Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 183
Filter
1.
eNeuro ; 5(1)2018.
Article in English | MEDLINE | ID: mdl-29516036

ABSTRACT

Stress is a potent etiological factor in the onset of major depressive disorder and posttraumatic stress disorder (PTSD). Therefore, significant efforts have been made to identify factors that produce resilience to the outcomes of a later stressor, in hopes of preventing untoward clinical outcomes. The NMDA receptor antagonist ketamine has recently emerged as a prophylactic capable of preventing neurochemical and behavioral outcomes of a future stressor. Despite promising results of preclinical studies performed in male rats, the effects of proactive ketamine in female rats remains unknown. This is alarming given that stress-related disorders affect females at nearly twice the rate of males. Here we explore the prophylactic effects of ketamine on stress-induced anxiety-like behavior and the neural circuit-level processes that mediate these effects in female rats. Ketamine given one week prior to an uncontrollable stressor (inescapable tailshock; IS) reduced typical stress-induced activation of the serotonergic (5-HT) dorsal raphe nucleus (DRN) and eliminated DRN-dependent juvenile social exploration (JSE) deficits 24 h after the stressor. Proactive ketamine altered prelimbic cortex (PL) neural ensembles so that a later experience with IS now activated these cells, which it ordinarily would not. Ketamine acutely activated a PL to DRN (PL-DRN) circuit and inhibition of this circuit with Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) at the time of IS one week later prevented stress prophylaxis, suggesting that persistent changes in PL-DRN circuit activity are responsible, at least in part, for mediating long-term effects associated with ketamine.


Subject(s)
Excitatory Amino Acid Antagonists/administration & dosage , Ketamine/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Resilience, Psychological/drug effects , Stress, Psychological/prevention & control , Animals , Anxiety/prevention & control , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/physiology , Electroshock , Female , Neural Pathways/drug effects , Neural Pathways/physiology , Rats, Sprague-Dawley , Serotonin/physiology
2.
Neuroscience ; 309: 84-99, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-25772789

ABSTRACT

A consequence of normal aging is a greater susceptibility to memory impairments following an immune challenge such as infection, surgery, or traumatic brain injury. The neuroinflammatory response, produced by these challenges results in increased and prolonged production of pro-inflammatory cytokines in the otherwise healthy aged brain. Here we discuss the mechanisms by which long-lasting elevations in pro-inflammatory cytokines in the hippocampus produce memory impairments. Sensitized microglia are a primary source of this exaggerated neuroinflammatory response and appear to be a hallmark of the normal aging brain. We review the current understanding of the causes and effects of normal aging-induced microglial sensitization, including dysregulations of the neuroendocrine system, potentiation of neuroinflammatory responses following an immune challenge, and the impairment of memories. We end with a discussion of therapeutic approaches to prevent these deleterious effects.


Subject(s)
Aging/immunology , Hippocampus/immunology , Aging/drug effects , Aging/psychology , Animals , Hippocampus/drug effects , Humans , Memory/drug effects , Memory/physiology , Microglia/drug effects , Microglia/immunology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology
3.
Mol Psychiatry ; 20(12): 1525-37, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25644383

ABSTRACT

The initial reinforcing properties of drugs of abuse, such as cocaine, are largely attributed to their ability to activate the mesolimbic dopamine system. Resulting increases in extracellular dopamine in the nucleus accumbens (NAc) are traditionally thought to result from cocaine's ability to block dopamine transporters (DATs). Here we demonstrate that cocaine also interacts with the immunosurveillance receptor complex, Toll-like receptor 4 (TLR4), on microglial cells to initiate central innate immune signaling. Disruption of cocaine signaling at TLR4 suppresses cocaine-induced extracellular dopamine in the NAc, as well as cocaine conditioned place preference and cocaine self-administration. These results provide a novel understanding of the neurobiological mechanisms underlying cocaine reward/reinforcement that includes a critical role for central immune signaling, and offer a new target for medication development for cocaine abuse treatment.


Subject(s)
Cocaine/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Cocaine/administration & dosage , Dopamine Plasma Membrane Transport Proteins/metabolism , Interleukin-1beta/genetics , Male , Mice , Mice, Inbred C3H , Mutation , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neuroglia/drug effects , Neuroglia/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reinforcement, Psychology , Reward , Self Administration , Toll-Like Receptor 4/genetics , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
4.
Neuropharmacology ; 96(Pt A): 55-69, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25446571

ABSTRACT

Neuroimmune diseases have diverse symptoms and etiologies but all involve pathological inflammation that affects normal central nervous system signaling. Critically, many neuroimmune diseases also involve insufficient signaling/bioavailability of interleukin-10 (IL-10). IL-10 is a potent anti-inflammatory cytokine released by immune cells and glia, which drives the regulation of a variety of anti-inflammatory processes. This review will focus on the signaling pathways and function of IL-10, the current evidence for insufficiencies in IL-10 signaling/bioavailability in neuroimmune diseases, as well as the implications for IL-10-based therapies to treating such problems. We will review in detail four pathologies as examples of the common etiologies of such disease states, namely neuropathic pain (nerve trauma), osteoarthritis (peripheral inflammation), Parkinson's disease (neurodegeneration), and multiple sclerosis (autoimmune). A number of methods to increase IL-10 have been developed (e.g. protein administration, viral vectors, naked plasmid DNA, plasmid DNA packaged in polymers to enhance their uptake into target cells, and adenosine 2A agonists), which will also be discussed. In general, IL-10-based therapies have been effective at treating both the symptoms and pathology associated with various neuroimmune diseases, with more sophisticated gene therapy-based methods producing sustained therapeutic effects lasting for several months following a single injection. These exciting results have resulted in IL-10-targeted therapeutics being positioned for upcoming clinical trials for treating neuroimmune diseases, including neuropathic pain. Although further research is necessary to determine the full range of effects associated with IL-10-based therapy, evidence suggests IL-10 may be an invaluable target for the treatment of neuroimmune disease. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.


Subject(s)
Interleukin-10/therapeutic use , Multiple Sclerosis/drug therapy , Neuralgia/drug therapy , Neuroimmunomodulation , Osteoarthritis/drug therapy , Parkinson Disease/drug therapy , Signal Transduction , Animals , Drug Delivery Systems , Female , Humans , Inflammation/immunology , Interleukin-10/immunology , Male , Multiple Sclerosis/immunology , Neuralgia/immunology , Osteoarthritis/immunology , Parkinson Disease/immunology
5.
Neuroscience ; 280: 299-317, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25241065

ABSTRACT

CNS immune signaling contributes to deleterious opioid effects including hyperalgesia, tolerance, reward, and dependence/withdrawal. Such effects are mediated by opioid signaling at toll-like receptor 4 (TLR4), presumptively of glial origin. Whether CNS endothelial cells express TLR4 is controversial. If so, they would be well positioned for activation by blood-borne opioids, contributing to opioid-induced pro-inflammatory responses. These studies examined adult primary rat CNS endothelial cell responses to (-)-morphine or its mu opioid receptor (MOR)-inactive metabolite morphine-3-glucuronide (M3G), both known TLR4 agonists. We demonstrate that adult rat CNS endothelial cells express functional TLR4. M3G activated nuclear factor kappaB (NF-κB), increased tumor necrosis factor-α (TNFα) and cyclooxygenase-2 (COX2) mRNAs, and released prostaglandin E2 (PGE2) from these cells. (-)-Morphine-induced upregulation of TNFα mRNA and PGE2 release were unmasked by pre-treatment with nalmefene, a MOR antagonist without TLR4 activity (unlike CTAP, shown to have both MOR- and TLR4-activity), suggestive of an interplay between MOR and TLR4 co-activation by (-)-morphine. In support, MOR-dependent Protein Kinase A (PKA) opposed TLR4 signaling, as PKA inhibition (H-89) also unmasked (-)-morphine-induced TNFα and COX2 mRNA upregulation. Intrathecal injection of CNS endothelial cells, stimulated in vitro with M3G, produced TLR4-dependent tactile allodynia. Further, cortical suffusion with M3G in vivo induced TLR4-dependent vasodilation. Finally, endothelial cell TLR4 activation by lipopolysaccharide and/or M3G was blocked by the glial inhibitors AV1013 and propentofylline, demonstrating endothelial cells as a new target of such drugs. These data indicate that (-)-morphine and M3G can activate CNS endothelial cells via TLR4, inducing proinflammatory, biochemical, morphological, and behavioral sequelae. CNS endothelial cells may have previously unanticipated roles in opioid-induced effects, in phenomena blocked by presumptive glial inhibitors, as well as TLR4-mediated phenomena more broadly.


Subject(s)
Central Nervous System/drug effects , Endothelial Cells/drug effects , Morphine Derivatives/pharmacology , Morphine/pharmacology , Narcotics/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Central Nervous System/physiology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Endothelial Cells/physiology , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Male , NF-kappa B/metabolism , Neuroglia/drug effects , Neuroglia/physiology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , Toll-Like Receptor 4/agonists , Tumor Necrosis Factor-alpha/metabolism , Vasodilation/drug effects , Vasodilation/physiology
6.
Mol Psychiatry ; 19(6): 699-709, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24342992

ABSTRACT

The limited success in understanding the pathophysiology of major depression may result from excessive focus on the dysfunctioning of neurons, as compared with other types of brain cells. Therefore, we examined the role of dynamic alterations in microglia activation status in the development of chronic unpredictable stress (CUS)-induced depressive-like condition in rodents. We report that following an initial period (2-3 days) of stress-induced microglial proliferation and activation, some microglia underwent apoptosis, leading to reductions in their numbers within the hippocampus, but not in other brain regions, following 5 weeks of CUS exposure. At that time, microglia displayed reduced expression of activation markers as well as dystrophic morphology. Blockade of the initial stress-induced microglial activation by minocycline or by transgenic interleukin-1 receptor antagonist overexpression rescued the subsequent microglial apoptosis and decline, as well as the CUS-induced depressive-like behavior and suppressed neurogenesis. Similarly, the antidepressant drug imipramine blocked the initial stress-induced microglial activation as well as the CUS-induced microglial decline and depressive-like behavior. Treatment of CUS-exposed mice with either endotoxin, macrophage colony-stimulating factor or granulocyte-macrophage colony-stimulating factor, all of which stimulated hippocampal microglial proliferation, partially or completely reversed the depressive-like behavior and dramatically increased hippocampal neurogenesis, whereas treatment with imipramine or minocycline had minimal or no anti-depressive effects, respectively, in these mice. These findings provide direct causal evidence that disturbances in microglial functioning has an etiological role in chronic stress-induced depression, suggesting that microglia stimulators could serve as fast-acting anti-depressants in some forms of depressive and stress-related conditions.


Subject(s)
Behavior, Animal/physiology , Brain/physiopathology , Depressive Disorder/physiopathology , Microglia/physiology , Neurogenesis/physiology , Stress, Psychological/physiopathology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Chronic Disease , Depressive Disorder/drug therapy , Depressive Disorder/etiology , Depressive Disorder/pathology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Male , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Neurogenesis/drug effects , Rats , Stress, Psychological/complications , Stress, Psychological/drug therapy , Stress, Psychological/pathology , Uncertainty
7.
J Neurosci ; 32(33): 11187-200, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22895704

ABSTRACT

Opioid action was thought to exert reinforcing effects solely via the initial agonism of opioid receptors. Here, we present evidence for an additional novel contributor to opioid reward: the innate immune pattern-recognition receptor, toll-like receptor 4 (TLR4), and its MyD88-dependent signaling. Blockade of TLR4/MD2 by administration of the nonopioid, unnatural isomer of naloxone, (+)-naloxone (rats), or two independent genetic knock-outs of MyD88-TLR4-dependent signaling (mice), suppressed opioid-induced conditioned place preference. (+)-Naloxone also reduced opioid (remifentanil) self-administration (rats), another commonly used behavioral measure of drug reward. Moreover, pharmacological blockade of morphine-TLR4/MD2 activity potently reduced morphine-induced elevations of extracellular dopamine in rat nucleus accumbens, a region critical for opioid reinforcement. Importantly, opioid-TLR4 actions are not a unidirectional influence on opioid pharmacodynamics, since TLR4(-/-) mice had reduced oxycodone-induced p38 and JNK phosphorylation, while displaying potentiated analgesia. Similar to our recent reports of morphine-TLR4/MD2 binding, here we provide a combination of in silico and biophysical data to support (+)-naloxone and remifentanil binding to TLR4/MD2. Collectively, these data indicate that the actions of opioids at classical opioid receptors, together with their newly identified TLR4/MD2 actions, affect the mesolimbic dopamine system that amplifies opioid-induced elevations in extracellular dopamine levels, therefore possibly explaining altered opioid reward behaviors. Thus, the discovery of TLR4/MD2 recognition of opioids as foreign xenobiotic substances adds to the existing hypothesized neuronal reinforcement mechanisms, identifies a new drug target in TLR4/MD2 for the treatment of addictions, and provides further evidence supporting a role for central proinflammatory immune signaling in drug reward.


Subject(s)
Analgesics, Opioid/administration & dosage , Conditioning, Operant/drug effects , Reinforcement, Psychology , Toll-Like Receptor 4/metabolism , Analgesics, Opioid/blood , Analysis of Variance , Animals , Conditioning, Operant/physiology , Dopamine/metabolism , Dose-Response Relationship, Drug , Drug Administration Routes , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Microdialysis , Mitogen-Activated Protein Kinase 1/metabolism , Models, Molecular , Myeloid Differentiation Factor 88/deficiency , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Pain Threshold/drug effects , Pain Threshold/physiology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Self Administration , Signal Transduction/drug effects , Time Factors , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/deficiency
8.
Neuroscience ; 197: 132-44, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21958863

ABSTRACT

Uncontrollable stress can interfere with instrumental learning and induce anxiety in humans and rodents. While evidence supports a role for serotonin (5-HT) and serotonin 2C receptors (5-HT(2C)R) in the behavioral consequences of uncontrollable stress, the specific sites of action are unknown. These experiments sought to delineate the role of 5-HT and 5-HT(2C)R in the dorsal striatum (DS) and the lateral/basolateral amygdala (BLA) in the expression of stress-induced instrumental escape deficits and exaggerated fear, as these structures are critical to instrumental learning and fear behaviors. Using in vivo microdialysis, we first demonstrated that prior uncontrollable, but not controllable, stress sensitizes extracellular 5-HT in the dorsal striatum, a result that parallels prior work in the BLA. Additionally, rats were implanted with bi-lateral cannula in either the DS or the BLA and exposed to uncontrollable tail shock stress. One day later, rats were injected with 5-HT(2C)R antagonist (SB242084) and fear and instrumental learning behaviors were assessed in a shuttle box. Separately, groups of non-stressed rats received an intra-DS or an intra-BLA injection of the 5-HT(2C)R agonist (CP809101) and behavior was observed. Intra-DS injections of the 5-HT(2C)R antagonist prior to fear/escape tests completely blocked the stress-induced interference with instrumental escape learning; a partial block was observed when injections were in the BLA. Antagonist administration in either region did not influence stress-induced fear behavior. In the absence of prior stress, intra-DS administration of the 5-HT(2C)R agonist was sufficient to interfere with escape behavior without enhancing fear, while intra-BLA administration of the 5-HT(2C)R agonist increased fear behavior but had no effect on escape learning. Results reveal a novel role of the 5-HT(2C)R in the DS in the expression of instrumental escape deficits produced by uncontrollable stress and demonstrate that the involvement of 5-HT(2C)R activation in stress-induced behaviors is regionally specific.


Subject(s)
Conditioning, Operant/physiology , Corpus Striatum/metabolism , Helplessness, Learned , Receptor, Serotonin, 5-HT2C/metabolism , Stress, Psychological/metabolism , Amygdala/metabolism , Animals , Behavior, Animal , Male , Microdialysis , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley
9.
Neuroscience ; 170(1): 138-48, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20600641

ABSTRACT

Control over an aversive experience can greatly impact the organism's response to subsequent stressors. We compared the effects of escapable (ES) and yoked inescapable (IS) electric tail shocks on the hypothalamic-pituitary-adrenal (HPA) axis hormonal (corticosterone and adrenocorticotropic hormone (ACTH)), neural (c-fos mRNA) and behavioral (struggling) response to subsequent restraint. We found that although the HPA axis response during restraint of both previously stressed groups were higher than stress-naïve rats and not different from each other, lack of control over the tailshock experience led to an increase in restraint-induced struggling behavior of the IS rats compared to both stress-naïve and ES rats. Additionally, c-fos expression in the basolateral amygdala was increased selectively in the IS group, and relative c-fos mRNA expression in the basolateral amygdala positively correlated with struggling behavior. Restraint-induced c-fos expression in the medial prefrontal cortex, a brain area critical for mediating some of the differential neurochemical and behavioral effects of ES and IS, was surprisingly similar in both ES and IS groups, lower than that of stress-naïve rats, and did not correlate with struggling behavior. Our findings indicate that basolateral amygdala activity may be connected with the differential effects of ES and IS on subsequent behavioral responses to restraint, without contributing to the concurrent HPA axis hormone response.


Subject(s)
Escape Reaction/physiology , Gene Expression Regulation , Immobilization/physiology , Proto-Oncogene Proteins c-fos/biosynthesis , Stress, Psychological/metabolism , Amygdala , Animals , Cohort Studies , Electric Stimulation/methods , Immobilization/methods , Immobilization/psychology , Male , Proto-Oncogene Proteins c-fos/genetics , Rats , Rats, Sprague-Dawley , Restraint, Physical/methods , Restraint, Physical/physiology , Restraint, Physical/psychology , Stress, Psychological/genetics , Stress, Psychological/psychology
10.
Neuroscience ; 169(4): 1888-900, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20547213

ABSTRACT

Glutamate neurotransmission is highly regulated, largely by glutamate transporters. In the spinal cord, the glutamate transporter GLT-1 is primarily responsible for glutamate clearance. Downregulation of GLT-1 can occur in activated astrocytes, and is associated with increased extracellular glutamate and neuroexcitation. Among other conditions, astrocyte activation occurs following repeated opioids and in models of chronic pain. If GLT-1 downregulation occurs in these states, GLT-1 could be a pharmacological target for improving opioid efficacy and controlling chronic pain. The present studies explored whether daily intrathecal treatment of rats with ceftriaxone, a beta-lactam antibiotic that upregulates GLT-1 expression, could prevent development of hyperalgesia and allodynia following repeated morphine, reverse pain arising from central or peripheral neuropathy, and reduce glial activation in these models. Ceftriaxone pre-treatment attenuated the development of hyperalgesia and allodynia in response to repeated morphine, and prevented associated astrocyte activation. In a model of multiple sclerosis (experimental autoimmune encephalomyelitis; EAE), ceftriaxone reversed tactile allodynia and halted the progression of motor weakness and paralysis. Similarly, ceftriaxone reversed tactile allodynia induced by chronic constriction nerve injury (CCI). EAE and CCI each significantly reduced the expression of membrane-bound, dimerized GLT-1 protein in lumbar spinal cord, an effect normalized by ceftriaxone. Lastly, ceftriaxone normalized CCI- and EAE-induced astrocyte activation in lumbar spinal cord. Together, these data indicate that increasing spinal GLT-1 expression attenuates opioid-induced paradoxical pain, alleviates neuropathic pain, and suppresses associated glial activation. GLT-1 therefore may be a therapeutic target that could improve available treatment options for patients with chronic pain.


Subject(s)
Ceftriaxone/pharmacology , Ceftriaxone/therapeutic use , Excitatory Amino Acid Transporter 2/biosynthesis , Glutamic Acid/metabolism , Pain, Intractable/drug therapy , Spinal Cord/drug effects , Up-Regulation/physiology , Animals , Disease Models, Animal , Male , Pain, Intractable/metabolism , Pain, Intractable/physiopathology , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord/physiopathology , Up-Regulation/drug effects
11.
Neuroscience ; 168(2): 551-63, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20381591

ABSTRACT

Opioids have been discovered to have Toll-like receptor (TLR) activity, beyond actions at classical opioid receptors. This raises the question whether other pharmacotherapies for pain control may also possess TLR activity, contributing to or opposing their clinical effects. We document that tricyclics can alter TLR4 and TLR2 signaling. In silico simulations revealed that several tricyclics docked to the same binding pocket on the TLR accessory protein, myeloid differentiation protein 2 (MD-2), as do opioids. Eight tricyclics were tested for effects on TLR4 signaling in HEK293 cells over-expressing human TLR4. Six exhibited mild (desipramine), moderate (mianserin, cyclobenzaprine, imiprimine, ketotifen) or strong (amitriptyline) TLR4 inhibition, and no TLR4 activation. In contrast, carbamazepine and oxcarbazepine exhibited mild and strong TLR4 activation, respectively, and no TLR4 inhibition. Amitriptyline but not carbamazepine also significantly inhibited TLR2 signaling in a comparable cell line. Live imaging of TLR4 activation in RAW264.7 cells and TLR4-dependent interleukin-1 release from BV-2 microglia revealed that amitriptyline blocked TLR4 signaling. Lastly, tricyclics with no (carbamazepine), moderate (cyclobenzeprine), and strong (amitriptyline) TLR4 inhibition were tested intrathecally (rats) and amitriptyline tested systemically in wildtype and knockout mice (TLR4 or MyD88). While tricyclics had no effect on basal pain responsivity, they potentiated morphine analgesia in rank-order with their potency as TLR4 inhibitors. This occurred in a TLR4/MyD88-dependent manner as no potentiation of morphine analgesia by amitriptyline occurred in these knockout mice. This suggests that TLR2 and TLR4 inhibition, possibly by interactions with MD2, contributes to effects of tricyclics in vivo. These studies provide converging lines of evidence that several tricyclics or their active metabolites may exert their biological actions, in part, via modulation of TLR4 and TLR2 signaling and suggest that inhibition of TLR4 and TLR2 signaling may potentially contribute to the efficacy of tricyclics in treating chronic pain and enhancing the analgesic efficacy of opioids.


Subject(s)
Heterocyclic Compounds, 3-Ring/pharmacology , Lymphocyte Antigen 96/physiology , Toll-Like Receptors/physiology , Analgesics, Opioid/pharmacology , Animals , Cells, Cultured , Humans , Interleukin-1/metabolism , Ligands , Lymphocyte Antigen 96/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Models, Molecular , Morphine/pharmacology , Myeloid Differentiation Factor 88/genetics , Pain Measurement , Protein Binding , Rats , Signal Transduction , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology , Toll-Like Receptors/agonists , Toll-Like Receptors/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
12.
Neuroscience ; 167(3): 880-93, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20178837

ABSTRACT

Opioid-induced glial activation and its proinflammatory consequences have been associated with both reduced acute opioid analgesia and the enhanced development of tolerance, hyperalgesia and allodynia following chronic opioid administration. Intriguingly, recent evidence demonstrates that these effects can result independently from the activation of classical, stereoselective opioid receptors. Here, a structurally disparate range of opioids cause activation of signaling by the innate immune receptor toll like receptor 4 (TLR4), resulting in proinflammatory glial activation. In the present series of studies, we demonstrate that the (+)-isomers of methadone and morphine, which bind with negligible affinity to classical opioid receptors, induced upregulation of proinflammatory cytokine and chemokine production in rat isolated dorsal spinal cord. Chronic intrathecal (+)-methadone produced hyperalgesia and allodynia, which were associated with significantly increased spinal glial activation (TLR4 mRNA and protein) and the expression of multiple chemokines and cytokines. Statistical analysis suggests that a cluster of cytokines and chemokines may contribute to these nociceptive behavioral changes. Acute intrathecal (+)-methadone and (+)-morphine were also found to induce microglial, interleukin-1 and TLR4/myeloid differentiation factor-2 (MD-2) dependent enhancement of pain responsivity. In silico docking analysis demonstrated (+)-naloxone sensitive docking of (+)-methadone and (+)-morphine to human MD-2. Collectively, these data provide the first evidence of the pro-nociceptive consequences of small molecule xenobiotic activation of spinal TLR4 signaling independent of classical opioid receptor involvement.


Subject(s)
Analgesics, Opioid/toxicity , Inflammation Mediators/toxicity , Inflammation/chemically induced , Lymphocyte Antigen 96/drug effects , Spinal Cord/drug effects , Toll-Like Receptor 4/drug effects , Animals , Gliosis/chemically induced , Gliosis/metabolism , Gliosis/physiopathology , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Immunity, Innate/drug effects , Immunity, Innate/physiology , Inflammation/metabolism , Inflammation/physiopathology , Injections, Spinal , Interleukin-1/metabolism , Isomerism , Lymphocyte Antigen 96/metabolism , Male , Methadone/toxicity , Microglia/drug effects , Microglia/metabolism , Morphine/toxicity , Organ Culture Techniques , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord/physiopathology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
13.
Neuroscience ; 165(4): 1031-8, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-19909791

ABSTRACT

Experience with behavioral control over tailshock (escapable shock, ES) has been shown to block the behavioral and neurochemical changes produced by later uncontrollable tail shock (inescapable shock, IS). The present experiments tested, in rats, whether the protective effect of control over tailshock extends beyond reducing the behavioral and neurochemical impact of a subsequent tailshock experience to stressors that are quite different. Social defeat (SD) was chosen as the second stress experience because it has few if any cues in common with tailshock. SD produced shuttlebox escape learning deficits ("learned helplessness") and reduced juvenile social investigation 24 h later, as does IS. IS is notable for inducing a large increase in dorsal raphe nucleus (DRN) serotonergic (5-HT) activity as measured by extracellular levels of 5-HT within the DRN, and SD did so as well. ES occurring 7 days before SD blocked this SD-induced DRN activation, as well as the SD-induced interference with shuttlebox escape and reduction in social investigation. Prior exposure to yoked IS did not reduce the DRN 5-HT activation or later behavioral effects produced by SD, and thus the proactive stress-blunting effects of ES can be attributed to it's controllability. Thus, ES confers a very general protection to the impact of a subsequent stress experience.


Subject(s)
Behavior, Animal/physiology , Helplessness, Learned , Stress, Psychological/physiopathology , Animals , Dominance-Subordination , Electroshock , Escape Reaction/physiology , Exploratory Behavior/physiology , Extracellular Space/metabolism , Learning/physiology , Male , Raphe Nuclei/physiopathology , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Time Factors
14.
Neuroscience ; 165(2): 569-83, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-19833175

ABSTRACT

Morphine-3-glucoronide (M3G) is a major morphine metabolite detected in cerebrospinal fluid of humans receiving systemic morphine. M3G has little-to-no affinity for opioid receptors and induces pain by unknown mechanisms. The pain-enhancing effects of M3G have been proposed to significantly and progressively oppose morphine analgesia as metabolism ensues. We have recently documented that morphine activates toll-like receptor 4 (TLR4), beyond its classical actions on mu-opioid receptors. This suggests that M3G may similarly activate TLR4. This activation could provide a novel mechanism for M3G-mediated pain enhancement, as (a) TLR4 is predominantly expressed by microglia in spinal cord and (b) TLR4 activation releases pain-enhancing substances, including interleukin-1 (IL-1). We present in vitro evidence that M3G activates TLR4, an effect blocked by TLR4 inhibitors, and that M3G activates microglia to produce IL-1. In vivo, intrathecal M3G (0.75 microg) induced potent allodynia and hyperalgesia, blocked or reversed by interleukin-1 receptor antagonist, minocycline (microglial inhibitor), and (+)-and (-)-naloxone. This latter study extends our prior demonstrations that TLR4 signaling is inhibited by naloxone nonstereoselectively. These results with (+)-and (-)-naloxone also demonstrate that the effects cannot be accounted for by actions at classical, stereoselective opioid receptors. Hyperalgesia (allodynia was not tested) and in vitro M3G-induced TLR4 signaling were both blocked by 17-DMAG, an inhibitor of heat shock protein 90 (HSP90) that can contribute to TLR4 signaling. Providing further evidence of proinflammatory activation, M3G upregulated TLR4 and CD11b (microglial/macrophage activation marker) mRNAs in dorsal spinal cord as well as IL-1 protein in the lumbosacral cerebrospinal fluid. Finally, in silico and in vivo data support that the glucuronic acid moiety is capable of inducing TLR4/MD-2 activation and enhanced pain. These data provide the first evidence for a TLR4 and IL-1 mediated component to M3G-induced effects, likely of at least microglial origin.


Subject(s)
Central Nervous System Stimulants/adverse effects , Interleukin-1beta/metabolism , Lymphocyte Antigen 96/metabolism , Morphine Derivatives/adverse effects , Pain/chemically induced , Toll-Like Receptor 4/metabolism , Animals , CD11b Antigen/metabolism , Central Nervous System Stimulants/administration & dosage , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Injections, Spinal , Interleukin-1beta/cerebrospinal fluid , Male , Microglia/drug effects , Microglia/metabolism , Morphine Derivatives/administration & dosage , Pain/drug therapy , Pain/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Toll-Like Receptor 4/antagonists & inhibitors
15.
Neuroscience ; 164(4): 1821-32, 2009 Dec 29.
Article in English | MEDLINE | ID: mdl-19788917

ABSTRACT

Spinal cord microglial toll-like receptor 4 (TLR4) has been implicated in enhancing neuropathic pain and opposing morphine analgesia. The present study was initiated to explore TLR4-mediated pain modulation by intrathecal lipopolysaccharide, a classic TLR4 agonist. However, our initial study revealed that intrathecal lipopolysaccharide failed to induce low-threshold mechanical allodynia in naive rats, suggestive that TLR4 agonism may be insufficient to enhance pain. These studies explore the possibility that a second signal is required; namely, heat shock protein-90 (HSP90). This candidate was chosen for study given its known importance as a regulator of TLR4 signaling. A combination of in vitro TLR4 cell signaling and in vivo behavioral studies of pain modulation suggest that TLR4-enhancement of neuropathic pain and TLR4-suppression of morphine analgesia each likely require HSP90 as a cofactor for the effects observed. In vitro studies revealed that dimethyl sulfoxide (DMSO) enhances HSP90 release, suggestive that this may be a means by which DMSO enhances TLR4 signaling. While 2 and 100 microg lipopolysaccharide intrathecally did not induce mechanical allodynia across the time course tested, co-administration of 1 microg lipopolysaccharide with a drug that enhances HSP90-mediated TLR4 signaling now induced robust allodynia. In support of this allodynia being mediated via a TLR4/HSP90 pathway, it was prevented or reversed by intrathecal co-administration of a HSP90 inhibitor, a TLR4 inhibitor, a microglia/monocyte activation inhibitor (as monocyte-derived cells are the predominant cell type expressing TLR4), and interleukin-1 receptor antagonist (as this proinflammatory cytokine is a downstream consequence of TLR4 activation). Together, these results suggest for the first time that TLR4 activation is necessary but not sufficient to induce spinally mediated pain enhancement. Rather, the data suggest that TLR4-dependent pain phenomena may require contributions by multiple components of the TLR4 receptor complex.


Subject(s)
HSP90 Heat-Shock Proteins/physiology , Pain/physiopathology , Toll-Like Receptor 4/physiology , Analgesics, Opioid/pharmacology , Animals , Benzoquinones/pharmacology , Constriction, Pathologic/complications , Dimethyl Sulfoxide/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/biosynthesis , Injections, Spinal , Interleukin-1/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Lipopolysaccharides/pharmacology , Male , Microglia/physiology , Morphine/pharmacology , Pain/etiology , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/physiopathology , Physical Stimulation , Rats , Rats, Sprague-Dawley , Sciatic Nerve/physiopathology , Signal Transduction , Toll-Like Receptor 4/agonists
16.
Gene Ther ; 16(4): 470-5, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19262611

ABSTRACT

Traditional approaches to treating chronic neuropathic pain largely focus on manipulations directly altering neuronal activity or neuron-to-neuron communication. Recently, however, it has become clear that glial cells (including microglia and astroglia) play a significant role in pain expression in a variety of neuropathic pain models. Multiple aspects of the inflammatory response of glial cells, commonly observed in neuropathic pain conditions, have been implicated in pain expression. Thus, glial cell inflammation has emerged as a potential therapeutic target in neuropathic pain. Our laboratory has been exploring the use of an anti-inflammatory cytokine, interleukin-10 (IL-10), to control glial inflammatory activation thereby controlling neuropathic pain. IL-10 protein delivery is limited by a short half-life and an inability to cross into the central nervous system from the periphery, making a centrally delivered gene therapy approach attractive. We have recently characterized a non-viral gene therapy approach using two injections of naked DNA to achieve long-term (>3 months) control of neuropathic pain in a peripheral nerve injury model. Timing and dose requirements leading to long-term pain control are discussed in this review, as is recent work using microparticle-encapsulated DNA to achieve long-term therapeutic efficacy with a single injection.


Subject(s)
Genetic Therapy/methods , Pain Management , Animals , Chronic Disease , DNA/administration & dosage , Gene Transfer Techniques , Interleukin-10/genetics , Interleukin-10/physiology , Neuroglia/physiology
17.
Brain Behav Immun ; 23(1): 92-100, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18835435

ABSTRACT

Multiple Sclerosis (MS) is an autoimmune inflammatory disease that presents clinically with a range of symptoms including motor, sensory, and cognitive dysfunction as well as demyelination and lesion formation in brain and spinal cord. A variety of animal models of MS have been developed that share many of the pathological hallmarks of MS including motor deficits (ascending paralysis), demyelination and axonal damage of central nervous system (CNS) tissue. In recent years, neuropathic pain has been recognized as a prevalent symptom of MS in a majority of patients. To date, there have been very few investigations into sensory disturbances in animal models of MS. The current work contains the first assessment of hind paw mechanical allodynia (von Frey test) over the course of a relapsing-remitting myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis (MOG-EAE) rat model of MS and establishes the utility of this model in examining autoimmune induced sensory dysfunction. We demonstrate periods of both decreased responsiveness to touch that precedes the onset of hind limb paralysis, and increased responsiveness (allodynia) that occurs during the period of motor deficit amelioration traditionally referred to as symptom remission. Furthermore, we tested the ability of our recently characterized anti-inflammatory IL-10 gene therapy to treat the autoimmune inflammation induced behavioral symptoms and tissue histopathological changes. This therapy is shown here to reverse inflammation induced paralysis, to reduce disease associated reduction in sensitivity to touch, to prevent the onset of allodynia, to reverse disease associated loss of body weight, and to suppress CNS glial activation associated with disease progression in this model.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Therapy/methods , Interleukin-10/physiology , Multiple Sclerosis/therapy , Analysis of Variance , Animals , Body Weight/physiology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Glial Fibrillary Acidic Protein/metabolism , Hindlimb , Immunohistochemistry , Inflammation/immunology , Inflammation/therapy , Interleukin-10/genetics , Male , Motor Activity/physiology , Multiple Sclerosis/chemically induced , Multiple Sclerosis/physiopathology , Myelin Proteins , Myelin-Associated Glycoprotein , Myelin-Oligodendrocyte Glycoprotein , Neuralgia/physiopathology , Neuralgia/therapy , Paralysis/physiopathology , Paralysis/therapy , Rats , Rats, Inbred Strains , Spinal Cord/metabolism , Time Factors
18.
Neuroscience ; 154(4): 1178-86, 2008 Jul 17.
Article in English | MEDLINE | ID: mdl-18515010

ABSTRACT

The degree of behavioral control that an organism has over a stressor determines the behavioral and neurochemical sequelae of the stressor, with the presence of control preventing the typical outcomes that occur when the stressor is uncontrollable (e.g. failure to learn, exaggerated fear, dorsal raphe nucleus (DRN) 5-HT activation). Furthermore, an experience with a controllable stressor blocks the consequences of later uncontrollable stressors ("immunization"). These effects of control have been argued to be mediated by control-induced activation of ventral medial prefrontal cortex (mPFCv) output to the DRN. The experiments that have led to this interpretation have all involved the inactivation of the mPFCv with muscimol, showing that inactivation during the stressor eliminates the stressor-resistance produced by control, with the controllable stressor now acting as if it were uncontrollable. The present experiments in rats employed the opposite strategy, activating the mPFCv during the stressor. mPFCv microinjection of picrotoxin during the stressor eliminated the DRN 5-HT activation that normally occurs during the uncontrollable stressor, as well as the escape learning deficit and exaggerated fear that normally follows uncontrollable stress. Furthermore, mPFCv activation during an initial exposure to an uncontrollable stressor led the uncontrollable stressor to produce behavioral and neurochemical immunization when the subjects were later exposed to an uncontrollable stressor. That is, the conjoint activation of the mPFCv and exposure to an uncontrollable stressor led the uncontrollable stressor to act as if it were controllable. These results provide strong support for the argument that behavioral control produced stress-resistance by activating the mPFCv.


Subject(s)
Behavior, Animal/physiology , Prefrontal Cortex/physiology , Stress, Psychological/metabolism , Animals , Conditioning, Operant , GABA Antagonists/pharmacology , Learning/physiology , Male , Microdialysis , Microinjections , Picrotoxin/pharmacology , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Serotonin/metabolism
19.
Neuroscience ; 150(4): 754-63, 2007 Dec 19.
Article in English | MEDLINE | ID: mdl-18035502

ABSTRACT

The intra-hippocampal administration of interleukin-1beta (IL-1beta) as well as the induction of elevated but physiological levels of IL-1beta within the hippocampus interferes with the formation of long-term memory. There is evidence suggesting that the induction of prostaglandin (PG) formation by IL-1beta is involved in impairments in working and spatial memory following IL-1beta. The present experiments extend these findings by showing that PGs are responsible for memory deficits in contextual fear conditioning that occur following IL-1beta injection into the dorsal hippocampus of Sprague-Dawley rats. Cyclooxygenase (COX) inhibition blocked the disruption in contextual fear conditioning produced by IL-1beta and COX inhibition alone also disrupted contextual memory, suggesting an inverted U-shaped relationship between PG levels and memory. In addition to demonstrating the necessity of PGs in IL-1beta-mediated memory deficits, we also show that PGs injected directly into the dorsal hippocampus are sufficient to impair context memory and significantly reduce post-conditioning levels of BDNF within the hippocampus, suggesting a possible mechanism for the memory-impairing effects of PGs.


Subject(s)
Conditioning, Psychological/drug effects , Fear , Hippocampus/drug effects , Interleukin-1beta/adverse effects , Learning Disabilities/chemically induced , Prostaglandins/metabolism , Animals , Behavior, Animal/drug effects , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/administration & dosage , Gene Expression Regulation/drug effects , Hippocampus/physiology , Learning Disabilities/drug therapy , Learning Disabilities/physiopathology , Male , Naproxen/pharmacology , Rats , Rats, Sprague-Dawley
20.
Neuroscience ; 146(4): 1495-503, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17478046

ABSTRACT

Fear conditioning and fear extinction play key roles in the development and treatment of anxiety-related disorders, yet there is little information concerning experiential variables that modulate these processes. Here we examined the impact of exposure to a stressor in a different environment on subsequent fear conditioning and extinction, and whether the degree of behavioral control that the subject has over the stressor is of importance. Rats received a session of either escapable (controllable) tail shock (ES), yoked inescapable (uncontrollable) tail shock (IS), or control treatment (home cage, HC) 7 days before fear conditioning in which a tone and foot shock were paired. Conditioning was measured 24 h later. In a second experiment rats received ES, IS or HC 24 h after contextual fear conditioning. Extinction then occurred every day beginning 7 days later until a criterion was reached. Spontaneous recovery of fear was assessed 14 days after extinction. IS potentiated fear conditioning when given before fear conditioning, and potentiated fear responding during extinction when given after conditioning. Importantly, ES potently interfered with later fear conditioning, decreased fear responding during fear extinction, and prevented spontaneous recovery of fear. Additionally, we examined if the activation of the ventral medial prefrontal cortex (mPFCv) by ES is critical for the protective effects of ES on later fear conditioning. Inactivation of the mPFCv with muscimol at the time of the initial experience with control prevented ES-induced reductions in later contextual and auditory fear conditioning. Finally, we explored if the protective effects of ES extended to an unconditioned fear stimulus, ferret odor. Unlike conditioned fear, prior ES increased the fear response to ferret odor to the same degree as did IS.


Subject(s)
Conditioning, Classical/physiology , Extinction, Psychological/physiology , Fear , Helplessness, Learned , Stress, Physiological/physiopathology , Acoustic Stimulation/adverse effects , Analysis of Variance , Animals , Association Learning/drug effects , Association Learning/physiology , Behavior, Animal , Conditioning, Classical/drug effects , Electroshock/adverse effects , Escape Reaction/physiology , Extinction, Psychological/drug effects , Freezing Reaction, Cataleptic/drug effects , GABA Agonists/pharmacology , Male , Muscimol/pharmacology , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...