Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Gut Microbes ; 14(1): 2149023, 2022.
Article in English | MEDLINE | ID: mdl-36420990

ABSTRACT

The mechanisms by which early microbial colonizers of the neonate influence gut development are poorly understood. Bacterial bile salt hydrolase (BSH) acts as a putative colonization factor that influences bile acid signatures and microbe-host signaling pathways and we considered whether this activity can influence infant gut development. In silico analysis of the human neonatal gut metagenome confirmed that BSH enzyme sequences are present as early as one day postpartum. Gastrointestinal delivery of cloned BSH to immature gnotobiotic mice accelerated shortening of the colon and regularized gene expression profiles, with monocolonised mice more closely resembling conventionally raised animals. In situ expression of BSH decreased markers of cell proliferation (Ki67, Hes2 and Ascl2) and strongly increased expression of ALPI, a marker of cell differentiation and barrier function. These data suggest an evolutionary paradigm whereby microbial BSH activity potentially influences bacterial colonization and in-turn benefits host gastrointestinal maturation.


Subject(s)
Gastrointestinal Microbiome , Transcriptome , Female , Humans , Mice , Animals , Amidohydrolases/genetics , Amidohydrolases/metabolism , Gastrointestinal Tract/microbiology , Bacteria/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism
2.
Am J Physiol Endocrinol Metab ; 322(4): E366-E381, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35224983

ABSTRACT

This review aims to describe and compare porcine models of metabolic syndrome. This syndrome and its associated secondary comorbidities are set to become the greatest challenge to healthcare providers and policy makers in the coming century. However, an incomplete understanding of the pathogenesis has left significant knowledge gaps in terms of efficacious therapeutics. To further our comprehension and, in turn, management of metabolic syndrome, appropriate high-fidelity models of the disease complex are of great importance. In this context, our review aims to assess the most promising porcine models of metabolic syndrome currently available for their similarity to the human phenotype. In addition, we aim to highlight the strengths and shortcomings of each model in an attempt to identify the most appropriate application of each. Although no porcine model perfectly recapitulates the human metabolic syndrome, several pose satisfactory approximations. The Ossabaw miniature swine in particular represents a highly translatable model that develops each of the core parameters of the syndrome with many of the associated secondary comorbidities. Future high-fidelity porcine models of metabolic syndrome need to focus on secondary sequelae replication, which may require extended induction period to reveal.


Subject(s)
Metabolic Syndrome , Animals , Disease Models, Animal , Disease Progression , Metabolic Syndrome/etiology , Swine , Swine, Miniature
3.
Am J Physiol Heart Circ Physiol ; 318(3): H590-H603, 2020 03 01.
Article in English | MEDLINE | ID: mdl-32031871

ABSTRACT

Metabolic syndrome (MetS) is a composite of cardiometabolic risk factors, including obesity, dyslipidemia, hypertension, and insulin resistance, with a range of secondary sequelae such as nonalcoholic fatty liver disease and diastolic heart failure. This syndrome has been identified as one of the greatest global health challenges of the 21st century. Herein, we examine whether a porcine model of diet- and mineralocorticoid-induced MetS closely mimics the cardiovascular, metabolic, gut microbiota, and functional metataxonomic phenotype observed in human studies. Landrace pigs with deoxycorticosterone acetate-induced hypertension fed a diet high in fat, salt, and sugar over 12 wk were assessed for hyperlipidemia, hyperinsulinemia, and immunohistologic, echocardiographic, and hemodynamic parameters, as well as assessed for microbiome phenotype and function through 16S rRNA metataxonomic and metabolomic analysis, respectively. All MetS animals developed obesity, hyperlipidemia, insulin resistance, hypertension, fatty liver, structural cardiovascular changes including left ventricular hypertrophy and left atrial enlargement, and increased circulating saturated fatty acid levels, all in keeping with the human phenotype. A reduction in α-diversity and specific microbiota changes at phylum, family, and genus levels were also observed in this model. Specifically, this porcine model of MetS displayed increased abundances of proinflammatory bacteria coupled with increased circulating tumor necrosis factor-α and increased secondary bile acid-producing bacteria, which substantially impacted fibroblast growth factor-19 expression. Finally, a significant decrease in enteroprotective bacteria and a reduction in short-chain fatty acid-producing bacteria were also noted. Together, these data suggest that diet and mineralocorticoid-mediated development of biochemical and cardiovascular stigmata of metabolic syndrome in pigs leads to temporal gut microbiome changes that mimic key gut microbial population signatures in human cardiometabolic disease.NEW & NOTEWORTHY This study extends a prior porcine model of cardiometabolic syndrome to include systemic inflammation, fatty liver, and insulin sensitivity. Gut microbiome changes during evolution of porcine cardiometabolic disease recapitulate those in human subjects with alterations in gut taxa associated with proinflammatory bacteria, bile acid, and fatty acid pathways. This clinical scale model may facilitate design of future interventional trials to test causal relationships between gut dysbiosis and cardiometabolic syndrome at a systemic and organ level.


Subject(s)
Gastrointestinal Microbiome/physiology , Hypertension/microbiology , Insulin Resistance/physiology , Metabolic Syndrome/microbiology , Non-alcoholic Fatty Liver Disease/microbiology , Animals , Blood Glucose/metabolism , Cholesterol/blood , Diet, High-Fat , Disease Models, Animal , Echocardiography , Female , Hypertension/metabolism , Inflammation/metabolism , Inflammation/microbiology , Insulin/blood , Metabolic Syndrome/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Swine , Triglycerides/blood
4.
Neurosci Lett ; 711: 134409, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31374323

ABSTRACT

Administration of the mixed opioid agonist-antagonist butorphanol tartrate (BT) has been shown to robustly increase food intake in rodent models utilizing adult and young animals. BT at orexigenic doses increases c-Fos-immunoreactivity (IR) in brain areas associated with feeding for energy as well as for reward, including the paraventricular nucleus of the hypothalamus, central nucleus of the amygdala and nucleus of the solitary tract. Interestingly, aged rats given standard chow show a diminished feeding response to BT. It is not known, however, whether this weakened orexigenic response in aged animals extends to palatable tastants and whether it is accompanied by changes in brain activation. In the current study, we injected adult (11-12 months) and aged (26-27 months) rats with BT and studied the effect on intake of chow and palatable ingestants (liquid and solid). We found that BT produced only a moderate increase in consumption of bland or palatable chow as well as sweet solutions (both caloric and non-caloric) in aged rats, and that higher BT doses are required to generate such eating in old animals compared to adults. This blunted hyperphagia after BT is accompanied by diminished c-Fos IR in the central and basolateral amygdala, regions that process emotional aspects of behaviors, including food intake. Thus, aged rats exhibit diminished responsiveness to the feeding effects of BT, independent of the type of diet; and it appears to be due, in part, to diminished neural activity in central circuits involved in emotional behavior.


Subject(s)
Aging/physiology , Brain/drug effects , Butorphanol/pharmacology , Feeding Behavior/drug effects , Narcotic Antagonists/pharmacology , Animals , Brain/metabolism , Hyperphagia/chemically induced , Male , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley
5.
BMC Neurosci ; 16: 13, 2015 Mar 14.
Article in English | MEDLINE | ID: mdl-25887538

ABSTRACT

BACKGROUND: Obesity is a growing global concern with strong associations with cardiovascular disease, cancer and type-2 diabetes. Although various genome-wide association studies have identified more than 40 genes associated with obesity, these genes cannot fully explain the heritability of obesity, suggesting there may be other contributing factors, including epigenetic effects. RESULTS: We performed genome wide DNA methylation profiling comparing normal-weight and obese 9-13 year old children to investigate possible epigenetic changes correlated with obesity. Of note, obese children had significantly lower methylation levels at a CpG site located near coronin 7 (CORO7), which encodes a tryptophan-aspartic acid dipeptide (WD)-repeat containing protein most likely involved in Golgi complex morphology and function. Anatomical profiling of coronin 7 (Coro7) mRNA expression in mice revealed that it is highly expressed in appetite and energy balance regulating regions, including the hypothalamus, striatum and locus coeruleus, the main noradrenergic brain site. Interestingly, we found that food deprivation in mice downregulates hypothalamic Coro7 mRNA levels, and injecting ethanol, an appetite stimulant, increased the number of Coro7 expressing cells in the locus coeruleus. Finally, by employing the genetically-tractable Drosophila melanogaster model we were able to demonstrate an evolutionarily conserved metabolic function for the CORO7 homologue pod1. Knocking down the pod1 in the Drosophila adult nervous system increased their resistance to starvation. Furthermore, feeding flies a high-calorie diet significantly increased pod1 expression. CONCLUSION: We conclude that coronin 7 is involved in the regulation of energy homeostasis and this role stems, to some degree, from the effect on feeding for calories and reward.


Subject(s)
Body Weight/physiology , Drosophila Proteins/metabolism , Microfilament Proteins/metabolism , Adolescent , Animals , Appetite Stimulants/pharmacology , Brain/drug effects , Brain/metabolism , Child , Cohort Studies , Diet, High-Fat , Drosophila Proteins/genetics , Drosophila melanogaster , Ethanol/pharmacology , Female , Food Deprivation/physiology , Gene Knockdown Techniques , Humans , Male , Mice, Inbred C57BL , Microfilament Proteins/genetics , Obesity/genetics , RNA, Messenger/metabolism , Starvation/metabolism
6.
Neuroreport ; 25(12): 909-14, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-24893201

ABSTRACT

Centrally acting oxytocin (OT) inhibits feeding. Recent evidence suggests a link between OT and control of carbohydrate and saccharin intake, but it is unclear whether OT affects appetite for only carbohydrates, especially sweet ones, or sweet tastants irrespective of their carbohydrate content. Therefore, a blood-brain barrier penetrant OT receptor antagonist, L-368,899, was administered in mice and intake of liquid diets containing carbohydrates sucrose, glucose, fructose, polycose, or cornstarch (CS) or the noncarbohydrate, noncaloric sweetener saccharin was studied in episodic intake paradigms: one in which only one tastant was available and the other in which a choice between a carbohydrate (sucrose, glucose, or fructose) and saccharin was provided. We also used real-time PCR to examine hypothalamic Ot mRNA levels in mice provided short-term access to sucrose, CS, or saccharin. In the no-choice paradigm, L-368,899 increased the intake of all carbohydrates, whereas its effect on saccharin consumption showed only a trend. A 10 times lower dose (0.3 mg/kg) stimulated intake of sucrose than other carbohydrates. In the choice test, a very low 0.1 mg/kg dose of L-368,899 doubled the proportion of sucrose consumption relative to saccharin, but did not affect fructose or glucose preference. Ot gene expression increased after sucrose and CS, but not saccharin exposure compared with the controls; however, a higher level of significance was detected in the sucrose group. We conclude that OT inhibits appetite for carbohydrates. Sucrose consumption considerably enhances Ot gene expression and is particularly sensitive to OT receptor blockade, suggesting a special functional relationship between OT and sugar intake.


Subject(s)
Appetite/physiology , Dietary Carbohydrates/administration & dosage , Food Preferences/physiology , Oxytocin/metabolism , Saccharin/administration & dosage , Animals , Appetite/drug effects , Camphanes/pharmacology , Central Nervous System Agents/pharmacology , Choice Behavior/drug effects , Choice Behavior/physiology , Dose-Response Relationship, Drug , Food Preferences/drug effects , Hypothalamus/drug effects , Hypothalamus/physiology , Male , Mice, Inbred C57BL , Piperazines/pharmacology , RNA, Messenger/metabolism , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...