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.
Biol Reprod ; 95(6): 120, 2016 12.
Article in English | MEDLINE | ID: mdl-27733380

ABSTRACT

There is considerable evidence that implicates oxidative stress in the pathophysiology of human pregnancy complications. However, the role and the mechanism of maintaining an antioxidant prosurvival uterine environment during normal pregnancy is largely unresolved. Herein we report that the highly active uterine unfolded protein response plays a key role in promoting antioxidant activity in the uterine myocyte across gestation. The unfolded protein response (UPR) senses the accumulation of misfolded proteins in the endoplasmic reticulum (ER) and activates a signaling network that consists of the transmembrane protein kinase eukaryotic translation initiation factor 2 alpha kinase 3/PKR-like-ER kinase (EIF2AK3), which acts to decrease protein translation levels, allowing for a lowered need for protein folding during periods of ER stress. However, independent of its translational regulatory capacity, EIF2AK3-dependent signals elicit the activation of the transcription factor, nuclear factor erythroid 2-like 2 (NFE2L2) in response to oxidative stress. NFE2L2 binds to antioxidant response elements in the promoters of a variety of antioxidant genes that minimize the opportunities for generation of reactive oxygen intermediates. Our analysis demonstrates that in the absence of EIF2AK3, the uterine myocyte experiences increased levels of reactive oxygen species due to decreased NFE2L2 activation. Elevated levels of intracellular reactive oxygen species were observed in the EIF2AK3 null cells, and this was associated with the onset of apoptotic cell death. These findings confirm the prosurvival and antioxidant role of UPR-mediated EIF2AK3 activation in the context of the human uterine myocyte.


Subject(s)
Endometrium/metabolism , Muscle Cells/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Unfolded Protein Response/physiology , Uterus/metabolism , Animals , Apoptosis/physiology , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/physiology , Female , Humans , Mice , NF-E2-Related Factor 2/metabolism , Pregnancy , Protein Folding , eIF-2 Kinase/metabolism
2.
Brain Res ; 1537: 191-200, 2013 Nov 06.
Article in English | MEDLINE | ID: mdl-24076449

ABSTRACT

Insulin injections into the central nucleus of the amygdala (CeA) inhibit food intake but this response is lost quickly on feeding a high fat diet. The purpose of the studies described in this manuscript was to identify the potential mechanism for the development of this insulin resistance. High fat diets (HFD) induced PKCθ activation and blocked the stimulation of Akt but not mTOR phosphorylation in the amygdala in response to CeA insulin injections. Infusions of palmitic acid onto the CeA had identical effects to HFD on PKCθ expression and insulin signaling in the amygdala. CeA insulin also induced an increase in Akt phosphorylation in the hypothalamus but had no effect on hypothalamic mTOR phosphorylation. Feeding HFD but not CeA palmitate infusions reversed the hypothalamic Akt signaling response to CeA insulin. These data, which show the independence of Akt and mTOR signaling responses to insulin in the amygdala and the effect of insulin signaling in the CeA on hypothalamic Akt signaling, suggest that the amygdala might also have a significant role in regulating hypothalamic responses to dietary fat.


Subject(s)
Amygdala/metabolism , Diet, High-Fat , Fatty Acids/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Signal Transduction , Animal Feed , Animals , Dietary Fats/metabolism , Insulin Resistance , Male , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/physiology
3.
Obesity (Silver Spring) ; 21(4): 755-64, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23712979

ABSTRACT

OBJECTIVE: To investigate the signaling mechanisms that might underlie the loss of anorectic response to insulin injections into the central nucleus of the amygdala (CeA) within 3 days of feeding a high fat diet. DESIGN AND METHODS: Protein samples from amygdala and hypothalamus of rats fed high or low fat diets were subjected to a phosphorylation screening assay. The effects of dietary fat intake on the expression and activation of protein kinase C theta (PKCθ) in brain regions was studied. Finally, lentiviral vectors were used to overexpress rat PKCθ unilaterally or bilaterally into the CeA of rats and the effects on food intake, body weight and insulin stimulation of Akt phosphorylation were studied. RESULTS: The level of pMARCKS (Myristoylated alanine-rich C-kinase substrate), a major substrate of PKCθ, was increased 116% in amygdala of high fat diet fed rats but reduced in the hypothalamus. High fat diets increased the level of PKCθ in a region specific manner in the brain and this PKCθ was activated by membrane association. Overexpressing rat PKCθ either unilaterally or bilaterally into the CeA inhibited insulin stimulation of Akt signaling and blocked the anorectic response to insulin injected into the amygdala. Bilaterally injected PKCθ rats gained more weight and body fat and had increased food intake when fed a high fat diet compared to the control rats that received a lentiviral-Green Fluorescent Protein construct. CONCLUSION: The data suggest that insulin may have a physiological role within the amygdala to regulate energy balance.


Subject(s)
Amygdala/metabolism , Body Weight , Energy Intake , Insulin/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Adipose Tissue/metabolism , Amygdala/cytology , Amygdala/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Diet, Fat-Restricted , Diet, High-Fat , Dietary Fats/administration & dosage , Genetic Vectors , Hypothalamus/drug effects , Hypothalamus/metabolism , Immunohistochemistry , Insulin Resistance , Isoenzymes/genetics , Lentivirus/genetics , Male , Phenotype , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C-theta , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
4.
Brain Res ; 1498: 85-94, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23295188

ABSTRACT

The increase in PKCθ expression in the amygdala of rats fed high fat diet (HFD) has been related to the loss of the anorectic response to insulin injections into the central nucleus of the amygdala (CeA) in these animals. PKCθ overexpression in the CeA increases food intake, body weight and body fat and inhibits insulin stimulation of Akt signaling. To study the effects of bilateral overexpression of PKCθ in the CeA of rats on peripheral metabolism, rats were injected into the CeA or 3rd ventricle with a lentiviral (LV)-PKCθ construct or LV-Green fluorescent protein (GFP) construct as a control and fed either LFD or HFD. Insulin and glucose tolerance tests were undertaken and hepatic AMPK activation, Pepck, Srebp1c gene expression and lipid levels assayed. CeA LV-PKCθ injected rats increased food intake, body weight and body fat and increased hepatic, but not serum, triglyceride levels compared to control rats that received a CeA-LV-GFP construct. Hepatic AMP-kinase activity was reduced but expression of Pepck increased while serum insulin decreased, glucose tolerance improved and the hypoglycemic response to insulin was enhanced in CeA LV-PKCθ injected rats. In contrast, rats that received LV-PKCθ injections into the 3rd Ventricle did not show any changes in food intake or body weight although serum, but not hepatic, triglyceride levels were increased and glucose tolerance was impaired. The data suggest that activation of PKCθ in the CeA and hypothalamus have different effects on energy balance and peripheral metabolism and that insulin signaling in the amygdala regulates peripheral metabolism.


Subject(s)
Central Amygdaloid Nucleus/enzymology , Glucose/metabolism , Homeostasis/physiology , Hypothalamus/enzymology , Isoenzymes/metabolism , Protein Kinase C/metabolism , Adenylate Kinase/metabolism , Adipose Tissue/physiology , Animals , Body Weight/physiology , Diet, Fat-Restricted , Diet, High-Fat , Eating/physiology , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Isoenzymes/genetics , Lentivirus/genetics , Liver/metabolism , Male , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Protein Kinase C/genetics , Protein Kinase C-theta , Rats, Sprague-Dawley , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/metabolism
5.
Pflugers Arch ; 464(2): 227-37, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22644615

ABSTRACT

The chemoreception of dietary fat in the oral cavity has largely been attributed to activation of the somatosensory system that conveys the textural properties of fat. However, the ability of fatty acids, which are believed to represent the proximate stimulus for fat taste, to stimulate rat trigeminal neurons has remained unexplored. Here, we found that several free fatty acids are capable of activating trigeminal neurons with different kinetics. Further, a polyunsaturated fatty acid, linoleic acid (LA), activates trigeminal neurons by increasing intracellular calcium concentration and generating depolarizing receptor potentials. Ion substitution and pharmacological approaches reveal that intracellular calcium store depletion is crucial for LA-induced signaling in a subset of trigeminal neurons. Using pseudorabies virus (PrV) as a live cell tracer, we identified a subset of lingual nerve-innervated trigeminal neurons that respond to different subsets of fatty acids. Quantitative real-time PCR of several transient receptor potential channel markers in individual neurons validated that PrV labeled a subset but not the entire population of lingual-innervated trigeminal neurons. We further confirmed that the LA-induced intracellular calcium rise is exclusively coming from the release of calcium stores from the endoplasmic reticulum in this subset of lingual nerve-innervated trigeminal neurons.


Subject(s)
Calcium Signaling , Fatty Acids/pharmacology , Linoleic Acid/pharmacology , Membrane Potentials , Trigeminal Ganglion/physiology , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Herpesvirus 1, Suid , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley , Taste/physiology , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism
6.
Physiol Behav ; 106(2): 158-63, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22336738

ABSTRACT

We have previously shown that voluntary wheel running activity in mice is associated with an increase in the Endoplasmic Reticulum (ER) Unfolded Protein stress response in multiple regions of the brain. Mice that are given access to running wheels show large variations in individual running activity. In contrast, when food is placed on the lid of their cages, rather than within the cage, all mice must undertake significant physical activity in order to gain access to their food. Hence we investigated the effects of food location on food intake and growth of C57BL/6 mice and on the activity of the ER stress system in the brain as reflected in the expression of two marker genes, Xbp1 and Atf6. Mice that had food in cups within their cages and allowed access to running wheels showed the anticipated changes in food intake, body weight and ER stress in the hippocampus compared to mice with no access to running wheels. Locating the food on the lid had no effect on food intake but reduced weight gain significantly. Likewise, locating food on the lid increased the expression of both Xbp1 and Atf6 in the hippocampi in the absence of any running wheel activity. Voluntary wheel running activity was reduced in mice whose food was located on the cage lid and this running actually reduced the expression of the two marker ER stress genes. We conclude that the usual practice of providing food for mice on their cage lids provides a significant level of physical activity that alters the metabolic status and increases ER stress. As such, this may not be the optimal model for the majority of mouse studies that are reported in the literature and it may significantly alter the interpretation of the effect of wheel running activity on ER stress. The differential effects of food location on hippocampal Bdnf gene expression also suggest that BDNF does not directly regulate UPR activity but may be coordinately regulated in response to running activity.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Energy Metabolism/physiology , Feeding Behavior/physiology , Hippocampus/metabolism , Physical Exertion/physiology , Running/physiology , Activating Transcription Factor 6/biosynthesis , Animals , Body Weight , Brain-Derived Neurotrophic Factor/biosynthesis , DNA-Binding Proteins/biosynthesis , Eating , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/biosynthesis , Male , Mice , Mice, Inbred C57BL , Motor Activity , Regulatory Factor X Transcription Factors , Transcription Factors/biosynthesis , X-Box Binding Protein 1 , eIF-2 Kinase/biosynthesis
7.
Diabetes ; 60(1): 97-106, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980461

ABSTRACT

OBJECTIVE: The agouti-related protein (Agrp) is a powerful orexigenic peptide, but little is known about its transcriptional regulation. The objective of this study was to determine molecular mechanisms for the activation of hypothalamic Agrp and identify compounds that stimulate appetite. RESEARCH DESIGN AND METHODS: We used promoter analyses methods, hypothalamic cell culture and transfection, immunohistochemistry, luciferase-expressing transgenic mice, in vivo bioluminescence, anitisense RNA, mouse feeding studies, indirect calorimetry, real-time PCR, and Western blots. RESULTS: We found that the Krüppel-like factor 4 (Klf4) is a potent activator of Agrp by binding to a specific CACCC-box in its minimal promoter. We also found that an extract of tarragon, termed PMI-5011, activated hypothalamic Klf4 and Agrp. In vivo, PMI-5011 increased Agrp promoter activity in luciferase-expressing transgenic mice, increased hypothalamic Klf4 and Agrp expression, increased hypothalamic Orexin and melanin-concentrating hormone, increased food intake, reduced circulating insulin and leptin levels, attenuated energy expenditure, and enhanced body weight but only when using a high-fat diet. CONCLUSIONS: These data show that Klf4 augmented hypothalamic Agrp by binding to a specific CACCC-box onto its minimal promoter. In addition, the tarragon extract PMI-5011 activated Klf4 and orexigenic neuropeptides and reduced peripheral insulin and leptin levels leading to positive energy balance.


Subject(s)
Agouti-Related Protein/genetics , Appetite/physiology , Animal Feed , Animals , Body Composition , Calorimetry, Indirect/methods , DNA Primers , Energy Intake , Energy Metabolism , Feeding Behavior , Insulin/blood , Kruppel-Like Factor 4 , Leptin/blood , Luciferases/genetics , Mice , Mice, Transgenic , Polymerase Chain Reaction/methods , Promoter Regions, Genetic , RNA/genetics , RNA/isolation & purification , Transfection
8.
Brain Res ; 1349: 48-55, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20599830

ABSTRACT

Studies have demonstrated defects of DA and 5HT neurotransmission in dietary fat induced obese animals. In the present study, we used a perfusion system to assay the release of DA and 5HT from striatal slices preloaded with [(3)H]-DA or [(3)H]-5HT. The release of both DA and 5HT from striatal slices of rats fed a high fat diet for 10 days, but not 3 days, was reduced when compared to striatal slices taken from rats fed a low fat diet. Enterostatin, an endogenous pentapeptide inhibits dietary fat intake when administered peripherally and centrally in animals. The central mechanism for the action of enterostatin is not yet determined even though several mechanisms have been suggested. We have shown that enterostatin enhanced [(3)H]-DA release, but not [(3)H]-5HT release from striatal slices of rats that had been adapted to high fat diet for 10 days. The enterostatin-induced increase in [(3)H]-DA release was blocked by nomifensine. Enterostatin did not alter [(3)H]-DA or [(3)H]-5HT release from striatal slices of rats adapted to high fat or low fat diet feeding for 3 days. These findings suggest that enterostatin may inhibit dietary fat intake by blocking dopamine reuptake transport to increase central striatal DA release from rats that have acquired diminished dopamine signal after an adaptive period of fat consumption.


Subject(s)
Colipases/pharmacology , Corpus Striatum/drug effects , Dietary Fats/pharmacology , Dopamine/metabolism , Enzyme Precursors/pharmacology , Serotonin/metabolism , Analysis of Variance , Animals , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley , Tritium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...