF mice were identified. These lipid metabolites mainly belonged to glycerophospholipids, glycerolipids, sphingolipids, and saccharolipids. The gut microbiota colonization did not reverse these changed behavioral phenotypes, but could restore 25 key differential lipid metabolites. Discussion: These results showed that the absence of gut microbiota could influence host behaviors and lipid metabolism. Our findings could provide original and valuable data for future studies to further investigate the microbiota-gut-brain axis.


Subject(s)
Behavior, Animal , Gastrointestinal Microbiome/physiology , Lipid Metabolism , Prefrontal Cortex/metabolism , Animals , Anxiety/metabolism , Anxiety/microbiology , Depression/metabolism , Depression/microbiology , Male , Metabolomics , Mice, Inbred BALB C , Specific Pathogen-Free Organisms
2.
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
3.
Behav Brain Res ; 322(Pt A): 34-41, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28093256

ABSTRACT

BACKGROUNDS: Gut microbiota is increasingly recognized as an important environmental factor that could influence the brain function and behaviors through the microbiota-gut-brain axis. METHOD: Here, we used the germ-free (GF) mice to explore the effect of gut microbiota on hippocampal microRNA (miRNA) and messenger RNAs (mRNAs) expression. RESULTS: Behavioral tests showed that, compared to specific pathogen-free (SPF) mice, the GF mice displayed more center time, center distance and less latency to familiar food. Colonization of the GF mice with gut microbiota from SPF mice did not reverse these behaviors. However, 7 differentially expressed miRNAs and 139 mRNAs were significantly restored. Through microRNA Target Filter analysis, 4 of 7 restored miRNAs had 2232 target mRNAs. Among these target mRNAs, 21 target mRNAs levels were decreased. Further analysis showed that the most significant GO terms were metabolic process (GO: 0008152), binding (GO: 0005488) and cell part (GO: 0044464) for biological process, molecular function and cellular component, respectively, and the most significantly altered pathway was axon guidance (mmu04360). CONCLUSIONS: These findings indicated that colonization of gut microbiota to adolescent GF mice was not sufficient to reverse the behavioral alterations. Gut microbiota could significantly influence the expression levels of miRNAs and mRNAs in hippocampus. Our results could provide original and valuable data for researchers to further study the microbiota-gut-brain axis.


Subject(s)
Gastrointestinal Microbiome , Hippocampus/metabolism , MicroRNAs/metabolism , RNA, Messenger/metabolism , Animals , Anxiety/metabolism , Anxiety/microbiology , Exploratory Behavior , Feeding Behavior , Male , Mice, Inbred BALB C , Motor Activity , Psychological Tests , Specific Pathogen-Free Organisms , Stress, Psychological/metabolism , Stress, Psychological/microbiology
4.
Front Nutr ; 3: 42, 2016.
Article in English | MEDLINE | ID: mdl-28066771

ABSTRACT

There is an increasing interest in the effect of dietary polyphenols on the intestinal microbiota and the possible associations between this effect and the development of some cardiovascular diseases, such as atherosclerosis (AS). However, limited information is available on how these polyphenols affect the gut microbiota and AS development. This study was designed to evaluate the modulation of dietary tea polyphenols (TPs) on intestinal Bifidobacteria (IB) and its correlation with AS development in apolipoprotein E-deficient (ApoE-/-) mice. Fifty C57BL/6 ApoE-/- mice were randomized into one of the five treatment groups (n = 10/group): control group fed normal diet (CK); a group fed a high-fat diet (HFD); and the other three groups fed the same HFD supplemented with TPs in drinking water for 16 weeks. The total cholesterol and low-density lipoprotein cholesterol (LDL-C) were decreased significantly (P < 0.05) after TP interference. In addition, the TP diet also decreased the plaque area/lumen area (PA/LA) ratios (P < 0.01) in the TP diet group. Interestingly, copies of IB in the gut of ApoE-/- mice were notably increased with TP interference. This increase was dose dependent (P < 0.01) and negatively correlated with the PA/LA ratio (P < 0.05). We conclude that TPs could promote the proliferation of the IB, which is partially responsible for the reduction of AS plaque induced by HFD.

5.
Sci Rep ; 5: 10512, 2015 May 27.
Article in English | MEDLINE | ID: mdl-26015368

ABSTRACT

We have previously observed that knockout of Niemann-Pick C1-Like 1 (NPC1L1), a cholesterol transporter essential for intestinal cholesterol absorption, reduces the output of dry stool in mice. As the food intake remains unaltered in NPC1L1-knockout (L1-KO) mice, we hypothesized that NPC1L1 deficiency may alter the gut microbiome to reduce stool output. Consistently, here we demonstrate that the phyla of fecal microbiota differ substantially between L1-KO mice and their wild-type controls. Germ-free (GF) mice have reduced stool output. Inhibition of NPC1L1 by its inhibitor ezetimibe reduces stool output in specific pathogen-free (SPF), but not GF mice. In addition, we show that GF versus SPF mice have reduced intestinal absorption and increased fecal excretion of cholesterol, particularly after treatment with ezetimibe. This negative balance of cholesterol in GF mice is associated with reduced plasma and hepatic cholesterol, and likely caused by reduced expression of NPC1L1 and increased expression of ABCG5 and ABCG8 in small intestine. Expression levels of other genes in intestine and liver largely reflect a state of cholesterol depletion and a decrease in intestinal sensing of bile acids. Altogether, our findings reveal a broad role of microbiota in regulating whole-body cholesterol homeostasis and its response to a cholesterol-lowering drug, ezetimibe.


Subject(s)
Bacteria/isolation & purification , Cholesterol/metabolism , Gene Expression Regulation , Intestine, Small/microbiology , Microbiota , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Anticholesteremic Agents/pharmacology , Bacteria/genetics , Bile Acids and Salts/metabolism , Body Weight/drug effects , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Cytochrome P450 Family 7 , Diet, High-Fat , Ezetimibe/pharmacology , Feces/microbiology , Intestinal Absorption/drug effects , Intestine, Small/metabolism , Lipid Metabolism/genetics , Lipids/blood , Lipoproteins/genetics , Lipoproteins/metabolism , Liver/drug effects , Liver/metabolism , Male , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/genetics , Steroid Hydroxylases/metabolism , Up-Regulation
6.
Vascular ; 22(5): 328-35, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23929425

ABSTRACT

PURPOSE: To determine the effects of hypercholesterolemia in pregnant mice on the susceptibility to atherosclerosis in adult life through a new animal modeling approach. METHODS: Male offspring from apoE-/- mice fed with regular (R) or high (H) cholesterol chow during pregnancy were randomly subjected to regular (Groups R-R and H-R, n = 10) or high cholesterol diet (Groups R-H and H-H, n = 10) for 14 weeks. Plasma lipid profiles were determined in all rats. The abdominal aorta was examined for the severity of atherosclerotic lesions in offspring. RESULTS: Lipids significantly increased while high-density lipoprotein-cholesterol/low-density lipoprotein-cholesterol decreased in mothers fed high cholesterol chow after delivery compared with before pregnancy (p < 0.01). Groups R-H and H-R indicated dyslipidemia and significant atherosclerotic lesions. Group H-H demonstrated the highest lipids, lowest high-density lipoprotein-cholesterol/low-density lipoprotein-cholesterol, highest incidence (90%), plaque area to luminal area ratio (0.78 ± 0.02) and intima to media ratio (1.57 ± 0.05). CONCLUSION: Hypercholesterolemia in pregnant mice may increase susceptibility to atherosclerosis in their adult offspring.


Subject(s)
Aorta, Abdominal/pathology , Atherosclerosis/etiology , Atherosclerosis/pathology , Disease Susceptibility , Hypercholesterolemia/complications , Hypercholesterolemia/pathology , Lipids/blood , Animals , Disease Models, Animal , Female , Immunoenzyme Techniques , Male , Matrix Metalloproteinase 9/blood , Mice , Mice, Knockout , Pregnancy , Random Allocation , Risk Factors
7.
PLoS One ; 7(1): e30560, 2012.
Article in English | MEDLINE | ID: mdl-22291988

ABSTRACT

Cytochomosome P450 enzymes (CYP) are heme-containing monooxygenases responsible for oxidative metabolism of many exogenous and endogenous compounds including drugs. The species difference of CYP limits the extent to which data obtained from animals can be translated to humans in pharmacodynamics or pharmacokinetics studies. Transgenic expression of human CYP in animals lacking or with largely reduced endogenous CYP counterparts is recognized as an ideal strategy to correct CYP species difference. CYP3A is the most abundant CYP subfamily both in human and mammals. In this study, we designed a microRNA-based shRNA (miR-shRNA) simultaneously targeting four members of mouse CYP3A subfamily (CYP3A11, CYP3A16, CYP3A41 and CYP3A44), and transgenic mice expressing the designed miR-shRNA were generated by lentiviral transgenesis. Results showed that the CYP3A expression level in transgenic mice was markedly reduced compared to that in wild type or unrelated miR-shRNA transgenic mice, and was inversely correlated to the miR-shRNA expression level. The CYP3A expression levels in transgenic offspring of different generations were also remarkably lower compared to those of controls, and moreover the inhibition rate of CYP3A expression remained comparable over generations. The ratio of the targeted CYP3A transcriptional levels was comparable between knockdown and control mice of the same gender as detected by RT-PCR DGGE analysis. These data suggested that transgenic miR-shRNA suppressed CYP3A expression in a dose-dependent and inheritable manner, and transcriptional levels of the targeted CYP3As were suppressed to a similar extent. The observed knockdown efficacy was further confirmed by enzymatic activity analysis, and data showed that CYP3A activities in transgenic mice were markedly reduced compared to those in wild-type or unrelated miR-shRNA transgenic controls (1.11±0.71 vs 5.85±1.74, 5.9±2.4; P<0.01). This work laid down a foundation to further knock down the remaining murine CYP3As or CYPs of other subfamilies, and a basis to generate CYP knockdown animals of other species.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , MicroRNAs/genetics , MicroRNAs/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Animals , Cells, Cultured , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Dose-Response Relationship, Drug , Female , Gene Dosage/physiology , Genetic Vectors/genetics , Humans , Inheritance Patterns/drug effects , Inheritance Patterns/genetics , Inheritance Patterns/physiology , Lentivirus/genetics , Male , Mice , Mice, Transgenic , RNA Interference/drug effects , RNA Interference/physiology
8.
Comp Med ; 58(6): 580-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19149415

ABSTRACT

The objective of this study was to demonstrate that Bama miniature pigs are a suitable experimental animal model for the evaluation of drugs for man. To this end, in vitro lovastatin metabolism at the minipig liver microsomal level and in vivo pharmacokinetics were studied. Results were compared with those obtained from humans. Our data indicate that the main metabolites and enzyme kinetic parameters of lovastatin metabolism are similar in pigs and humans. Triacetyloleandomycin, a specific inhibitor of human CYP3A4, inhibited the metabolism of lovastatin in pig and human liver microsomes. In addition, the pharmacokinetic parameters and absolute bioavailability suggested that the absorption and elimination of lovastatin in Bama miniature pigs were similar to those in humans. Lovastatin was distributed across many organs in pigs, but the highest levels were found in the stomach, intestines, and liver. Within 96 h, 7% and 82% of the given dose was excreted in the urine and feces, respectively. In addition, no significant species differences in the plasma protein binding ratio of lovastatin and the rates of lovastatin hydrolysis to beta-hydroxyacid lovastatin were apparent. From these results, we conclude that Bama miniature pigs are suitable for use in drug evaluation studies.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Lovastatin/metabolism , Lovastatin/pharmacokinetics , Swine, Miniature/metabolism , Animals , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Humans , Hydrolysis , In Vitro Techniques , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Animal , Species Specificity , Swine , Troleandomycin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL