Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nutrients ; 15(22)2023 Nov 17.
Article in English | MEDLINE | ID: mdl-38004202

ABSTRACT

Diet-induced obesity impairs mitochondrial respiratory responses in tissues that are highly metabolically active, such as the heart. However, less is known about the impact of obesity on the respiratory activity of specific cell types, such as splenic B cells. B cells are of relevance, as they play functional roles in obesity-induced insulin resistance, inflammation, and responses to infection. Here, we tested the hypothesis that high-fat-diet (HFD)-induced obesity could impair the mitochondrial respiration of intact and permeabilized splenic CD19+ B cells isolated from C57BL/6J mice and activated ex vivo with lipopolysaccharide (LPS). High-resolution respirometry was used with intact and permeabilized cells. To reveal potential mechanistic targets by which HFD-induced obesity dysregulates B cell mitochondria, we conducted proteomic analyses and 3D serial block face scanning electron microscopy (SBFEM). High-resolution respirometry revealed that intact LPS-stimulated B cells of obese mice, relative to controls, displayed lower ATP-linked, as well as maximal uncoupled, respiration. To directly investigate mitochondrial function, we used permeabilized LPS-stimulated B cells, which displayed increased H2O2 emission and production with obesity. We also examined oxidative phosphorylation efficiency simultaneously, which revealed that oxygen consumption and ATP production were decreased in LPS-stimulated B cells with obesity relative to controls. Despite minimal changes in total respiratory complex abundance, in LPS-stimulated B cells of obese mice, three of the top ten most downregulated proteins were all accessory subunits of respiratory complex I. SBFEM showed that B cells of obese mice, compared to controls, underwent no change in mitochondrial cristae integrity but displayed increased mitochondrial volume that was linked to bioenergetic function. Collectively, these results establish a proof of concept that HFD-induced obesity dysregulates the mitochondrial bioenergetic metabolism of activated splenic B cells.


Subject(s)
Diet, High-Fat , Insulin Resistance , Animals , Mice , Diet, High-Fat/adverse effects , Lipopolysaccharides/metabolism , Proteomics , Hydrogen Peroxide/metabolism , Mice, Obese , Mice, Inbred C57BL , Mitochondria/metabolism , Obesity/metabolism , Adenosine Triphosphate/metabolism
2.
FASEB J ; 36(6): e22354, 2022 06.
Article in English | MEDLINE | ID: mdl-35616343

ABSTRACT

Resolvin E1 (RvE1), a specialized pro-resolving mediator (SPM), improves glucose homeostasis in inbred mouse models of obesity. However, an impediment toward translation is that obesity is a highly heterogenous disease in which individuals will respond very differently to interventions such as RvE1. Thus, there is a need to study SPMs in the context of modeling the heterogeneity of obesity that is observed in humans. We investigated how RvE1 controls the concentration of key circulating metabolic biomarkers using diversity outbred (DO) mice, which mimic human heterogeneity. We first demonstrate that weights of DO mice can be classified into distinct distributions of fat mass (i.e., modeling differing classes of obesity) in response to a high-fat diet and in the human population when examining body composition. Next, we show RvE1 administration based on body weight for four consecutive days after giving mice a high-fat diet led to approximately half of the mice responding positively for serum total gastric inhibitory polypeptide (GIP), glucagon, insulin, glucose, leptin, and resistin. Interestingly, RvE1 improved hyperleptinemia most effectively in the lowest class of fat mass despite adjusting the dose of RvE1 with increasing adiposity. Furthermore, leptin levels after RvE1 treatment were the lowest in those mice that were also RvE1 positive responders for insulin and resistin. Collectively, these results suggest a therapeutic fat mass-dependent window for RvE1, which should be considered in future clinical trials. Moreover, the data underscore the importance of studying SPMs with heterogenous mice as a step toward precision SPM administration in humans.


Subject(s)
Eicosapentaenoic Acid , Obesity , Animals , Collaborative Cross Mice , Disease Models, Animal , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Glucose , Humans , Insulins , Leptin , Mice , Obesity/drug therapy , Resistin
3.
Article in English | MEDLINE | ID: mdl-34619367

ABSTRACT

Eicosapentaenoic acid (EPA) ethyl esters are of interest given their clinical approval for lowering circulating triglycerides and cardiometabolic disease risk. EPA ethyl esters prevent metabolic complications driven by a high fat diet in male mice; however, their impact on female mice is less studied. Herein, we first investigated how EPA influences the metabolic profile of female C57BL/6J mice consuming a high fat diet. EPA lowered murine fat mass accumulation, potentially through increased biosynthesis of 8-hydroxyeicosapentaenoic acid (HEPE), as revealed by mass spectrometry and cell culture studies. EPA also reversed the effects of a high fat diet on circulating levels of insulin, glucose, and select inflammatory/metabolic markers. Next, we studied if the improved metabolic profile of obese mice consuming EPA was associated with a reduction in the abundance of key gut Gram-negative bacteria that contribute toward impaired glucose metabolism. Using fecal 16S-ribosomal RNA gene sequencing, we found EPA restructured the gut microbiota in a time-dependent manner but did not lower the levels of key Gram-negative bacteria. Interestingly, EPA robustly increased the abundance of the Gram-negative Akkermansia muciniphila, which controls glucose homeostasis. Finally, predictive functional profiling of microbial communities revealed EPA-mediated reversal of high fat diet-associated changes in a wide range of genes related to pathways such as Th-17 cell differentiation and PI3K-Akt signaling. Collectively, these results show that EPA ethyl esters prevent some of the deleterious effects of a high fat diet in female mice, which may be mediated mechanistically through 8-HEPE and the upregulation of intestinal Akkermansia muciniphila.


Subject(s)
Eicosapentaenoic Acid/pharmacology , Gastrointestinal Microbiome/genetics , Hydroxyeicosatetraenoic Acids/biosynthesis , Akkermansia/genetics , Akkermansia/growth & development , Animals , Cardiometabolic Risk Factors , Cell Differentiation/drug effects , Diet, High-Fat/adverse effects , Eicosapentaenoic Acid/metabolism , Female , Gastrointestinal Microbiome/drug effects , Glucose/metabolism , Humans , Hydroxyeicosatetraenoic Acids/blood , Male , Mass Spectrometry , Mice , Mice, Obese/genetics , Mice, Obese/microbiology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Ribosomal, 16S/genetics , Sex Characteristics , Th17 Cells/metabolism , Triglycerides/blood
4.
Toxicol Sci ; 183(1): 170-183, 2021 08 30.
Article in English | MEDLINE | ID: mdl-34175951

ABSTRACT

Ozone (O3) is a criteria air pollutant known to increase the morbidity and mortality of cardiopulmonary diseases. This occurs through a pulmonary inflammatory response characterized by increased recruitment of immune cells into the airspace, pro-inflammatory cytokines, and pro-inflammatory lipid mediators. Recent evidence has demonstrated sex-dependent differences in the O3-induced pulmonary inflammatory response. However, it is unknown if this dimorphic response is evident in pulmonary lipid mediator metabolism. We hypothesized that there are sex-dependent differences in lipid mediator production following acute O3 exposure. Male and female C57BL/6J mice were exposed to 1 part per million O3 for 3 h and were necropsied at 6 or 24 h following exposure. Lung lavage was collected for cell differential and total protein analysis, and lung tissue was collected for mRNA analysis, metabololipidomics, and immunohistochemistry. Compared with males, O3-exposed female mice had increases in airspace neutrophilia, neutrophil chemokine mRNA, pro-inflammatory eicosanoids such as prostaglandin E2, and specialized pro-resolving mediators (SPMs), such as resolvin D5 in lung tissue. Likewise, precursor fatty acids (arachidonic and docosahexaenoic acid; DHA) were increased in female lung tissue following O3 exposure compared with males. Experiments with ovariectomized females revealed that loss of ovarian hormones exacerbates pulmonary inflammation and injury. However, eicosanoid and SPM production were not altered by ovariectomy despite depleted pulmonary DHA concentrations. Taken together, these data indicate that O3 drives an increased pulmonary inflammatory and bioactive lipid mediator response in females. Furthermore, ovariectomy increases susceptibility to O3-induced pulmonary inflammation and injury, as well as decreases pulmonary DHA concentrations.


Subject(s)
Ozone , Sex Characteristics , Animals , Eicosanoids , Female , Lung , Male , Mice , Mice, Inbred C57BL , Ozone/toxicity
5.
Front Nutr ; 8: 799492, 2021.
Article in English | MEDLINE | ID: mdl-35004828

ABSTRACT

Resolvin E1 (RvE1) is an immunoresolvent that is synthesized from eicosapentaenoic acid and can bind the receptor ERV1/ChemR23. We previously showed activation of the RvE1-ChemR23 axis improves hyperglycemia and hyperinsulinemia of obese mice; however, it remains unclear how RvE1 controls glucose homeostasis. Here we investigated hepatic metabolic and inflammatory transcriptional targets of the RvE1-ChemR23 axis using lean and obese wild type (WT) and ChemR23 knockout (KO) mice. We conducted an in-depth transcriptional study by preforming whole gene-level and exon-level analyses, which provide insight into alternative splicing variants and miRNA regulation. Compared to controls, WT and KO obese mice in the absence of RvE1 displayed similar gene-level profiles, which entailed dysregulated pathways related to glucose homeostasis. Notably, obese WT mice relative to lean controls showed a robust decrease in pathways related to the biosynthesis of unsaturated fatty acids. At the exon-level, obese ChemR23 KOs compared to obese WT mice displayed changes in pathways related to hepatic lipid transport, cholesterol metabolism, and immunological functions such as complement cascades and platelet activation. Importantly, upon RvE1 administration to WT obese mice, we discovered upregulated genes in pathways relating to insulin sensitivity and downregulated genes related to regulators of TGF-ß signaling. This transcriptional profile was generally not recapitulated with obese ChemR23 KO mice administered RvE1. Collectively, gene and exon-level analyses suggest RvE1 controls the hepatic transcriptional profile related to glucose homeostasis, insulin sensitivity, and inflammation in a manner that is largely dependent on ChemR23. These studies will drive future mechanistic experiments on the RvE1-ChemR23 axis.

6.
Epigenet Insights ; 13: 2516865720970575, 2020.
Article in English | MEDLINE | ID: mdl-33313480

ABSTRACT

Deficiency of methyl donor nutrients folate, choline, and methionine (methyl deficiency) during gestation can impair fetal development and perturb DNA methylation. Here, we assessed genetic susceptibility to methyl deficiency by comparing effects in wildtype C57BL/6J (B6) mice to mutant mice carrying a 1.3 kb deletion at the H19/Igf2 Imprinting Control Region (ICR) (H19 ICRΔ2,3). The H19 ICRΔ2,3 mutation mimics microdeletions observed in Beckwith-Wiedemann syndrome (BWS) patients, who exhibit epimutations in cis that cause loss of imprinting and fetal overgrowth. Dams were treated during pregnancy with 1 of 4 methyl sufficient (MS) or methyl deficient (MD) diets, with or without the antibiotic commonly used to deplete folate producing gut microbes. As expected, after ~9 weeks of treatment, dams in MD and MD + antibiotic groups exhibited substantially reduced plasma folate concentrations. H19 ICRΔ2,3 mutant lines were more susceptible to adverse pregnancy outcomes caused by methyl deficiency (reduced birth rate and increased pup lethality) and antibiotic (decreased litter size and litter survival). Surprisingly, pup growth/development was only minimally affected by methyl deficiency, while antibiotic treatment caused inverse effects on B6 and H19 ICRΔ2,3 lines. B6 pups treated with antibiotic exhibited increased neonatal and weanling bodyweight, while both wildtype and mutant pups of heterozygous H19 ICRΔ2,3/+ dams exhibited decreased neonatal bodyweight that persisted into adulthood. Interestingly, only antibiotic-treated pups carrying the H19 ICRΔ2,3 mutation exhibited altered DNA methylation at the H19/Igf2 ICR, suggesting ICR epimutation was not sufficient to explain the altered phenotypes. These findings demonstrate that genetic mutation of the H19/Igf2 ICR increases offspring susceptibility to developmental perturbation in the methyl deficiency model, maternal and pup genotype play an essential role, and antibiotic treatment in the model also plays a key independent role.

7.
Nutrients ; 12(12)2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33276463

ABSTRACT

Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are n-3 polyunsaturated fatty acids (PUFAs) consumed in low abundance in the Western diet. Increased consumption of n-3 PUFAs may have beneficial effects for a wide range of physiological outcomes including chronic inflammation. However, considerable mechanistic gaps in knowledge exist about EPA versus DHA, which are often studied as a mixture. We suggest the novel hypothesis that EPA and DHA may compete against each other through overlapping mechanisms. First, EPA and DHA may compete for residency in membrane phospholipids and thereby differentially displace n-6 PUFAs, which are highly prevalent in the Western diet. This would influence biosynthesis of downstream metabolites of inflammation initiation and resolution. Second, EPA and DHA exert different effects on plasma membrane biophysical structure, creating an additional layer of competition between the fatty acids in controlling signaling. Third, DHA regulates membrane EPA levels by lowering its rate of conversion to EPA's elongation product n-3 docosapentaenoic acid. Collectively, we propose the critical need to investigate molecular competition between EPA and DHA in health and disease, which would ultimately impact dietary recommendations and precision nutrition trials.


Subject(s)
Docosahexaenoic Acids/metabolism , Eicosapentaenoic Acid/metabolism , Animals , Arachidonic Acid , Diet , Diet, Western , Dietary Supplements , Fatty Acids , Fatty Acids, Omega-3 , Fatty Acids, Omega-6 , Fatty Acids, Unsaturated , Humans , Inflammation , Phospholipids
8.
Front Immunol ; 11: 1997, 2020.
Article in English | MEDLINE | ID: mdl-32983141

ABSTRACT

Obesity is a major independent risk factor for increased morbidity and mortality upon infection with Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2), which is responsible for the current coronavirus disease pandemic (COVID-19). Therefore, there is a critical need to identify underlying metabolic factors associated with obesity that could be contributing toward increased susceptibility to SARS-CoV-2 in this vulnerable population. Here, we focus on the critical role of potent endogenous lipid metabolites known as specialized pro-resolving mediators (SPMs) that are synthesized from polyunsaturated fatty acids. SPMs are generated during the transition of inflammation to resolution and have a vital role in directing damaged tissues to homeostasis; furthermore, SPMs display anti-viral activity in the context of influenza infection without being immunosuppressive. We cover evidence from rodent and human studies to show that obesity, and its co-morbidities, induce a signature of SPM deficiency across immunometabolic tissues. We further discuss how the effects of obesity upon SARS-CoV-2 infection are likely exacerbated with environmental exposures that promote chronic pulmonary inflammation and augment SPM deficits. Finally, we highlight potential approaches to overcome the loss of SPMs using dietary and pharmacological interventions. Collectively, this mini-review underscores the need for mechanistic studies on how SPM deficiencies driven by obesity and environmental exposures may exacerbate the response to SARS-CoV-2.


Subject(s)
Betacoronavirus , Coronavirus Infections/epidemiology , Docosahexaenoic Acids/deficiency , Eicosapentaenoic Acid/metabolism , Linoleic Acid/deficiency , Lipoxins/deficiency , Obesity/epidemiology , Obesity/immunology , Pneumonia, Viral/epidemiology , COVID-19 , Comorbidity , Coronavirus Infections/diet therapy , Coronavirus Infections/virology , Disease Susceptibility , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/therapeutic use , Humans , Inflammation/metabolism , Linoleic Acid/therapeutic use , Lipoxins/therapeutic use , Morbidity , Obesity/metabolism , Pandemics , Pneumonia, Viral/diet therapy , Pneumonia, Viral/virology , Risk Factors , SARS-CoV-2
9.
FASEB J ; 34(8): 10640-10656, 2020 08.
Article in English | MEDLINE | ID: mdl-32579292

ABSTRACT

Eicosapentaenoic acid (EPA) has garnered attention after the success of the REDUCE-IT trial, which contradicted previous conclusions on EPA for cardiovascular disease risk. Here we first investigated EPA's preventative role on hyperglycemia and hyperinsulinemia. EPA ethyl esters prevented obesity-induced glucose intolerance, hyperinsulinemia, and hyperglycemia in C57BL/6J mice. Supporting NHANES analyses showed that fasting glucose levels of obese adults were inversely related to EPA intake. We next investigated how EPA improved murine hyperinsulinemia and hyperglycemia. EPA overturned the obesity-driven decrement in the concentration of 18-hydroxyeicosapentaenoic acid (18-HEPE) in white adipose tissue and liver. Treatment of obese inbred mice with RvE1, the downstream immunoresolvant metabolite of 18-HEPE, but not 18-HEPE itself, reversed hyperinsulinemia and hyperglycemia through the G-protein coupled receptor ERV1/ChemR23. To translate the findings, we determined if the effects of RvE1 were dependent on host genetics. RvE1's effects on hyperinsulinemia and hyperglycemia were divergent in diversity outbred mice that model human genetic variation. Secondary SNP analyses further confirmed extensive genetic variation in human RvE1/EPA-metabolizing genes. Collectively, the data suggest EPA prevents hyperinsulinemia and hyperglycemia, in part, through RvE1's activation of ERV1/ChemR23 in a host genetic manner. The studies underscore the need for personalized administration of RvE1 based on genetic/metabolic enzyme profiles.


Subject(s)
Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Hyperglycemia/genetics , Hyperglycemia/prevention & control , Hyperinsulinism/genetics , Hyperinsulinism/prevention & control , Adipose Tissue, White/drug effects , Animals , Glucose Intolerance/genetics , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Chemokine/genetics , Receptors, G-Protein-Coupled/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...