Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Int J Mol Sci ; 20(4)2019 Feb 22.
Article in English | MEDLINE | ID: mdl-30813227

ABSTRACT

The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor highly expressed in hepatocytes. Researchers have employed global and liver-specific conditional Ahr knockout mouse models to characterize the physiological roles of the AHR; however, the gestational timing of AHR loss in these models can complicate efforts to distinguish the direct and indirect effects of post-gestational AHR deficiency. Utilizing a novel tamoxifen-inducible AHR knockout mouse model, we analyzed the effects of hepatocyte-targeted AHR loss in adult mice. The data demonstrate that AHR deficiency significantly reduces weight gain and adiposity, and increases multilocular lipid droplet formation within perigonadal white adipose tissue (gWAT). Protein and mRNA expression of fibroblast growth factor 21 (FGF21), an important hepatokine that activates thermogenesis in brown adipose tissue (BAT) and gWAT, significantly increases upon AHR loss and correlates with a significant increase of BAT and gWAT respiratory capacity. Confirming the role of FGF21 in mediating these effects, this phenotype is reversed in mice concomitantly lacking AHR and FGF21 expression. Chromatin immunoprecipitation analyses suggest that the AHR may constitutively suppress Fgf21 transcription through binding to a newly identified xenobiotic response element within the Fgf21 promoter. The data demonstrate an important AHR-FGF21 regulatory axis that influences adipose biology and may represent a "druggable" therapeutic target for obesity and its related metabolic disorders.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Cell Respiration , Fibroblast Growth Factors/metabolism , Gonads/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Thermogenesis , Adipose Tissue, White/drug effects , Adiposity/drug effects , Animals , Body Weight/drug effects , Cell Respiration/drug effects , Diet, High-Fat , Drinking , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Female , Fibroblast Growth Factors/genetics , Gonads/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Droplets/metabolism , Liver/drug effects , Liver/metabolism , Mice , Mice, Knockout , Phenotype , Physical Conditioning, Animal , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Tamoxifen/pharmacology , Thermogenesis/drug effects , Transcription, Genetic/drug effects
2.
Addict Biol ; 23(1): 55-68, 2018 01.
Article in English | MEDLINE | ID: mdl-27862692

ABSTRACT

Cocaine use disorder is a chronic relapsing condition characterized by compulsive drug seeking and taking even after prolonged abstinence periods. Subsequent exposure to drug-associated cues can promote intense craving and lead to relapse in abstinent humans and rodent models. The responsiveness to these cocaine-related cues, or 'cue reactivity', can trigger relapse and cocaine-seeking behaviors; cue reactivity is measurable in cocaine-dependent humans as well as rodent models. Cue reactivity is thought to be predictive of cocaine craving and relapse. Here we report that PPARγ agonism during abstinence from cocaine self-administration reduced previously active lever pressing in Sprague Dawley rats during cue-reactivity tests, while administration of the PPARγ antagonist, GW9662, reversed this effect. PPARγ agonism also normalized nuclear ERK activity in the medial prefrontal cortex and hippocampus which was reversed with GW9662. Our results support the utility of PPARγ agonism as a relapse prevention strategy to maintain abstinence in the presence of cocaine-associated cues.


Subject(s)
Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Drug-Seeking Behavior/drug effects , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Pioglitazone/pharmacology , Anilides/pharmacology , Animals , Behavior, Animal/drug effects , Cocaine-Related Disorders , Craving/drug effects , Cues , Locomotion/drug effects , MAP Kinase Signaling System , Rats , Rats, Sprague-Dawley , Recurrence , Self Administration
3.
J Neurosci ; 34(11): 4054-63, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24623782

ABSTRACT

Cognitive impairment is a quintessential feature of Alzheimer's disease (AD) and AD mouse models. The peroxisome proliferator-activated receptor-γ (PPARγ) agonist rosiglitazone improves hippocampus-dependent cognitive deficits in some AD patients and ameliorates deficits in the Tg2576 mouse model for AD amyloidosis. Tg2576 cognitive enhancement occurs through the induction of a gene and protein expression profile reflecting convergence of the PPARγ signaling axis and the extracellular signal-regulated protein kinase (ERK) cascade, a critical mediator of memory consolidation. We therefore tested whether PPARγ and ERK associated in protein complexes that subserve cognitive enhancement through PPARγ agonism. Coimmunoprecipitation of hippocampal extracts revealed that PPARγ and activated, phosphorylated ERK (pERK) associated in Tg2576 in vivo, and that PPARγ agonism facilitated recruitment of PPARγ to pERK during memory consolidation. Furthermore, the amount of PPARγ recruited to pERK correlated with the cognitive reserve in humans with AD and in Tg2576. Our findings implicate a previously unidentified PPARγ-pERK complex that provides a molecular mechanism for the convergence of these pathways during cognitive enhancement, thereby offering new targets for therapeutic development in AD.


Subject(s)
Alzheimer Disease/metabolism , Cognition/physiology , MAP Kinase Signaling System/physiology , Memory/physiology , PPAR gamma/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Anilides/pharmacology , Animals , Cognition/drug effects , Female , Hippocampus/physiology , Humans , Injections, Intraventricular , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Middle Aged , Nootropic Agents/pharmacology , PPAR gamma/antagonists & inhibitors , Phosphorylation/physiology , Rosiglitazone , Thiazolidinediones/pharmacology
4.
J Neurosci ; 34(3): 1028-36, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24431460

ABSTRACT

Hippocampal network hyperexcitability is considered an early indicator of Alzheimer's disease (AD) memory impairment. Some AD mouse models exhibit similar network phenotypes. In this study we focused on dentate gyrus (DG) granule cell spontaneous and evoked properties in 9-month-old Tg2576 mice that model AD amyloidosis and cognitive deficits. Using whole-cell patch-clamp recordings, we found that Tg2576 DG granule cells exhibited spontaneous EPSCs that were higher in frequency but not amplitude compared with wild-type mice, suggesting hyperactivity of DG granule cells via a presynaptic mechanism. Further support of a presynaptic mechanism was revealed by increased I-O relationships and probability of release in Tg2576 DG granule cells. Since we and others have shown that activation of the peroxisome proliferator-activated receptor gamma (PPARγ) axis improves hippocampal cognition in mouse models for AD as well as benefitting memory performance in some humans with early AD, we investigated how PPARγ agonism affected synaptic activity in Tg2576 DG. We found that PPARγ agonism normalized the I-O relationship of evoked EPSCs, frequency of spontaneous EPSCs, and probability of release that, in turn, correlated with selective expression of DG proteins essential for presynaptic SNARE function that are altered in patients with AD. These findings provide evidence that DG principal cells may contribute to early AD hippocampal network hyperexcitability via a presynaptic mechanism, and that hippocampal cognitive enhancement via PPARγ activation occurs through regulation of presynaptic vesicular proteins critical for proper glutamatergic neurotransmitter release, synaptic transmission, and short-term plasticity.


Subject(s)
Dentate Gyrus/physiology , Nootropic Agents/pharmacology , PPAR gamma/agonists , PPAR gamma/physiology , Presynaptic Terminals/physiology , Thiazolidinediones/pharmacology , Amyloid beta-Protein Precursor/genetics , Animals , Dentate Gyrus/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Presynaptic Terminals/drug effects , Protein Interaction Maps/drug effects , Protein Interaction Maps/physiology , Protein Transport/drug effects , Protein Transport/physiology , Rosiglitazone
5.
J Neurophysiol ; 113(6): 1712-26, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25540218

ABSTRACT

Early cognitive impairment in Alzheimer's disease (AD) correlates with medial temporal lobe dysfunction, including two areas essential for memory formation: the entorhinal cortex and dentate gyrus (DG). In the Tg2576 animal model for AD amyloidosis, activation of the peroxisome proliferator-activated receptor-gamma (PPARγ) with rosiglitazone (RSG) ameliorates hippocampus-dependent cognitive impairment and restores aberrant synaptic activity at the entorhinal cortex to DG granule neuron inputs. It is unknown, however, whether intrinsic firing properties of DG granule neurons in these animals are affected by amyloid-ß pathology and if they are sensitive to RSG treatment. Here, we report that granule neurons from 9-mo-old wild-type and Tg2576 animals can be segregated into two cell types with distinct firing properties and input resistance that correlate with less mature type I and more mature type II neurons. The DG type I cell population was greater than type II in wild-type littermates. In the Tg2576 animals, the type I and type II cell populations were nearly equal but could be restored to wild-type levels through cognitive enhancement with RSG. Furthermore, Tg2576 cell firing frequency and spike after depolarization were decreased in type I and increased in type II cells, both of which could also be restored to wild-type levels upon RSG treatment. That these parameters were restored by PPARγ activation emphasizes the therapeutic value of RSG against early AD cognitive impairment.


Subject(s)
Action Potentials , Alzheimer Disease/metabolism , Neurons/physiology , PPAR gamma/metabolism , Alzheimer Disease/physiopathology , Animals , Cognition , Entorhinal Cortex/cytology , Entorhinal Cortex/drug effects , Entorhinal Cortex/physiopathology , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , PPAR gamma/agonists , Rosiglitazone , Thiazolidinediones/pharmacology
6.
Mol Cell Proteomics ; 12(12): 3640-52, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24008390

ABSTRACT

Chronic hepatic disease damages the liver, and the resulting wound-healing process leads to liver fibrosis and the subsequent development of cirrhosis. The leading cause of hepatic fibrosis and cirrhosis is infection with hepatitis C virus (HCV), and of the patients with HCV-induced cirrhosis, 2% to 5% develop hepatocellular carcinoma (HCC), with a survival rate of 7%. HCC is one of the leading causes of cancer-related death worldwide, and the poor survival rate is largely due to late-stage diagnosis, which makes successful intervention difficult, if not impossible. The lack of sensitive and specific diagnostic tools and the urgent need for early-stage diagnosis prompted us to discover new candidate biomarkers for HCV and HCC. We used aptamer-based fractionation technology to reduce serum complexity, differentially labeled samples (six HCV and six HCC) with fluorescent dyes, and resolved proteins in pairwise two-dimensional difference gel electrophoresis. DeCyder software was used to identify differentially expressed proteins and spots picked, and MALDI-MS/MS was used to determine that ApoA1 was down-regulated by 22% (p < 0.004) in HCC relative to HCV. Differential expression quantified via two-dimensional difference gel electrophoresis was confirmed by means of (18)O/(16)O stable isotope differential labeling with LC-MS/MS zoom scans. Technically independent confirmation was demonstrated by triple quadrupole LC-MS/MS selected reaction monitoring (SRM) assays with three peptides specific to human ApoA1 (DLATVYVDVLK, WQEEMELYR, and VSFLSALEEYTK) using (18)O/(16)O-labeled samples and further verified with AQUA peptides as internal standards for quantification. In 50 patient samples (24 HCV and 26 HCC), all three SRM assays yielded highly similar differential expression of ApoA1 in HCC and HCV patients. These results validated the SRM assays, which were independently confirmed by Western blotting. Thus, ApoA1 is a candidate member of an SRM biomarker panel for early diagnosis, prognosis, and monitoring of HCC. Future multiplexing of SRM assays for other candidate biomarkers is envisioned to develop a biomarker panel for subsequent verification and validation studies.


Subject(s)
Apolipoprotein A-I/genetics , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Hepatitis C, Chronic/genetics , Liver Cirrhosis/genetics , Liver Neoplasms/genetics , Adult , Amino Acid Sequence , Apolipoprotein A-I/blood , Aptamers, Peptide/chemistry , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/etiology , Early Diagnosis , Electrophoresis, Gel, Two-Dimensional , Fluorescent Dyes/chemistry , Hepacivirus/isolation & purification , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/diagnosis , Humans , Isotope Labeling , Liver Cirrhosis/blood , Liver Cirrhosis/diagnosis , Liver Cirrhosis/etiology , Liver Neoplasms/blood , Liver Neoplasms/diagnosis , Liver Neoplasms/etiology , Male , Middle Aged , Molecular Sequence Data , Oxygen Isotopes , Sensitivity and Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
Neurobiol Dis ; 72 Pt A: 92-103, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25237037

ABSTRACT

Insulin is a key hormone regulating metabolism. Insulin binding to cell surface insulin receptors engages many signaling intermediates operating in parallel and in series to control glucose, energy, and lipids while also regulating mitogenesis and development. Perturbations in the function of any of these intermediates, which occur in a variety of diseases, cause reduced sensitivity to insulin and insulin resistance with consequent metabolic dysfunction. Chronic inflammation ensues which exacerbates compromised metabolic homeostasis. Since insulin has a key role in learning and memory as well as directly regulating ERK, a kinase required for the type of learning and memory compromised in early Alzheimer's disease (AD), insulin resistance has been identified as a major risk factor for the onset of AD. Animal models of AD or insulin resistance or both demonstrate that AD pathology and impaired insulin signaling form a reciprocal relationship. Of note are human and animal model studies geared toward improving insulin resistance that have led to the identification of the nuclear receptor and transcription factor, peroxisome proliferator-activated receptor gamma (PPARγ) as an intervention tool for early AD. Strategic targeting of alternate nodes within the insulin signaling network has revealed disease-stage therapeutic windows in animal models that coalesce with previous and ongoing clinical trial approaches. Thus, exploiting the connection between insulin resistance and AD provides powerful opportunities to delineate therapeutic interventions that slow or block the pathogenesis of AD.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Insulin Resistance , Insulin/metabolism , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Encephalitis/metabolism , Humans , Learning/physiology , MAP Kinase Signaling System , PPAR gamma/metabolism
8.
J Neurosci ; 32(47): 16725-35a, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23175826

ABSTRACT

We previously reported that the peroxisome proliferator-activated receptor γ (PPARγ) agonist rosiglitazone (RSG) improved hippocampus-dependent cognition in the Alzheimer's disease (AD) mouse model, Tg2576. RSG had no effect on wild-type littermate cognitive performance. Since extracellular signal-regulated protein kinase mitogen-activated protein kinase (ERK MAPK) is required for many forms of learning and memory that are affected in AD, and since both PPARγ and ERK MAPK are key mediators of insulin signaling, the current study tested the hypothesis that RSG-mediated cognitive improvement induces a hippocampal PPARγ pattern of gene and protein expression that converges with the ERK MAPK signaling axis in Tg2576 AD mice. In the hippocampal PPARγ transcriptome, we found significant overlap between peroxisome proliferator response element-containing PPARγ target genes and ERK-regulated, cAMP response element-containing target genes. Within the Tg2576 dentate gyrus proteome, RSG induced proteins with structural, energy, biosynthesis and plasticity functions. Several of these proteins are known to be important for cognitive function and are also regulated by ERK MAPK. In addition, we found the RSG-mediated augmentation of PPARγ and ERK2 activity during Tg2576 cognitive enhancement was reversed when hippocampal PPARγ was pharmacologically antagonized, revealing a coordinate relationship between PPARγ transcriptional competency and phosphorylated ERK that is reciprocally affected in response to chronic activation, compared with acute inhibition, of PPARγ. We conclude that the hippocampal transcriptome and proteome induced by cognitive enhancement with RSG harnesses a dysregulated ERK MAPK signal transduction pathway to overcome AD-like cognitive deficits in Tg2576 mice. Thus, PPARγ represents a signaling system that is not crucial for normal cognition yet can intercede to restore neural networks compromised by AD.


Subject(s)
Hippocampus/physiology , MAP Kinase Signaling System/physiology , Nootropic Agents/pharmacology , PPAR gamma/physiology , Signal Transduction/physiology , Thiazolidinediones/pharmacology , Amyloid beta-Peptides/metabolism , Animals , Blotting, Western , Cell Nucleus/physiology , Conditioning, Psychological , Electroshock , Fear , Female , Injections, Intraventricular , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , PPAR gamma/antagonists & inhibitors , Polymerase Chain Reaction , Rosiglitazone , Tandem Mass Spectrometry , Transcriptome/physiology
9.
J Proteome Res ; 9(8): 4306-12, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20568695

ABSTRACT

We describe a method for ratio estimations in (18)O-water labeling experiments acquired from low resolution isotopically resolved data. The method is implemented in a software package specifically designed for use in experiments making use of zoom-scan mode data acquisition. Zoom-scan mode data allow commonly used ion trap mass spectrometers to attain isotopic resolution, which makes them amenable to use in labeling schemes such as (18)O-water labeling, but algorithms and software developed for high resolution instruments may not be appropriate for the lower resolution data acquired in zoom-scan mode. The use of power spectrum analysis is proposed as a general approach that may be uniquely suited to these data types. The software implementation uses a power spectrum to remove high-frequency noise and band-filter contributions from coeluting species of differing charge states. From the elemental composition of a peptide sequence, we generate theoretical isotope envelopes of heavy-light peptide pairs in five different ratios; these theoretical envelopes are correlated with the filtered experimental zoom scans. To automate peptide quantification in high-throughput experiments, we have implemented our approach in a computer program, MassXplorer. We demonstrate the application of MassXplorer to two model mixtures of known proteins and to a complex mixture of mouse kidney cortical extract. Comparison with another algorithm for ratio estimations demonstrates the increased precision and automation of MassXplorer.


Subject(s)
Algorithms , Isotope Labeling/methods , Proteins/analysis , Proteomics/methods , Software , Spectrum Analysis/methods , Tandem Mass Spectrometry/methods , Animals , Cattle , Chromatography, Liquid , Computational Biology/methods , Cytochromes c/analysis , Horses , Kidney Cortex/chemistry , Mice , Oxygen Isotopes/analysis , Proteins/metabolism , Serum Albumin, Bovine/analysis
10.
J Neuroendocrinol ; 32(10): e12904, 2020 10.
Article in English | MEDLINE | ID: mdl-33000549

ABSTRACT

Neural progestin receptors (PR) function in reproduction, neural development, neuroprotection, learning, memory and the anxiety response. In the absence of progestins, PR can be activated by dopamine (DA) in the rodent hypothalamus to elicit female sexual behaviour. The present study investigated mechanisms of DA activation of PR by testing the hypothesis that proteins from DA-treated hypothalami interact with PR in the absence of progestins. Ovariectomised, oestradiol-primed mice were infused with a D1-receptor agonist, SKF38393 (SKF), into the third ventricle 30 minutes prior to death. Proteins from SKF-treated hypothalami were pulled-down with glutathione S-transferase-tagged mouse PR-A or PR-B and the interactomes were analysed by mass spectrometry. The largest functional group to interact with PR-A in a DA-dependent manner was synaptic proteins. To test the hypothesis that DA activation of PR regulates synaptic proteins, we developed oestradiol-induced PR-expressing hypothalamic-like neurones derived from human-induced pluripotent stem cells (hiPSCs). Similar to progesterone (P4), SKF treatment of hiPSCs increased synapsin1/2 expression. This SKF-dependent effect was blocked by the PR antagonist RU486, suggesting that PR are necessary for this DA-induced increase. The second largest DA-dependent PR-A protein interactome comprised metabolic regulators involved in glucose metabolism, lipid synthesis and mitochondrial energy production. Interestingly, hypothalamic proteins interacted with PR-A, but not PR-B, in an SKF-dependent manner, suggesting that DA promotes the interaction of multiple hypothalamic proteins with PR-A. These in vivo and in vitro results indicate novel mechanisms by which DA can differentially activate PR isoforms in the absence of P4 and provide a better understanding of ligand-independent PR activation in reproductive, metabolic and mental health disorders in women.


Subject(s)
Dopamine/pharmacology , Nerve Tissue Proteins/metabolism , Receptors, Progesterone/metabolism , Animals , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Progesterone/pharmacology , Protein Binding/drug effects , Protein Isoforms/drug effects , Protein Isoforms/metabolism , Receptors, Progesterone/drug effects , Signal Transduction/drug effects
11.
Eur J Heart Fail ; 11(7): 638-47, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19502378

ABSTRACT

AIMS: Activation of the receptor for advanced glycation end products (RAGE) is associated with long-term complications in diabetes mellitus. In this study, we tested whether RAGE activation in the diabetic myocardium is implicated in the development of cardiac dysfunction. METHODS AND RESULTS: Using MRI and conductance catheter techniques, we evaluated cardiac function in a type 2 diabetic mouse model (db/db), and assessed the effect of blocking RAGE with a RAGE antibody. Gene expressions were evaluated in samples of myocardial tissue. Diabetic db/db mice demonstrated an accelerated age-dependent deterioration in cardiac function associated with altered expression of genes related to cardiac structure and function. Blockage of RAGE signalling prevented the reduction in systolic function (preload recruitable stroke work: 109.8 +/- 13.8 vs. 94.5 +/- 14.9 mmHg/microL, P = 0.04) and development of increased LV diastolic chamber stiffness (0.18 +/- 0.05 vs. 0.27 +/- 0.07 mmHg, P = 0.01). The cardiac expression of collagen (col1a1) was reduced by approximately 45% and the expression of myosin was switched from the foetal isoform (MHCbeta) to the adult isoform (MHCalpha). CONCLUSION: Activation of RAGE is a significant pathogenetic mechanism for the development of cardiac dysfunction in type 2 diabetes. The underlying mechanisms involve not only the passive biophysical properties of the myocardium but also myocyte function.


Subject(s)
Cardiomyopathies/etiology , Cardiomyopathies/prevention & control , Diabetes Mellitus, Type 2/complications , Glycation End Products, Advanced/antagonists & inhibitors , Heart Function Tests , Analysis of Variance , Animals , Cardiomyopathies/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase/metabolism , Risk Factors , Systole
12.
Front Aging Neurosci ; 11: 38, 2019.
Article in English | MEDLINE | ID: mdl-30930764

ABSTRACT

A major aspect of mammalian aging is the decline in functional competence of many self-renewing cell types, including adult-born neuronal precursors in a process termed neurogenesis. Adult neurogenesis is limited to specific brain regions in the mammalian brain, such as the subgranular zone (SGZ) of the hippocampus. Alterations in adult neurogenesis appear to be a common hallmark in different neurodegenerative diseases including Alzheimer's disease (AD). We and others have shown that PPARγ agonism improves cognition in preclinical models of AD as well as in several pilot clinical trials. Context discrimination is recognized as a cognitive task supported by proliferation and differentiation of adult-born neurons in the dentate gyrus of the hippocampus that we and others have previously shown declines with age. We therefore postulated that PPARγ agonism would positively impact context discrimination in middle-aged mice via mechanisms that influence proliferation and differentiation of adult-born neurons arising from the SGZ. To achieve our objective, 8-months old mice were left untreated or treated with the FDA-approved PPARγ agonist, rosiglitazone then tested for context discrimination learning and memory, followed by immunofluorescence evaluation of hippocampal SGZ cell proliferation and neuron survival. We found that PPARγ agonism enhanced context discrimination performance in middle-aged mice concomitant with stimulated SGZ cell proliferation, but not new neuron survival. Focal cranial irradiation that destroys neurogenesis severely compromised context discrimination in middle-aged mice yet rosiglitazone treatment significantly improved cognitive performance through an anti-inflammatory mechanism and resurrection of the neurogenic niche. Thus, we have evidence for divergent mechanisms by which PPARγ agonism impinges upon aging-related versus cranial irradiation-induced deficits in context discrimination learning and memory.

13.
Endocrinology ; 149(10): 5272-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18566116

ABSTRACT

In vitro studies reveal that nuclear receptor coactivators enhance the transcriptional activity of steroid receptors, including estrogen (ER) and progestin receptors (PR), through ligand-dependent interactions. Whereas work from our laboratory and others shows that steroid receptor coactivator-1 (SRC-1) is essential for efficient ER and PR action in brain, very little is known about receptor-coactivator interactions in brain. In the present studies, pull-down assays were used to test the hypotheses that SRC-1 from hypothalamic and hippocampal tissue physically associate with recombinant PR or ER in a ligand-dependent manner. SRC-1, from hypothalamus or hippocampus, interacted with PR-A and PR-B in the presence of an agonist, but not in the absence of ligand or in the presence of a selective PR modulator, RU486. Interestingly, SRC-1 from brain associated more with PR-B, the stronger transcriptional activator, than with PR-A. In addition, SRC-1 from brain, which was confirmed by mass spectrometry, interacted with ERalpha and ERbeta in the presence of agonist but not when unliganded or in the presence of the selective ER modulator, tamoxifen. Furthermore, SRC-1 from hypothalamus, but not hippocampus, interacted more with ERalpha than ERbeta, suggesting distinct expression patterns of other cofactors in these brain regions. These findings suggest that interactions of SRC-1 from brain with PR and ER are dependent on ligand, receptor subtype, and brain region to manifest the pleiotropic functional consequences that underlie steroid-regulated behaviors. The present findings reveal distinct contrasts with previous cell culture studies and emphasize the importance of studying receptor-coactivator interactions using biologically relevant tissue.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Histone Acetyltransferases/metabolism , Hypothalamus/metabolism , Receptors, Progesterone/metabolism , Transcription Factors/metabolism , Animals , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Glutathione Transferase/genetics , Hippocampus/metabolism , Hormone Antagonists/pharmacology , Ligands , Mass Spectrometry , Mifepristone/pharmacology , Nuclear Receptor Coactivator 1 , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tamoxifen/pharmacology
14.
Mol Cell Endocrinol ; 294(1-2): 52-60, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-18694803

ABSTRACT

PKC signaling is critical for follicular development and the induction of ovulatory genes including Pgr, Prkg2, and Cyp11a1 (SCC). We investigated PKC signaling mechanisms in the JC-410 porcine granulosa cell line stably expressing an SCC-luciferase reporter gene containing 2kb of the porcine SCC promoter. Addition of phorbol 12-myristate 13-acetate (PMA), which activates protein kinase C, induced the promoter approximately 6-fold over the basal levels in 4h. This effect was predominantly mediated by the PKC beta and delta isoforms. PMA-mediated induction of the SCC promoter was sensitive to inhibition of ERK1/2 or JNK. Inhibition of p38 MAP kinase or Src tyrosine kinase did not alter the PMA-mediated inducibility of the promoter. SCC promoter induction in response to PMA treatment required basal EGF-receptor activity, but did not involve ectodomain shedding. Western blot analyses using phospho-specific antibodies showed that PMA treatment of JC-410 cells induced phosphorylation of MEK1/2, ERK1/2, and its downstream target p90 RSK at 15min. We also documented the rapid phosphorylation of JNK1/2 in response to PMA treatment. Phosphorylation of ERK and JNK was robust and sustained in contrast to activation of PKA and EGF-receptor signaling in these cells. Pretreatment of JC-410 granulosa cells with IGF-1 had a synergistic effect on PMA-mediated induction of the SCC promoter. We demonstrated the importance of PMA activation of ERK signaling and the synergism with IGF-1 by showing similar responses for Prkg2 expression in primary granulosa cells. In conclusion, our studies demonstrated PMA activation of ERK and JNK signaling which is relevant in the regulation of gene expression during follicular development, ovulation, and luteinization.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Granulosa Cells/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Protein Kinase C/metabolism , Animals , Cell Line , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Cholesterol Side-Chain Cleavage Enzyme/genetics , Colforsin/pharmacology , Enzyme Activation/drug effects , Enzyme Induction/drug effects , ErbB Receptors/metabolism , Female , Granulosa Cells/cytology , Granulosa Cells/drug effects , Humans , Insulin-Like Growth Factor I/pharmacology , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Promoter Regions, Genetic , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Swine , Tetradecanoylphorbol Acetate/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Proteome Sci ; 6: 15, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18501002

ABSTRACT

BACKGROUND: Proteomic methodologies increasingly have been applied to the kidney to map the renal cortical proteome and to identify global changes in renal proteins induced by diseases such as diabetes. While progress has been made in establishing a renal cortical proteome using 1-D or 2-DE and mass spectrometry, the number of proteins definitively identified by mass spectrometry has remained surprisingly small. Low coverage of the renal cortical proteome as well as our interest in diabetes-induced changes in proteins found in the renal cortex prompted us to perform an in-depth proteomic analysis of mouse renal cortical tissue. RESULTS: We report a large scale analysis of mouse renal cortical proteome using SCX prefractionation strategy combined with HPLC - tandem mass spectrometry. High-confidence identification of ~2,000 proteins, including cytoplasmic, nuclear, plasma membrane, extracellular and unknown/unclassified proteins, was obtained by separating tryptic peptides of renal cortical proteins into 60 fractions by SCX prior to LC-MS/MS. The identified proteins represented the renal cortical proteome with no discernible bias due to protein physicochemical properties, subcellular distribution, biological processes, or molecular function. The highest ranked molecular functions were characteristic of tubular epithelium, and included binding, catalytic activity, transporter activity, structural molecule activity, and carrier activity. Comparison of this renal cortical proteome with published human urinary proteomes demonstrated enrichment of renal extracellular, plasma membrane, and lysosomal proteins in the urine, with a lack of intracellular proteins. Comparison of the most abundant proteins based on normalized spectral abundance factor (NSAF) in this dataset versus a published glomerular proteome indicated enrichment of mitochondrial proteins in the former and cytoskeletal proteins in the latter. CONCLUSION: A whole tissue extract of the mouse kidney cortex was analyzed by an unbiased proteomic approach, yielding a dataset of ~2,000 unique proteins identified with strict criteria to ensure a high level of confidence in protein identification. As a result of extracting all proteins from the renal cortex, we identified an exceptionally wide range of renal proteins in terms of pI, MW, hydrophobicity, abundance, and subcellular location. Many of these proteins, such as low-abundance proteins, membrane proteins and proteins with extreme values in pI or MW are traditionally under-represented in 2-DE-based proteomic analysis.

16.
J Alzheimers Dis ; 64(s1): S427-S453, 2018.
Article in English | MEDLINE | ID: mdl-29710715

ABSTRACT

Insulin resistance can occur when the body is unable to respond to insulin even in excess. In the brain, insulin manages glucose metabolism in regions such as the hippocampus and plays a key role in directly regulating ERK, a kinase required for the type of memory compromised in early Alzheimer's disease (AD). Human imaging studies show that brain glucose utilization declines with age and is notably impaired in subjects with early AD. Likewise, animal models of AD or insulin resistance, or both, demonstrate that dysfunctional insulin signaling and insulin resistance in the brain have reciprocity with neuroinflammation and aberrant accumulation of amyloid-ß (Aß), pathological hallmarks in AD. As such, the association between brain insulin activity and AD has led to clinical trials testing the efficacy of insulin and insulin-sensitizing drugs to intervene in AD. Based on recent inquiries to ClinicalTrials.gov, we evaluated thirty-three clinical studies related to AD and insulin. The search filtered for interventional clinical trials to test FDA-approved drugs or substances that impinge upon the insulin signaling pathway. Insulin, metformin, and thiazolidinediones were the three main interventions assessed. Overall, these strategies are expected to negate the effects of brain insulin resistance by targeting insulin signaling pathways involved in neuroinflammation, metabolic homeostasis, synaptic functional and structural integrity. The goal of this review is to provide an update on insulin and ERK signaling in relation to memory, its decline in early AD, and provide an overview of clinical trials related to insulin for early AD intervention.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Insulin/metabolism , Animals , Humans
17.
Diabetes ; 55(5): 1252-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16644679

ABSTRACT

Evidence of diabetes-induced nuclear factor-kappaB (NF-kappaB) activation has been provided with DNA binding assays or nuclear localization with immunohistochemistry, but few studies have explored mechanisms involved. We examined effects of diabetes on proteins comprising NF-kappaB canonical and noncanonical activation pathways in the renal cortex of diabetic mice. Plasma concentrations of NF-kappaB-regulated cytokines were increased after 1 month of hyperglycemia, but most returned to control levels or lower by 3 months, when the same cytokines were increased significantly in renal cortex. Cytosolic content of NF-kappaB canonical pathway proteins did not differ between experimental groups after 3 months of diabetes, while NF-kappaB noncanonical pathway proteins were affected, including increased phosphorylation of inhibitor of kappaB kinase-alpha and several fold increases in NF-kappaB-inducing kinase and RelB, which were predominantly located in tubular epithelial cells. Nuclear content of all NF-kappaB pathway proteins was decreased by diabetes, with the largest change in RelB and p50 (approximately twofold decrease). Despite this decrease, measurable increases in protein binding to DNA in diabetic versus control nuclear extracts were observed with electrophoretic mobility shift assay. These results provide evidence for chronic NF-kappaB activation in the renal cortex of db/db mice and suggest a novel, diabetes-linked mechanism involving both canonical and noncanonical NF-kappaB pathway proteins.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Kidney Cortex/metabolism , NF-kappa B/metabolism , Animals , Cell Nucleus/immunology , Cell Nucleus/metabolism , Chemokines/biosynthesis , Cytokines/biosynthesis , Cytosol/immunology , Cytosol/metabolism , DNA/metabolism , Diabetes Mellitus, Experimental/immunology , Mice , NF-kappa B/classification
18.
Neurobiol Aging ; 58: 1-13, 2017 10.
Article in English | MEDLINE | ID: mdl-28688899

ABSTRACT

Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder worldwide. While the causes of AD are not known, several risk factors have been identified. Among these, type two diabetes (T2D), a chronic metabolic disease, is one of the most prevalent risk factors for AD. Insulin resistance, which is associated with T2D, is defined as diminished or absent insulin signaling and is reflected by peripheral blood hyperglycemia and impaired glucose clearance. In this study, we used complementary approaches to probe for peripheral insulin resistance, central nervous system (CNS) insulin sensitivity and energy homeostasis in Tg2576 and 3xTg-AD mice, two widely used animal models of AD. We report that CNS insulin signaling abnormalities are evident months before peripheral insulin resistance. In addition, we find that brain energy metabolism is differentially altered in both mouse models, with 3xTg-AD mice showing more extensive changes. Collectively, our data suggest that early AD may reflect engagement of different signaling networks that influence CNS metabolism, which in turn may alter peripheral insulin signaling.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Central Nervous System/metabolism , Energy Metabolism/physiology , Homeostasis/physiology , Insulin Resistance , Insulin/metabolism , Aging/metabolism , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Female , Glucose/metabolism , Hyperglycemia/metabolism , Male , Mice, Transgenic , Risk Factors , Signal Transduction/physiology
19.
eNeuro ; 4(5)2017.
Article in English | MEDLINE | ID: mdl-28955722

ABSTRACT

Progestins bind to the progestin receptor (PR) isoforms, PR-A and PR-B, in brain to influence development, female reproduction, anxiety, and stress. Hormone-activated PRs associate with multiple proteins to form functional complexes. In the present study, proteins from female mouse hypothalamus that associate with PR were isolated using affinity pull-down assays with glutathione S-transferase-tagged mouse PR-A and PR-B. Using complementary proteomics approaches, reverse phase protein array (RPPA) and mass spectrometry, we identified hypothalamic proteins that interact with PR in a ligand-dependent and isoform-specific manner and were confirmed by Western blot. Synaptic proteins, including synapsin-I and synapsin-II, interacted with agonist-bound PR isoforms, suggesting that both isoforms function in synaptic plasticity. In further support, synaptogyrin-III and synapsin-III associated with PR-A and PR-B, respectively. PR also interacted with kinases, including c-Src, mTOR, and MAPK1, confirming phosphorylation as an integral process in rapid effects of PR in the brain. Consistent with a role in transcriptional regulation, PR associated with transcription factors and coactivators in a ligand-specific and isoform-dependent manner. Interestingly, both PR isoforms associated with a key regulator of energy homeostasis, FoxO1, suggesting a novel role for PR in energy metabolism. Because many identified proteins in this PR interactome are synaptic proteins, we tested the hypothesis that progestins function in synaptic plasticity. Indeed, progesterone enhanced synaptic density, by increasing synapsin-I-positive synapses, in rat primary cortical neuronal cultures. This novel combination of RPPA and mass spectrometry allowed identification of PR action in synaptic remodeling and energy homeostasis and reveals unique roles for progestins in brain function and disease.


Subject(s)
Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Progesterone/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Estradiol/pharmacology , Female , Gene Expression Regulation/drug effects , Glutathione Transferase/metabolism , Ligands , Mice , Mice, Inbred C57BL , Neurons/drug effects , Ovariectomy , Protein Binding , Protein Isoforms/metabolism , Receptors, Progesterone/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic
20.
Exp Neurol ; 295: 1-17, 2017 09.
Article in English | MEDLINE | ID: mdl-28522250

ABSTRACT

BACKGROUND: Cognitive impairment in humans with Alzheimer's disease (AD) and in animal models of Aß-pathology can be ameliorated by treatments with the nuclear receptor peroxisome proliferator-activated receptor-gamma (PPARγ) agonists, such as rosiglitazone (RSG). Previously, we demonstrated that in the Tg2576 animal model of AD, RSG treatment rescued cognitive deficits and reduced aberrant activity of granule neurons in the dentate gyrus (DG), an area critical for memory formation. METHODS: We used a combination of mass spectrometry, confocal imaging, electrophysiology and split-luciferase assay and in vitro phosphorylation and Ingenuity Pathway Analysis. RESULTS: Using an unbiased, quantitative nano-LC-MS/MS screening, we searched for potential molecular targets of the RSG-dependent rescue of DG granule neurons. We found that S226 phosphorylation of fibroblast growth factor 14 (FGF14), an accessory protein of the voltage-gated Na+ (Nav) channels required for neuronal firing, was reduced in Tg2576 mice upon treatment with RSG. Using confocal microscopy, we confirmed that the Tg2576 condition decreased PanNav channels at the AIS of the DG, and that RSG treatment of Tg2576 mice reversed the reduction in PanNav channels. Analysis from previously published data sets identified correlative changes in action potential kinetics in RSG-treated T2576 compared to untreated and wildtype controls. In vitro phosphorylation and mass spectrometry confirmed that the multifunctional kinase GSK-3ß, a downstream target of insulin signaling highly implicated in AD, phosphorylated FGF14 at S226. Assembly of the FGF14:Nav1.6 channel complex and functional regulation of Nav1.6-mediated currents by FGF14 was impaired by a phosphosilent S226A mutation. Bioinformatics pathway analysis of mass spectrometry and biochemistry data revealed a highly interconnected network encompassing PPARγ, FGF14, SCN8A (Nav 1.6), and the kinases GSK-3 ß, casein kinase 2ß, and ERK1/2. CONCLUSIONS: These results identify FGF14 as a potential PPARγ-sensitive target controlling Aß-induced dysfunctions of neuronal activity in the DG underlying memory loss in early AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Fibroblast Growth Factors/drug effects , PPAR gamma/agonists , Amino Acid Sequence , Animals , Axons/metabolism , Dentate Gyrus/metabolism , Female , Fibroblast Growth Factors/genetics , HEK293 Cells , Humans , Insulin Resistance , Male , Mice , Mice, Knockout , Mutation/genetics , Phosphorylation , Rosiglitazone , Sodium Channels/genetics , Sodium Channels/metabolism , Thiazolidinediones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL