Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters

Complementary Medicines
Database
Language
Affiliation country
Publication year range
1.
Mol Cell Neurosci ; 80: 52-57, 2017 04.
Article in English | MEDLINE | ID: mdl-28214555

ABSTRACT

Hypothalamic inflammation contributes to metabolic dysregulation and the onset of obesity. Dietary saturated fats activate microglia via a nuclear factor-kappa B (NFκB) mediated pathway to release pro-inflammatory cytokines resulting in dysfunction or death of surrounding neurons. Fatty acid binding proteins (FABPs) are lipid chaperones regulating metabolic and inflammatory pathways in response to fatty acids. Loss of FABP4 in peripheral macrophages via either molecular or pharmacologic mechanisms results in reduced obesity-induced inflammation via a UCP2-redox based mechanism. Despite the widespread appreciation for the role of FABP4 in mediating peripheral inflammation, the expression of FABP4 and a potential FABP4-UCP2 axis regulating microglial inflammatory capacity is largely uncharacterized. To that end, we hypothesized that microglial cells express FABP4 and that inhibition would upregulate UCP2 and attenuate palmitic acid (PA)-induced pro-inflammatory response. Gene expression confirmed expression of FABP4 in brain tissue lysate from C57Bl/6J mice and BV2 microglia. Treatment of microglial cells with an FABP inhibitor (HTS01037) increased expression of Ucp2 and arginase in the presence or absence of PA. Moreover, cells exposed to HTS01037 exhibited attenuated expression of inducible nitric oxide synthase (iNOS) compared to PA alone indicating reduced NFκB signaling. Hypothalamic tissue from mice lacking FABP4 exhibit increased UCP2 expression and reduced iNOS, tumor necrosis factor-alpha (TNF-α), and ionized calcium-binding adapter molecule 1 (Iba1; microglial activation marker) expression compared to wild type mice. Further, this effect is negated in microglia lacking UCP2, indicating the FABP4-UCP2 axis is pivotal in obesity induced neuroinflammation. To our knowledge, this is the first report demonstrating a FABP4-UCP2 axis with the potential to modulate the microglial inflammatory response.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Microglia/metabolism , Uncoupling Protein 2/genetics , Animals , Anti-Inflammatory Agents/pharmacology , Arginase/metabolism , Brain/cytology , Calcium-Binding Proteins/metabolism , Cell Line, Transformed , Fatty Acid-Binding Proteins/antagonists & inhibitors , Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Hypothalamus/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/drug effects , Nitric Oxide Synthase Type II/metabolism , Palmitic Acid/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/metabolism , Uncoupling Protein 2/metabolism
2.
Mol Cell Neurosci ; 75: 93-100, 2016 09.
Article in English | MEDLINE | ID: mdl-27449757

ABSTRACT

Palmitic acid (PA), an abundant dietary saturated fatty acid, contributes to obesity and hypothalamic dysregulation in part through increase in oxidative stress, insulin resistance, and neuroinflammation. Increased production of reactive oxygen species (ROS) as a result of PA exposure contributes to the onset of neuronal apoptosis. Additionally, high fat diets lead to changes in hypothalamic gene expression profiles including suppression of the anti-apoptotic protein B cell lymphoma 2 (Bcl-2) and upregulation of the pro-apoptotic protein B cell lymphoma 2 associated X protein (Bax). Orexin A (OXA), a hypothalamic peptide important in obesity resistance, also contributes to neuroprotection. Prior studies have demonstrated that OXA attenuates oxidative stress induced cell death. We hypothesized that OXA would be neuroprotective against PA induced cell death. To test this, we treated an immortalized hypothalamic cell line (designated mHypoA-1/2) with OXA and PA. We demonstrate that OXA attenuates PA-induced hypothalamic cell death via reduced caspase-3/7 apoptosis, stabilization of Bcl-2 gene expression, and reduced Bax/Bcl-2 gene expression ratio. We also found that OXA inhibits ROS production after PA exposure. Finally, we show that PA exposure in mHypoA-1/2 cells significantly reduces basal respiration, maximum respiration, ATP production, and reserve capacity. However, OXA treatment reverses PA-induced changes in intracellular metabolism, increasing basal respiration, maximum respiration, ATP production, and reserve capacity. Collectively, these results support that OXA protects against PA-induced hypothalamic dysregulation, and may represent one mechanism through which OXA can ameliorate effects of obesogenic diet on brain health.


Subject(s)
Apoptosis , Neurons/drug effects , Neuroprotective Agents/pharmacology , Orexins/pharmacology , Palmitic Acid/toxicity , Adenosine Triphosphate/metabolism , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Cell Respiration , Hypothalamus/cytology , Mice , Neurons/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/metabolism
3.
J Cardiovasc Transl Res ; 9(4): 368-73, 2016 08.
Article in English | MEDLINE | ID: mdl-27184805

ABSTRACT

There is conflicting clinical evidence whether administration of coenzyme Q10 (CoQ10) improves function following coronary artery bypass graft surgery (CABG). Using a swine model of hibernating myocardium, we tested whether daily CoQ10 would improve contractile function by MRI at 4-week post-CABG. Twelve pigs underwent a thoracotomy and had a constrictor placed on the left anterior descending (LAD). At 12 weeks, they underwent off-pump bypass and received daily dietary supplements of either CoQ10 (10 mg/kg/day) or placebo. At 4-week post-CABG, circumferential strain measurements in the hibernating LAD region from placebo and CoQ10 groups were not different and increased to a similar extent with dobutamine (-14.7 ± 0.6 versus -14.8 ± 0.1, respectively (NS)). Post-sacrifice, oxidant stress markers were obtained in the mitochondrial isolates and protein carbonyl in the placebo, and CoQ10 groups were 6.14 ± 0.36 and 5.05 ± 0.32 nmol/mg, respectively (NS). In summary, CoQ10 did not improve contractile reserve or reduce oxidant stress at 4-week post-CABG.


Subject(s)
Cardiotonic Agents/pharmacology , Coronary Artery Bypass, Off-Pump , Coronary Artery Disease/drug therapy , Coronary Artery Disease/surgery , Myocardial Contraction/drug effects , Myocardial Stunning/drug therapy , Myocardial Stunning/surgery , Ubiquinone/analogs & derivatives , Animals , Biomarkers/metabolism , Biomechanical Phenomena , Coronary Artery Disease/metabolism , Coronary Artery Disease/physiopathology , Disease Models, Animal , Female , Magnetic Resonance Imaging , Mitochondria, Heart/metabolism , Myocardial Stunning/metabolism , Myocardial Stunning/physiopathology , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress , Protein Carbonylation , Recovery of Function , Stress, Mechanical , Sus scrofa , Time Factors , Ubiquinone/pharmacology
4.
Neurosci Lett ; 606: 140-4, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26306651

ABSTRACT

Excess dietary saturated fatty acids such as palmitic acid (PA) induce peripheral and hypothalamic inflammation. Hypothalamic inflammation, mediated in part by microglial activation, contributes to metabolic dysregulation. In rodents, high fat diet-induced microglial activation results in nuclear translocation of nuclear factor-kappa B (NFκB), and increased central pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6). The hypothalamic neuropeptide orexin A (OXA, hypocretin 1) is neuroprotective in brain. In cortex, OXA can also reduce inflammation and neurodegeneration through a microglial-mediated pathway. Whether hypothalamic orexin neuroprotection mechanisms depend upon microglia is unknown. To address this issue, we evaluated effects of OXA and PA on inflammatory response in immortalized murine microglial and hypothalamic neuronal cell lines. We demonstrate for the first time in microglial cells that exposure to PA increases gene expression of orexin-1 receptor but not orexin-2 receptor. Pro-inflammatory markers IL-6, TNF-α, and inducible nitric oxide synthase in microglial cells are increased following PA exposure, but are reduced by pretreatment with OXA. The anti-inflammatory marker arginase-1 is increased by OXA. Finally, we show hypothalamic neurons exposed to conditioned media from PA-challenged microglia have increased cell survival only when microglia were pretreated with OXA. These data support the concept that OXA may act as an immunomodulatory regulator of microglia, reducing pro-inflammatory cytokines and increasing anti-inflammatory factors to promote a favorable neuronal microenvironment.


Subject(s)
Hypothalamus/drug effects , Microglia/drug effects , Neurons/drug effects , Orexins/metabolism , Palmitic Acid/pharmacology , Animals , Arginase/metabolism , Cells, Cultured , Culture Media, Conditioned , Cytokines/metabolism , Dietary Fats , Hypothalamus/cytology , Hypothalamus/metabolism , Interleukin-6/metabolism , Mice , Microglia/metabolism , Neurons/cytology , Nitric Oxide Synthase Type II/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
5.
Ageing Res Rev ; 20: 63-73, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25462194

ABSTRACT

The hypothalamic neuropeptides orexin A and B (hypocretin 1 and 2) are important homeostatic mediators of central control of energy metabolism and maintenance of sleep/wake states. Dysregulation or loss of orexin signaling has been linked to narcolepsy, obesity, and age-related disorders. In this review, we present an overview of our current understanding of orexin function, focusing on sleep disorders, energy balance, and aging, in both rodents and humans. We first discuss animal models used in studies of obesity and sleep, including loss of function using transgenic or viral-mediated approaches, gain of function models using exogenous delivery of orexin receptor agonist, and naturally-occurring models in which orexin responsiveness varies by individual. We next explore rodent models of orexin in aging, presenting evidence that orexin loss contributes to age-related changes in sleep and energy balance. In the next section, we focus on clinical importance of orexin in human obesity, sleep, and aging. We include discussion of orexin loss in narcolepsy and potential importance of orexin in insomnia, correlations between animal and human studies of age-related decline, and evidence for orexin involvement in age-related changes in cognitive performance. Finally, we present a summary of recent studies of orexin in neurodegenerative disease. We conclude that orexin acts as an integrative homeostatic signal influencing numerous brain regions, and that this pivotal role results in potential dysregulation of multiple physiological processes when orexin signaling is disrupted or lost.


Subject(s)
Aging/physiology , Intracellular Signaling Peptides and Proteins , Neuropeptides , Obesity , Sleep Wake Disorders , Animals , Disease Models, Animal , Energy Metabolism , Homeostasis/physiology , Humans , Hypothalamus/metabolism , Hypothalamus/physiopathology , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/metabolism , Neuropeptides/pharmacology , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/pharmacology , Obesity/drug therapy , Obesity/metabolism , Orexins , Signal Transduction , Sleep/drug effects , Sleep/physiology , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/metabolism
6.
J Vis Exp ; (86)2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24797379

ABSTRACT

The ability to multiplex assays in studies of complex cellular mechanisms eliminates the need for repetitive experiments, provides internal controls, and decreases waste in costs and reagents. Here we describe optimization of a multiplex assay to assess apoptosis following a palmitic acid (PA) challenge in an in vitro hypothalamic model, using both fluorescent and luminescent based assays to measure viable cell counts and caspase-3/7 activity in a 96-well microtiter plate format. Following PA challenge, viable cells were determined by a resazurin-based fluorescent assay. Caspase-3/7 activity was then determined using a luminogenic substrate, DEVD, and normalized to cell number. This multiplexing assay is a useful technique for determining change in caspase activity following an apoptotic stimulus, such as saturated fatty acid challenge. The saturated fatty acid PA can increase hypothalamic oxidative stress and apoptosis, indicating the potential importance of assays such as that described here in studying the relationship between saturated fatty acids and neuronal function.


Subject(s)
Caspase 3/metabolism , Caspase 7/metabolism , Hypothalamus/cytology , Hypothalamus/enzymology , Palmitic Acid/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/analysis , Caspase 7/analysis , Cell Line , Hypothalamus/drug effects , Mice
7.
Neurosci Lett ; 524(1): 30-4, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22796468

ABSTRACT

Current data support the idea that hypothalamic neuropeptide orexin A (OxA; hypocretin 1) mediates resistance to high fat diet-induced obesity. We previously demonstrated that OxA elevates spontaneous physical activity (SPA), that rodents with high SPA have higher endogenous orexin sensitivity, and that OxA-induced SPA contributes to obesity resistance in rodents. Recent reports show that OxA can confer neuroprotection against ischemic damage, and may decrease lipid peroxidation. This is noteworthy as independent lines of evidence indicate that diets high in saturated fats can decrease SPA, increase hypothalamic apoptosis, and lead to obesity. Together data suggest OxA may protect against obesity both by inducing SPA and by modulation of anti-apoptotic mechanisms. While OxA effects on SPA are well characterized, little is known about the short- and long-term effects of hypothalamic OxA signaling on intracellular neuronal metabolic status, or the physiological relevance of such signaling to SPA. To address this issue, we evaluated the neuroprotective effects of OxA in a novel immortalized primary embryonic rat hypothalamic cell line. We demonstrate for the first time that OxA increases cell viability during hydrogen peroxide challenge, decreases hydrogen peroxide-induced lipid peroxidative stress, and decreases caspase 3/7 induced apoptosis in an in vitro hypothalamic model. Our data support the hypothesis that OxA may promote obesity resistance both by increasing SPA, and by influencing survival of OxA-responsive hypothalamic neurons. Further identification of the individual mediators of the anti-apoptotic and peroxidative effects of OxA on target neurons could lead to therapies designed to maintain elevated SPA and increase obesity resistance.


Subject(s)
Apoptosis , Hypothalamus/cytology , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Lipid Peroxidation , Neuropeptides/physiology , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Cell Survival , Embryo, Mammalian , Hydrogen Peroxide/pharmacology , Hypothalamus/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Neurons/cytology , Neuropeptides/pharmacology , Orexin Receptors , Orexins , Oxidative Stress , RNA, Messenger/metabolism , Rats , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL