Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Am J Physiol Regul Integr Comp Physiol ; 326(4): R311-R318, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38344803

ABSTRACT

Aphagic hibernators such as the golden-mantled ground squirrel (GMGS; Callospermophilus lateralis) can fast for months and exhibit profound seasonal fluctuations in body weight, food intake, and behavior. Brain-derived neurotrophic factor (BDNF) regulates cellular and systemic metabolism via mechanisms that are conserved across mammalian species. In this study, we characterized regional changes in BDNF with hibernation, hypothermia, and seasonal cycle in GMGS. Analysis of BDNF protein concentrations by ELISA revealed overlapping seasonal patterns in the hippocampus and hypothalamus, where BDNF levels were highest in summer and lowest in winter. BDNF is the primary ligand for receptor tyrosine kinase B (TrkB), and BDNF/TrkB signaling in the brain potently regulates energy expenditure. To examine the functional relevance of seasonal variation in BDNF, hibernating animals were injected with the small molecule TrkB agonist 7,8-dihydroxyflavone (DHF) daily for 2 wk. When compared with vehicle, DHF-treated animals exhibited fewer torpor bouts and shorter bout durations. These results suggest that activating BDNF/TrkB disrupts hibernation and raise intriguing questions related to the role of BDNF as a potential regulatory mechanism or downstream response to seasonal changes in body temperature and environment.NEW & NOTEWORTHY Golden-mantled ground squirrels exhibit dramatic seasonal fluctuations in metabolism and can fast for months while hibernating. Brain-derived neurotrophic factor is an essential determinant of cellular and systemic metabolism, and in this study, we characterized seasonal fluctuations in BDNF expression and then administered the small molecule BDNF mimetic 7,8-dihydroxyflavone (DHF) in hibernating squirrels. The results indicate that activating BDNF/TrkB signaling disrupts hibernation, with implications for synaptic homeostasis in prolonged hypometabolic states.


Subject(s)
Hibernation , Animals , Hibernation/physiology , Brain-Derived Neurotrophic Factor/metabolism , Seasons , Body Temperature/physiology , Sciuridae/metabolism
2.
Diabetes ; 72(2): 245-260, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36367881

ABSTRACT

Preferential energy storage in subcutaneous adipose tissue (SAT) confers protection against obesity-induced pathophysiology in females. Females also exhibit distinct immunological responses, relative to males. These differences are often attributed to sex hormones, but reciprocal interactions between metabolism, immunity, and gonadal steroids remain poorly understood. We systematically characterized adipose tissue hypertrophy, sex steroids, and inflammation in male and female mice after increasing durations of high-fat diet (HFD)-induced obesity. After observing that sex differences in adipose tissue distribution before HFD were correlated with lasting protection against inflammation in females, we hypothesized that a priori differences in the ratio of subcutaneous to visceral fat might mediate this relationship. To test this, male and female mice underwent SAT lipectomy (LPX) or sham surgery before HFD challenge, followed by analysis of glial reactivity, adipose tissue inflammation, and reproductive steroids. Because LPX eliminated female resistance to the proinflammatory effects of HFD without changing circulating sex hormones, we conclude that sexually dimorphic organization of subcutaneous and visceral fat determines susceptibility to inflammation in obesity.


Subject(s)
Neuroinflammatory Diseases , Sex Characteristics , Female , Male , Mice , Animals , Tissue Distribution , Obesity/metabolism , Inflammation , Gonadal Steroid Hormones
3.
Nat Commun ; 12(1): 4623, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34330904

ABSTRACT

Visceral obesity increases risk of cognitive decline in humans, but subcutaneous adiposity does not. Here, we report that beige adipocytes are indispensable for the neuroprotective and anti-inflammatory effects of subcutaneous fat. Mice lacking functional beige fat exhibit accelerated cognitive dysfunction and microglial activation with dietary obesity. Subcutaneous fat transplantation also protects against chronic obesity in wildtype mice via beige fat-dependent mechanisms. Beige adipocytes restore hippocampal synaptic plasticity following transplantation, and these effects require the anti-inflammatory cytokine interleukin-4 (IL4). After observing beige fat-mediated induction of IL4 in meningeal T-cells, we investigated the contributions of peripheral lymphocytes in donor fat. There was no sign of donor-derived lymphocyte trafficking between fat and brain, but recipient-derived lymphocytes were required for the effects of transplantation on cognition and microglial morphology. These findings indicate that beige adipocytes oppose obesity-induced cognitive impairment, with a potential role for IL4 in the relationship between beige fat and brain function.


Subject(s)
Adipocytes, Beige/metabolism , Adipose Tissue, Beige/metabolism , Adiposity , Obesity/metabolism , Subcutaneous Fat/metabolism , Adipocytes, Beige/cytology , Animals , Anti-Inflammatory Agents/metabolism , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Diet, High-Fat/adverse effects , Humans , Interleukin-4/metabolism , Mice, Obese , Neuronal Plasticity/physiology , Neuroprotective Agents/metabolism , Obesity/etiology , Obesity/physiopathology , Subcutaneous Fat/transplantation , T-Lymphocytes/metabolism
4.
Brain Behav ; 11(2): e01973, 2021 02.
Article in English | MEDLINE | ID: mdl-33382528

ABSTRACT

INTRODUCTION: Several clinical studies have tested the efficacy of insulin-sensitizing drugs for cognitive enhancement in Alzheimer's disease (AD) patients, as type 2 diabetes (T2D) is a well-recognized risk factor for AD. Pilot studies assessing FDA-approved diabetes drugs in subjects with early-stage disease have found cognitive benefit in subjects comorbid for insulin resistance. In AD mouse models with concomitant insulin resistance, we have shown that 4 weeks of RSG can reverse peripheral and central insulin resistance concomitant with rescue of hippocampus-dependent fear learning and memory and hippocampal circuitry deficits in 9-month-old (9MO) Tg2576 mice with no effect in wild-type (WT) mice. Bioinformatics analysis of genomic and proteomic data reveals an intimate link between PPARγ and MAPK/ERK signaling in the hippocampus. We then demonstrated a direct interaction between PPARγ and phospho-ERK in vitro and in vivo during memory consolidation. The translational value of this discovery is evidenced by the positive correlational relationship between human AD postmortem brain levels of pERK-PPARγ nuclear complexes with cognitive reserve. METHODS: We tested whether insulin sensitizer therapy could rescue spatial navigation, context discrimination, and object recognition learning and memory in aged wild-type and Tg2576 mice in addition to hippocampus-dependent contextual fear learning and memory, as we have previously reported. RESULTS: We found that rosiglitazone treatment improved cognitive domains that predominantly rely upon the dorsal hippocampus rather than those that additionally engage the ventral hippocampus. CONCLUSION: These results suggest that insulin sensitizer therapy with rosiglitazone improved age- and AD-related learning and memory deficits in circuit selective ways.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Aged , Alzheimer Disease/drug therapy , Animals , Cognition , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Hippocampus/metabolism , Humans , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , PPAR gamma/metabolism , Proteomics , Rosiglitazone/pharmacology
5.
Pharmacol Biochem Behav ; 197: 172999, 2020 10.
Article in English | MEDLINE | ID: mdl-32702397

ABSTRACT

Antipsychotic drugs (APDs) have a variety of important therapeutic applications for neuropsychiatric disorders. However, they are routinely prescribed off-label across all age categories, a controversial practice given their potential for producing metabolic and extrapyramidal side effects. Evidence also suggests that chronic treatment with some APDs may lead to impairments in cognition and decreases in brain volume, although these findings are controversial. The purpose of the studies described here was to evaluate one of the most commonly prescribed APDs, quetiapine, for chronic effects on recognition memory, brain-derived neurotrophic factor (BDNF), its precursor proBDNF, as well as relevant downstream signaling molecules that are known to influence neuronal plasticity and cognition. Multiple cohorts of adult rats were treated with quetiapine (25.0 mg/kg/day) for 30 or 90 days in their drinking water then evaluated for drug effects on motor function in a catalepsy assessment, recognition memory in a spontaneous novel object recognition (NOR) task, and BDNF-related signaling molecules in the post mortem hippocampus via Western Blot. The results indicated that oral quetiapine at a dose that did not induce catalepsy, led to time-dependent impairments in NOR performance, increases in the proBDNF/BDNF ratio, and decreases in Akt and CREB phosphorylation in the hippocampus. These results indicate that chronic treatment with quetiapine has the potential to adversely affect recognition memory and neurotrophin-related signaling molecules that support synaptic plasticity and cognitive function. Given the widespread use this APD across multiple conditions and patient populations, such long-term effects observed in animals should be considered.


Subject(s)
Antipsychotic Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Quetiapine Fumarate/pharmacology , Recognition, Psychology/drug effects , Signal Transduction/drug effects , Administration, Oral , Animals , Antipsychotic Agents/administration & dosage , Behavior, Animal/drug effects , Catalepsy , Cognition/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Quetiapine Fumarate/administration & dosage , Rats , Rats, Wistar
6.
J Clin Invest ; 130(4): 1961-1976, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31935195

ABSTRACT

Induction of the inflammasome protein cryopyrin (NLRP3) in visceral adipose tissue (VAT) promotes release of the proinflammatory cytokine IL-1ß in obesity. Although this mechanism contributes to peripheral metabolic dysfunction, effects on the brain remain unexplored. We investigated whether visceral adipose NLRP3 impairs cognition by activating microglial IL-1 receptor 1 (IL-1R1). After observing protection against obesity-induced neuroinflammation and cognitive impairment in NLRP3-KO mice, we transplanted VAT from obese WT or NLRP3-KO donors into lean recipient mice. Transplantation of VAT from a WT donor (TRANSWT) increased hippocampal IL-1ß and impaired cognition, but VAT transplants from comparably obese NLRP3-KO donors (TRANSKO) had no effect. Visceral adipose NLRP3 was required for deficits in long-term potentiation (LTP) in transplant recipients, and LTP impairment in TRANSWT mice was IL-1 dependent. Flow cytometric and gene expression analyses revealed that VAT transplantation recapitulated the effects of obesity on microglial activation and IL-1ß gene expression, and visualization of hippocampal microglia revealed similar effects in vivo. Inducible ablation of IL-1R1 in CX3CR1-expressing cells eliminated cognitive impairment in mice with dietary obesity and in transplant recipients and restored immunoquiescence in hippocampal microglia. These results indicate that visceral adipose NLRP3 impairs memory via IL-1-mediated microglial activation and suggest that NLRP3/IL-1ß signaling may underlie correlations between visceral adiposity and cognitive impairment in humans.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Cognition , Hippocampus/metabolism , Intra-Abdominal Fat/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Obesity/metabolism , Receptors, Interleukin-1 Type I/metabolism , Animals , CX3C Chemokine Receptor 1/genetics , Hippocampus/pathology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intra-Abdominal Fat/pathology , Intra-Abdominal Fat/transplantation , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Obesity/genetics , Obesity/pathology , Receptors, Interleukin-1 Type I/genetics , Signal Transduction/genetics
7.
Pharmacol Biochem Behav ; 189: 172853, 2020 02.
Article in English | MEDLINE | ID: mdl-31945381

ABSTRACT

Antipsychotic drugs (APDs) are essential for the treatment of schizophrenia and other neuropsychiatric illnesses such as bipolar disease. However, they are also extensively prescribed off-label for many other conditions, a practice that is controversial given their potential for long-term side effects. There is clinical and preclinical evidence that chronic treatment with some APDs may lead to impairments in cognition and decreases in brain volume, although the molecular mechanisms of these effects are unknown. The purpose of the rodent studies described here was to evaluate a commonly prescribed APD, risperidone, for chronic effects on recognition memory, brain-derived neurotrophic factor (BDNF), its precursor proBDNF, as well as relevant downstream signaling molecules that are known to influence neuronal plasticity and cognition. Multiple cohorts of adult rats were treated with risperidone (2.5 mg/kg/day) or vehicle (dilute acetic acid solution) in their drinking water for 30 or 90 days. Subjects were then evaluated for drug effects on recognition memory in a spontaneous novel object recognition task and protein levels of BDNF-related signaling molecules in the hippocampus and prefrontal cortex. The results indicated that depending on the treatment period, a therapeutically relevant daily dose of risperidone impaired recognition memory and increased the proBDNF/BDNF ratio in the hippocampus and prefrontal cortex. Risperidone treatment also led to a decrease in Akt and CREB phosphorylation in the prefrontal cortex. These results indicate that chronic treatment with a commonly prescribed APD, risperidone, has the potential to adversely affect recognition memory and neurotrophin-related signaling molecules that support synaptic plasticity and cognitive function.


Subject(s)
Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Recognition, Psychology/drug effects , Risperidone/administration & dosage , Risperidone/pharmacology , Signal Transduction/drug effects , Administration, Oral , Animals , Antipsychotic Agents/blood , Behavior, Animal/drug effects , Catalepsy/chemically induced , Catalepsy/diagnosis , Cognition/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Hippocampus/metabolism , Male , Nerve Growth Factors/metabolism , Neuronal Plasticity/drug effects , Phosphorylation/drug effects , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Risperidone/blood
8.
J Neurosci ; 39(21): 4179-4192, 2019 05 22.
Article in English | MEDLINE | ID: mdl-30886019

ABSTRACT

Obesity and insulin resistance elicit blood-brain barrier (BBB) breakdown in humans and animal models, but the relative contributions of the two pathologies remain poorly understood. These studies initially addressed the temporal progression of cerebrovascular dysfunction relative to dietary obesity or diet-induced insulin resistance in male mice. Obesity increased BBB permeability to the low molecular weight fluorophore sodium fluorescein (NaFl), whereas diet-induced insulin resistance increased permeability to both NaFl and Evans blue, which forms a high molecular weight complex with serum albumin. Serial section transmission electron microscopy analysis of hippocampal capillaries revealed that diabetes promotes involution of tight junctions, fenestration of endothelial cells, and pericyte regression. Chronic activation of adenosine receptor 2a (Adora2a) erodes tight junctions between endothelial cells of the cerebral vasculature in other models of chronic neuropathology, and we observed that acute Adora2a antagonism normalized BBB permeability in wild-type mice with diet-induced insulin resistance. Experiments in mice with inducible deletion of Adora2a in endothelial cells revealed protection against BBB breakdown with diet-induced insulin resistance, despite comparable metabolic dysfunction relative to nontransgenic littermates. Protection against BBB breakdown was associated with decreased vascular inflammation, recovery of hippocampal synaptic plasticity, and restoration of hippocampus-dependent memory. These findings indicate that Adora2a-mediated signaling in vascular endothelial cells disrupts the BBB in dietary obesity, and implicate cerebrovascular dysfunction as the underlying mechanism for deficits in synaptic plasticity and cognition with obesity and insulin resistance.SIGNIFICANCE STATEMENT The blood-brain barrier (BBB) restricts the entry of circulating factors into the brain, but obesity promotes BBB breakdown in humans and animal models. We used transgenic mice with resistance to BBB breakdown to investigate the role of neurovascular dysfunction in high-fat diet (HFD)-induced cognitive impairment. Transgenic mice with inducible ablation of Adora2a in endothelial cells were protected against BBB breakdown on HFD, despite comparable metabolic impairments relative to normal mice. Transgenic mice were also resistant to HFD-induced cognitive dysfunction and were protected against deficits in hippocampal synaptic plasticity. These findings indicate that Adora2a-mediated signaling in endothelial cells mediates obesity-induced BBB breakdown, and implicate cerebrovascular dysfunction as the mechanism for deficits in synaptic plasticity and cognition with obesity and diabetes.


Subject(s)
Capillary Permeability/physiology , Cognitive Dysfunction/metabolism , Insulin Resistance/physiology , Receptor, Adenosine A2A/metabolism , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Cognitive Dysfunction/pathology , Diet, High-Fat/adverse effects , Endothelial Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/metabolism
9.
Toxicology ; 406-407: 92-103, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29894704

ABSTRACT

Organophosphates (OPs) are found in hundreds of valuable agricultural, industrial, and commercial compounds; however, they have also been associated with a variety of harmful effects in humans. The acute toxicity of OPs is attributed to the inhibition of the enzyme acetylcholinesterase (AChE); however, this mechanism may not account for all of the deleterious neurologic effects of OPs, especially at doses that produce no overt signs of acute toxicity. In this study, the effects of two weeks of daily subcutaneous exposure to the OP-nerve agent diisopropylfluorophosphate (DFP) in doses ranging from 0.125-0.500 mg/kg on whole brain volume, white matter, and gray matter integrity were evaluated in post mortem tissues using histology and magnetic resonance imaging (MRI) methods. The effects of DFP on axonal transport in the brains of living rats were evaluated using a manganese-enhanced MRI (MEMRI) method. DFP was associated with dose-dependent impairments in red blood cell and brain AChE (down to 29 and 18% of control, respectively at the highest dose), 24 h after the last injection. However, there were no visible signs of cholinergic toxicity noted in any portion of the study. Moreover, histological and MRI analysis of post mortem brains did not reveal any pronounced alterations of whole brain, white matter, or gray matter volumes associated with DFP. Electron microscopy did reveal a DFP-related increase in structural disruptions of myelinated axons (i.e., decompactions) in the fimbria region on the corpus callosum. MEMRI indicated that DFP was also associated with dose-dependent decreases in axonal transport in the brains of living rats, an effect that was also present after a 30-day (DFP-free) washout period, when AChE was not significantly inhibited. These results indicate that repeated exposures to the nerve agent, DFP at doses that are below the threshold for acute toxicity, can result in alterations in myelin structure and persistent decreases in axonal transport in the rodent brain. These observations could explain some of the long-term neurological deficits that have been observed in humans who have been repeatedly exposed to OPs.


Subject(s)
Axonal Transport/drug effects , Axons/drug effects , Brain/drug effects , Cholinesterase Inhibitors/toxicity , Isoflurophate/toxicity , Nerve Fibers, Myelinated/drug effects , Animals , Axonal Transport/physiology , Axons/pathology , Axons/ultrastructure , Brain/pathology , Brain/ultrastructure , Cholinesterase Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Isoflurophate/administration & dosage , Male , Nerve Fibers, Myelinated/pathology , Nerve Fibers, Myelinated/ultrastructure , Rats , Rats, Wistar
10.
Biochem Pharmacol ; 151: 180-187, 2018 05.
Article in English | MEDLINE | ID: mdl-29175423

ABSTRACT

While impairments of cognition in schizophrenia have the greatest impact on long-term functional outcome, the currently prescribed treatments, antipsychotic drugs (APDs), do not effectively improve cognition. Moreover, while more than 20 years have been devoted to the development of new drugs to treat cognitive deficits in schizophrenia, none have been approved to date. One area that has not been given proper attention at the preclinical or clinical stage of drug development is the chronic medication history of the test subject. Hence, very little is known about how chronic treatment with drugs that affect multiple receptors like APDs influence the response to a potential pro-cognitive agent. Therefore, the purpose of this study was to evaluate the α7 nicotinic acetylcholine receptor (α7 nAChR) partial agonist, tropisetron in rats chronically treated with APDs with distinct pharmacological profiles. Rats were treated orally with either risperidone (2.5 mg/kg/day) or quetiapine (25.0 mg/kg/day) for 30 or 90 days and then an acute injection of vehicle or tropisetron (3.0 mg/kg) was administered before training in a novel object recognition (NOR) task. After a 48 h delay (when recollection of the familiar object was impaired in vehicle-treated animals) neither 30 nor 90 days of risperidone or quetiapine treatment improved NOR performance. In contrast, tropisetron markedly improved NOR performance in rats treated with either APD for 30 or 90 days. These animal data reinforce the argument that two commonly prescribed APDs are not pro-cognitive agents and that α7 nAChR ligands like tropisetron have potential as adjunctive treatments in schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Quetiapine Fumarate/pharmacology , Recognition, Psychology/drug effects , Risperidone/pharmacology , Tropisetron/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Drug Discovery , Drug Evaluation, Preclinical , Drug Partial Agonism , Male , Quetiapine Fumarate/administration & dosage , Quetiapine Fumarate/blood , Rats, Wistar , Risperidone/administration & dosage , Risperidone/blood , Tropisetron/administration & dosage , Tropisetron/blood
11.
Neurotoxicology ; 62: 111-123, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28600141

ABSTRACT

Chlorpyrifos (CPF) is an extensively used organophosphorus pesticide that has recently come under increasing scrutiny due to environmental health concerns particularly its association with neurodevelopmental defects. While the insecticidal actions and acute toxicity of CPF are attributed to its oxon metabolite (CPO) which potently inhibits the cholinergic enzyme acetylcholinesterase (AChE), there is significant evidence that CPF, CPO, and other organophosphates may affect a variety of neuronal targets and processes that are not directly related to AChE. Previously, in adult rat sciatic nerves ex vivo and postnatal neurons from rats in vitro we observed that CPF and CPO impaired the movements of vesicles and mitochondria in axons. Here, in embryonic neurons from rats in culture, we evaluated 24h exposures to CPF and CPO across picomolar to micromolar concentrations for effects on fast axonal transport of membrane bound organelles (MBOs) that contained the amyloid precursor protein (APP) tagged with the fluorescent marker, Dendra2 (APPDendra2). The most notable observations of this study were concentration-dependent decreases in the velocity and percentage of MBOs moving in the anterograde direction, an increase in the number of stationary MBOs, and an increased frequency of pauses associated with both CPF and CPO. These effects occurred at concentrations that did not significantly inhibit AChE activity, they were not blocked by cholinergic receptor antagonists, and they were not associated with compromised cell viability. These effects of CPF and CPO may be significant given the importance of axonal transport to neuronal development as well the function of fully developed neurons.


Subject(s)
Axonal Transport/drug effects , Chlorpyrifos/analogs & derivatives , Chlorpyrifos/pharmacology , Cholinesterase Inhibitors/pharmacology , Neurons/drug effects , Organelles/metabolism , Acetylcholinesterase/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Atropine/pharmacology , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Doublecortin Domain Proteins , Embryo, Mammalian , Ganglionic Blockers/pharmacology , L-Lactate Dehydrogenase/metabolism , Mecamylamine/pharmacology , Microtubule-Associated Proteins/metabolism , Muscarinic Antagonists/pharmacology , Neuropeptides/metabolism , Organelles/drug effects , Rats , Rats, Sprague-Dawley
12.
J Pharmacol Exp Ther ; 356(3): 645-55, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26718240

ABSTRACT

The extensive use of organophosphates (OPs) is an ongoing environmental health concern due to multiple reports of OP-related neurologic abnormalities. The mechanism of the acute toxicity of OPs has been attributed to inhibition of acetylcholinesterase (AChE), but there is growing evidence that this may not account for all the long-term neurotoxic effects of OPs. In previous experiments (using ex vivo and in vitro model systems) we observed that the insecticide OP chlorpyrifos impaired the movements of vesicles and mitochondria in axons. Here, using a time-lapse imaging technique, we evaluated the OP-nerve agent diisopropylfluorophosphate (DFP) across a wide range of concentrations (subnanomolar to micromolar) for effects on fast axonal transport of membrane-bound organelles (MBOs) that contain the amyloid precursor protein (APP) tagged with the fluorescent marker Dendra2 (APPDendra2). Both 1 and 24 hours of exposure to DFP and a positive control compound, colchicine, resulted in a decrease in the velocity of anterograde and retrograde movements of MBOs and an increase in the number of stationary MBOs. These effects occurred at picomolar (100 pM) to low nanomolar (0.1 nM) concentrations that were not associated with compromised cell viability or cytoskeletal damage. Moreover, the effects of DFP on axonal transport occurred at concentrations that did not inhibit AChE activity, and they were not blocked by cholinergic receptor antagonists. Given the fundamental importance of axonal transport to neuronal function, these observations may explain some of the long-term neurologic deficits that have been observed in humans who have been exposed to OPs.


Subject(s)
Axons/drug effects , Cell Membrane/drug effects , Cerebral Cortex/drug effects , Isoflurophate/toxicity , Organelles/drug effects , Animals , Axons/metabolism , Biological Transport/drug effects , Biological Transport/physiology , Cell Membrane/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cholinesterase Inhibitors/toxicity , Female , Organelles/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley
13.
Article in English | MEDLINE | ID: mdl-26313131

ABSTRACT

A sensitive method to simultaneously quantitate quetiapine and norquetiapine in rat plasma and brain tissue was developed using a one-step liquid-liquid extraction for sample preparation and LC-MS/MS for detection. The method provided a linear range of 1.0-500.0ng/mL for each analyte in plasma and 3.0-1500.0ng/g in brain tissue. The method was validated with precision within 15% relative standard deviation (RSD), accuracy within 15% relative error (RE), matrix effects within 10% and a consistent recovery. This method has been successfully applied in a preclinical study of quetiapine and norquetiapine to simultaneously determine their concentrations in rat plasma and brain tissue.


Subject(s)
Chromatography, High Pressure Liquid/methods , Quetiapine Fumarate/blood , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Limit of Detection , Male , Rats , Rats, Wistar , Reproducibility of Results
14.
Biochem Pharmacol ; 97(4): 388-398, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26231940

ABSTRACT

The challenges associated with developing more effective treatments for neurologic and psychiatric illness such as Alzheimer's disease and schizophrenia are considerable. Both the symptoms and the pathophysiology of these conditions are complex and poorly understood and the clinical presentations across different patients can be very heterogeneous. Moreover, it has become apparent that the reductionist approach to drug discovery for these illnesses that has dominated the field for decades (i.e., the development of highly selective compounds or other treatment modalities focused on a very specific pathophysiologic target) has not been widely successful. Accordingly, a variety of new strategies have emerged including the development of "multitarget-directed ligands" (MTDLs), the development and/or identification of compounds that exhibit "multifunctional" activity (e.g., pro-cognitive plus neuroprotective, pro-cognitive plus antipsychotic activity), "repurposing" strategies for existing compounds that have other clinical indications, and novel "adjunctive" treatment strategies that might enhance the efficacy of the currently available treatments. Interestingly, a variety of ligands at nicotinic acetylcholine receptors (nAChRs) appear to have the potential to fulfill one or more of these desirable properties (i.e., multifunctional, repurposing, or adjunctive treatment potential). The purpose of this review (while not all-inclusive) is to provide an overview of a variety of nAChR ligands that demonstrate potential in these categories, particularly, "multifunctional" properties. Due to their densities in the mammalian brain and the amount of literature available, the review will focus on ligands of the high affinity α4ß2 nAChR and the low affinity α7 nAChR.


Subject(s)
Cholinergic Agents/therapeutic use , Mental Disorders/drug therapy , Receptors, Nicotinic/metabolism , Drug Discovery , Ligands
15.
Neurotoxicology ; 47: 17-26, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25614231

ABSTRACT

The toxicity of the class of chemicals known as the organophosphates (OP) is most commonly attributed to the inhibition of the enzyme acetylcholinesterase. However, there is significant evidence that this mechanism may not account for all of the deleterious neurologic and neurobehavioral symptoms of OP exposure, especially those associated with levels that produce no overt signs of acute toxicity. In the study described here we evaluated the effects of the commonly used OP-pesticide, chlorpyrifos (CPF) on axonal transport in the brains of living rats using manganese (Mn(2+))-enhanced magnetic resonance imaging (MEMRI) of the optic nerve (ON) projections from the retina to the superior colliculus (SC). T1-weighted MEMRI scans were evaluated at 6 and 24h after intravitreal injection of Mn(2+). As a positive control for axonal transport deficits, initial studies were conducted with the tropolone alkaloid colchicine administered by intravitreal injection. In subsequent studies both single and repeated exposures to CPF were evaluated for effects on axonal transport using MEMRI. As expected, intravitreal injection of colchicine (2.5µg) produced a robust decrease in transport of Mn(2+) along the optic nerve (ON) and to the superior colliculus (SC) (as indicated by the reduced MEMRI contrast). A single subcutaneous (s.c.) injection of CPF (18.0mg/kg) was not associated with significant alterations in the transport of Mn(2+). Conversely, 14-days of repeated s.c. exposure to CPF (18.0mg/kg/day) was associated with decreased transport of Mn(2+) along the ONs and to the SC, an effect that was also present after a 30-day (CPF-free) washout period. These results indicate that repeated exposures to a commonly used pesticide, CPF can result in persistent alterations in axonal transport in the living mammalian brain. Given the fundamental importance of axonal transport to neuronal function, these observations may (at least in part) explain some of the long term neurological deficits that have been observed in humans who have been repeatedly exposed to doses of OPs not associated with acute toxicity.


Subject(s)
Axonal Transport/drug effects , Brain/drug effects , Chlorpyrifos/toxicity , Insecticides/toxicity , Acetylcholinesterase/analysis , Animals , Brain/enzymology , Brain/metabolism , Contrast Media , Magnetic Resonance Imaging , Male , Manganese , Optic Nerve/drug effects , Optic Nerve/enzymology , Optic Nerve/metabolism , Rats , Rats, Wistar , Visual Pathways/drug effects , Visual Pathways/enzymology , Visual Pathways/metabolism
16.
J Physiol ; 592(15): 3201-14, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24879866

ABSTRACT

There is much interest in α7 nicotinic acetylcholine receptors (nAChRs) in CNS function since they are found throughout peripheral tissues as well as being highly expressed in brain regions implicated in attention, learning and memory. As such, the role of these receptors in many aspects of CNS function and disease is being actively investigated. To date, only one null mouse model (A7KO) is available which is non-conditional and constitutive. Since α7 nAChRs are present on neurons and glia (including astrocytes), as well as being developmentally regulated, there is an unmet need for the technical capability to control α7 nAChR gene expression. Therefore we have generated mice in which the fourth exon of the α7 nAChR gene (Chrna7) is flanked by loxP sites (B6-Chrna7(LBDEx4007Ehs)) which we refer to as floxed α7 nAChR conditional knockout or α7nAChR(flox). We validated the chosen approach by mating α7nAChR(flox) with mice expressing Cre recombinase driven by the glial acidic fibrillary protein (GFAP)-Cre promoter (GFAP-A7KO) to test whether α7nAChR(flox), GFAP-A7KO and appropriate littermate controls performed equally in our standard Rodent In Vivo Assessment Core battery to assess general health, locomotion, emotional and cognitive behaviours. Neither α7nAChR(flox) nor GFAP-A7KO exhibited significant differences from littermate controls in any of the baseline behavioural assessments we conducted, similar to the 'first generation' non-conditional A7KO mice. We also determined that α7 nAChR binding sites were absent on GFAP-positive astrocytes in hippocampal slices obtained from GFAP-A7KO offspring from α7nAChR(flox) and GFAP-Cre crosses. Finally, we validated that Cre recombinase (Cre)-mediated excision led to functional, cell- and tissue-specific loss of α7 nAChRs by demonstrating that choline-induced α7 nAChR currents were present in Cre-negative, but not synapsin promoter-driven Cre-positive, CA1 pyramidal neurons. Additionally, electrophysiological characterization of α7 nAChR-mediated current traces was similar in terms of amplitude and time constants of decay (during desensitization) for the α7nAChR(flox) and wild-type (WT) mice. Thus, we have in vivo and in vitro evidence that the Chrna7 exon 4 targeting strategy does not alter behavioural, cognitive, or electrophysiological properties compared to WT and that Cre-mediated excision is an effective approach to delete α7 nAChR expression in a cell-specific manner.


Subject(s)
Gene Targeting/methods , Neurons/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , Action Potentials , Animals , Astrocytes/metabolism , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Exons , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Integrases/genetics , Integrases/metabolism , Maze Learning , Mice , Mice, Knockout , Phenotype , Promoter Regions, Genetic , alpha7 Nicotinic Acetylcholine Receptor/metabolism
17.
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
18.
Neurobiol Aging ; 34(9): 2081-90, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23582659

ABSTRACT

Amyloid-ß peptide (Aß) fragment misfolding may play a crucial role in the progression of Alzheimer's disease (AD) pathophysiology as well as epigenetic mechanisms at the DNA and histone level. We hypothesized that histone H3 homeostasis is disrupted in association with the appearance of soluble Aß at an early stage in AD progression. We identified, localized, and compared histone H3 modifications in multiple model systems (neural-like SH-SY5Y, primary neurons, Tg2576 mice, and AD neocortex), and narrowed our focus to investigate 3 key motifs associated with regulating transcriptional activation and inhibition: acetylated lysine 14, phosphorylated serine 10 and dimethylated lysine 9. Our results in vitro and in vivo indicate that multimeric soluble Aß may be a potent signaling molecule indirectly modulating the transcriptional activity of DNA by modulating histone H3 homeostasis. These findings reveal potential loci of transcriptional disruption relevant to AD. Identifying genes that undergo significant epigenetic alterations in response to Aß could aid in the understanding of the pathogenesis of AD, as well as suggesting possible new treatment strategies.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/physiology , Histones/metabolism , Homeostasis/genetics , Neocortex/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Animals , Cells, Cultured , DNA/genetics , Epigenesis, Genetic/genetics , Female , Humans , Male , Mice , Mice, Transgenic , Molecular Targeted Therapy , Neuroblastoma/metabolism , Neurons/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Solubility , Transcriptional Activation
19.
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
20.
Curr Drug Targets ; 13(5): 613-22, 2012 May.
Article in English | MEDLINE | ID: mdl-22300028

ABSTRACT

One of the early signs of Alzheimer's disease is the impairment in hippocampus-based episodic memory function, which is improved through the enhancement of cholinergic transmission. Several studies suggest that α7 nicotinic receptor (nAChR) activation represents a useful therapeutic strategy for the cognitive impairments associated with early Alzheimer's disease as the α7 subtype of nicotinic acetylcholine receptors are expressed by basal forebrain cholinergic projection neurons as well as by their targets in the hippocampus. The current model for the cholinergic deficit in Alzheimer's disease posits that inappropriate accumulation of misfolded oligomeric aggregates of ß-amyloid peptide leads to the dysfunction of the signaling mechanisms that support the cholinergic phenotype; this is manifested as an altered function of nicotinic acetylcholine receptors and the nerve-growth factor trophic support system that results in the loss of cholinergic markers and eventually cholinergic neurons from the basal forebrain cholinergic system. A view was confounded by the fact that α7 nAChRs and ß-amyloid peptides have been shown to interact in vitro and in vivo, including human post-mortem AD brain. This review will begin with a brief overview of the basal forebrain cholinergic system, followed by a discussion of the current knowledge of the cholinergic deficit in Alzheimer's disease, then a summary of the cholinergic phenotype observed in transgenic Alzheimer's disease mouse models. We will also present our recent findings that support our hypothesis that the α7 nicotinic acetylcholine receptor performs both the neurotrophic and neuroprotective roles in the maintenance of the cholinergic phenotype and discusses potential mechanisms and implications for Alzheimer's disease therapy.


Subject(s)
Alzheimer Disease/physiopathology , Cognition Disorders/physiopathology , Receptors, Nicotinic/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cholinergic Neurons/metabolism , Cognition Disorders/etiology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Transgenic , Prosencephalon/metabolism , Prosencephalon/pathology , alpha7 Nicotinic Acetylcholine Receptor
SELECTION OF CITATIONS
SEARCH DETAIL