Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 13: 1084108, 2022.
Article in English | MEDLINE | ID: mdl-36588734

ABSTRACT

The reduced antidepressant and antihyperalgesic effects of selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine during maintenance treatment has been reported, but little is known about the molecular mechanism of this phenomenon. In three comorbid pain and depression animal models (genetic predisposition, chronic social stress, arthritis), we showed that the fluoxetine's antidepressant and antihyperalgesic effects were reduced during the maintenance treatment. Fluoxetine exposure induced upregulation of the 5-hydroxytryptamine 1A (5-HT1A) auto-receptor and indoleamine 2,3 dioxygenase 1 (IDO1, a rate-limiting enzyme of tryptophan metabolism) in the brainstem dorsal raphe nucleus (DRN), which shifted the tryptophan metabolism away from the 5-HT biosynthesis. Mechanistically, IDO1 upregulation was downstream to fluoxetine-induced 5-HT1A receptor expression because 1) antagonism of the 5-HT1A receptor with WAY100635 or 5-HT1A receptor knockout blocked the IDO1 upregulation, and 2) inhibition of IDO1 activity did not block the 5-HT1A receptor upregulation following fluoxetine exposure. Importantly, inhibition of either the 5-HT1A receptor or IDO1 activity sustained the fluoxetine's antidepressant and antihyperalgesic effects, indicating that 5-HT1A-mediated IDO1 upregulation in the brainstem DRN contributed to the reduced antidepressant and antihyperalgesic effects of fluoxetine. These results suggest a new strategy to improving the therapeutic efficacy of SSRI during maintenance treatment.

2.
Anesth Analg ; 129(3): 890-895, 2019 09.
Article in English | MEDLINE | ID: mdl-31425234

ABSTRACT

BACKGROUND: As opioid abuse and addiction have developed into a major national health crisis, prescription of opioids for pain management has become more controversial. However, opioids do help some patients by providing pain relief and improving the quality of life. To better understand the addictive properties of opioids under chronic pain conditions, we used a conditioned place preference (CPP) paradigm to examine the rewarding properties of morphine in rats with persistent nociception. METHODS: Spared nerve injury (SNI) model was used to induce persistent nociception in rats. Nociceptive behavior was assessed by von Frey test. CPP test was used to examine the rewarding properties of morphine. RESULTS: Our findings are as follows: (1) SNI rats did not show a difference compared with sham rats in magnitude of morphine-induced CPP 1 day after last morphine injection (2-way analysis of variance; for SNI versus sham, F[1,42] = 0.014, P = .91; and 95% confidence intervals for difference of means, -5.9 [-58 to 46], 0.76 [-51 to 53], and 0.90 [-51 to 53] for 2.5, 5, and 10 mg/kg, respectively); (2) increasing morphine dosage (2.5, 5, and 10 mg/kg) did not further increase the magnitude of CPP in both sham and SNI rats (for dosage: F[2,42] = 0.94, P = .40); and (3) morphine-induced CPP persisted in sham rats but extinguished in SNI rats when tested at 8 days after last morphine injection (for sham versus SNI: Bonferroni correction, P < .006 for both 5 and 10 mg/kg doses; and 95% confidence intervals for difference of means, 80.3 [19.7-141] and 87.0 [26.3-148] for 5 and 10 mg/kg, respectively). CONCLUSIONS: Our data provide new evidence supporting the notion that the brain's reward circuitry changes in the context of persistent pain. This observational study suggests that future investigation into the neurobiology of opioid reward requires consideration of the circumstances in which opioid analgesics are administered.


Subject(s)
Analgesics, Opioid/administration & dosage , Conditioning, Operant/drug effects , Morphine/administration & dosage , Nociception/drug effects , Animals , Conditioning, Operant/physiology , Male , Nociception/physiology , Rats , Rats, Sprague-Dawley
3.
Pain ; 159(8): 1518-1528, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29613911

ABSTRACT

Clinical evidence indicates that cognitive impairment is a common comorbid condition of chronic pain. However, the cellular basis for chronic pain-mediated cognitive impairment remains unclear. We report here that rats exhibited memory deficits after spared nerve injury (SNI). We found that levels of stable microtubule (MT) were increased in the hippocampus of the rats with memory deficits. This increase in stable MT is marked by α-tubulin hyperacetylation. Paclitaxel, a pharmacological MT stabilizer, increased the level of stable MT in the hippocampus and induced learning and memory deficits in normal rats. Furthermore, paclitaxel reduced long-term potentiation in hippocampal slices and increased stable MT (evidenced by α-tubulin hyperacetylation) levels in hippocampal neuronal cells. Intracerebroventricular infusion of nocodazole, an MT destabilizer, ameliorated memory deficits in rats with SNI-induced nociceptive behavior. Expression of HDAC6, an α-tubulin deacetylase, was reduced in the hippocampus in rats with cognitive impairment. These findings indicate that peripheral nerve injury (eg, SNI) affects the MT dynamic equilibrium, which is critical to neuronal structure and synaptic plasticity.


Subject(s)
Cognitive Dysfunction/etiology , Hippocampus/metabolism , Microtubules/metabolism , Neuralgia/complications , Animals , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/psychology , Disease Models, Animal , Hippocampus/drug effects , Learning/drug effects , Long-Term Potentiation/drug effects , Male , Memory/drug effects , Microtubules/drug effects , Neuralgia/metabolism , Neuralgia/psychology , Nocodazole/pharmacology , Paclitaxel/pharmacology , Rats , Rats, Sprague-Dawley , Tubulin Modulators/pharmacology
4.
Neuroimage ; 174: 364-379, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29518566

ABSTRACT

Methamphetamine (meth), and other psychostimulants such as cocaine, present a persistent problem for society with chronic users being highly prone to relapse. We show, in a chronic methamphetamine administration model, that discontinuation of drug for more than a week produces much larger changes in overall meth-induced brain connectivity and cerebral blood volume (CBV) response than changes that occur immediately following meth administration. Areas showing the largest changes were hippocampal, limbic striatum and sensorimotor cortical regions as well as brain stem areas including the pedunculopontine tegmentum (PPTg) and pontine nuclei - regions known to be important in mediating reinstatement of drug-taking after abstinence. These changes occur concomitantly with behavioral sensitization and appear to be mediated through increases in dopamine D1 and D3 and decreases in D2 receptor protein and mRNA expression. We further identify a novel region of dorsal caudate/putamen, with a low density of calbindin neurons, that has an opposite hemodynamic response to meth than the rest of the caudate/putamen and accumbens and shows very strong correlation with dorsal CA1 and CA3 hippocampus. This correlation switches following meth abstinence from CA1/CA3 to strong connections with ventral hippocampus (ventral subiculum) and nucleus accumbens. These data provide novel evidence for temporal alterations in brain connectivity where chronic meth can subvert hippocampal - striatal interactions from cognitive control regions to regions that mediate drug reinstatement. Our results also demonstrate that the signs and magnitudes of the induced CBV changes following challenge with meth or a D3-preferring agonist are a complementary read out of the relative changes that occur in D1, D2 and D3 receptors using protein or mRNA levels.


Subject(s)
Corpus Striatum/drug effects , Corpus Striatum/physiology , Hippocampus/drug effects , Hippocampus/physiology , Methamphetamine/administration & dosage , Sensorimotor Cortex/drug effects , Sensorimotor Cortex/physiology , Animals , Brain/blood supply , Brain/drug effects , Brain/physiology , Brain Mapping , Cerebral Blood Volume , Conditioning, Classical , Corpus Striatum/blood supply , Drug-Seeking Behavior , Hippocampus/blood supply , Locomotion/drug effects , Magnetic Resonance Imaging , Male , Neural Pathways/blood supply , Neural Pathways/drug effects , Neural Pathways/physiology , Rats, Sprague-Dawley , Sensorimotor Cortex/blood supply
5.
IUBMB Life ; 70(2): 143-152, 2018 02.
Article in English | MEDLINE | ID: mdl-29341457

ABSTRACT

Neuropathic pain is a complex, chronic pain condition and the treatment is a major clinical challenge. Recent studies have shown that two FDA approved drugs dexmedetomidine (DEX) and midazolam (MZL), may be useful in treating neuropathic pain, but the mechanism is not fully dementated. Here, we investigated the effects and mechanisms of DEX and MZL treatment in the peripheral nerve injury model. Intramuscular injection with DEX and MZL attenuated the development of mechanical allodynia and thermal hyperalgesia in rats with chronic constriction injury (CCI). Concurrently, the expression of NMDA receptor subunit 2B (NR2B), GABA (A) receptor subunit alpha1 (GABAA-α1), and Sonic Hedgehog (SHH) displayed different temporal patterns in the thalamus and the ipsilateral dorsal horn of the spinal cord after CCI. Such that (1) NR2B expression was decreased on day 1 and 14, whereas GABAA-α1 expression was increased on day 1 in the thalamus, and NR2B expression was decreased on day 1, whereas GABAA-α1 expression was increased on day 1 and day 30 in the ipsilateral spinal cord dorsal horn after DEX treatment. (2) NR2B expression was increased on day 1, then decreased on day 14 and returned to baseline on day30, whereas GABAA-α1 expression was no significant changes on day 1, 14, 30 in the thalamus, and NR2B expression was decreased on day 14 and 30, whereas GABAA-α1 expression was no changes on day 1 and 14 but increased on day 30 after MZL treatment. Furthermore, the mechanical allodynia was significantly attenuated after PUR administration. Meanwhile the expression of NR2B was significantly decreased, and the expression of GABAA-α1 was significantly increased, in the thalamus and in the ipsilateral spinal cord dorsal horn when detected on postoperative day 1, 7, and 14. Our findings indicate that DEX and MZL have different mechanisms in CCI rats, suggesting different strategies could be considered in managing neuropathic pain in different individuals. © 2018 IUBMB Life, 70(2):143-152, 2018.


Subject(s)
Dexmedetomidine/pharmacology , Midazolam/pharmacology , Peripheral Nerve Injuries/drug therapy , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Disease Models, Animal , Hedgehog Proteins/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Male , Neuralgia/drug therapy , Peripheral Nerve Injuries/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/metabolism , Thalamus/drug effects , Thalamus/metabolism
6.
J Pain ; 19(6): 626-634, 2018 06.
Article in English | MEDLINE | ID: mdl-29366880

ABSTRACT

Orofacial neuropathic pain caused by trigeminal nerve injury is a debilitating condition with limited therapeutic options. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels regulate neuronal excitability and are involved in the development and maintenance of chronic pain. However, the effect of HCN channel activity in the Gasserian ganglion on trigeminal neuropathic pain has not been examined. We evaluated nociceptive behaviors after microinjection of the HCN channel blockers ZD7288 or ivabradine into the Gasserian ganglion in rats with trigeminal nerve injury. Both blockers dose-dependently ameliorated evoked and spontaneous nociceptive behavior in rats with trigeminal neuropathic pain. Moreover, the clinically available HCN channel blocker ivabradine showed a prolonged antinociceptive effect. In the Gasserian ganglion, HCN1 and HCN2 are major HCN isoforms. After trigeminal nerve injury, the counts of HCN1 as well as HCN2 immuno-positive punctae were increased in the ipsilateral Gasserian ganglions. These results indicate that the increased HCN channel activity in the Gasserian ganglion directly contributes to neuropathic pain resulting from trigeminal nerve injury. PERSPECTIVE: Trigeminal nerve damage-induced orofacial pain is severe and more resistant to standard pharmacological treatment than other types of neuropathic pain. Our study suggests that targeting HCN channel activities in the Gasserian ganglion may provide an alternative treatment of trigeminal neuropathy including trigeminal neuralgia.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Neuralgia/metabolism , Trigeminal Ganglion/metabolism , Trigeminal Neuralgia/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley
7.
Pain Med ; 19(4): 686-692, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29177411

ABSTRACT

Objective: Correlation between radiologic structural abnormalities and clinical symptoms in low back pain patients is poor. There is an unmet clinical need to image inflammation in pain conditions to aid diagnosis and guide treatment. Ferumoxytol, an ultrasmall superparamagnetic iron oxide (USPIO) nanoparticle, is clinically used to treat iron deficiency anemia and showed promise in imaging tissue inflammation in human. We explored whether ferumoxytol can be used to identify tissue and nerve inflammation in pain conditions in animals and humans. Methods: Complete Freud's adjuvant (CFA) or saline was injected into mice hind paws to establish an inflammatory pain model. Ferumoxytol (20 mg/kg) was injected intravenously. Magnetic resonance imaging (MRI) was performed prior to injection and 72 hours postinjection. The changes in the transverse relaxation time (T2) before and after ferumoxytol injection were compared between mice that received CFA vs saline injection. In the human study, we administered ferumoxytol (4 mg/kg) to a human subject with clinical symptoms of lumbar radiculopathy and compared the patient with a healthy subject. Results: Mice that received CFA exhibited tissue inflammation and pain behaviors. The changes in T2 before and after ferumoxytol injection were significantly higher in mice that received CFA vs saline (20.8 ± 3.6 vs 2.2 ± 2.5, P = 0.005). In the human study, ferumoxytol-enhanced MRI identified the nerve root corresponding to the patient's symptoms, but the nerve root was not impinged by structural abnormalities, suggesting the potential superiority of this approach over conventional structural imaging techniques. Conclusions: Ferumoxytol-enhanced MRI can identify tissue and nerve inflammation and may provide a promising diagnostic tool in assessing pain conditions in humans.


Subject(s)
Contrast Media , Diagnostic Imaging/methods , Ferrosoferric Oxide , Inflammation/diagnostic imaging , Pain/diagnostic imaging , Radiculopathy/diagnostic imaging , Adult , Animals , Humans , Image Interpretation, Computer-Assisted , Magnetic Resonance Imaging/methods , Male , Metal Nanoparticles , Mice , Mice, Inbred C57BL , Middle Aged
8.
Nat Neurosci ; 20(9): 1213-1216, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28714953

ABSTRACT

Chemotherapy-induced pain is a dose-limiting condition that affects 30% of patients undergoing chemotherapy. We found that gut microbiota promotes the development of chemotherapy-induced mechanical hyperalgesia. Oxaliplatin-induced mechanical hyperalgesia was reduced in germ-free mice and in mice pretreated with antibiotics. Restoring the microbiota of germ-free mice abrogated this protection. These effects appear to be mediated, in part, by TLR4 expressed on hematopoietic cells, including macrophages.


Subject(s)
Antineoplastic Agents/toxicity , Gastrointestinal Microbiome/physiology , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Pain/chemically induced , Pain/metabolism , Animals , Cells, Cultured , Female , Gastrointestinal Microbiome/drug effects , Hyperalgesia/microbiology , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organoplatinum Compounds/toxicity , Oxaliplatin , Pain/microbiology , Pain Measurement/methods , Random Allocation , Rats , Rats, Sprague-Dawley
9.
Anesth Analg ; 125(3): 1032-1041, 2017 09.
Article in English | MEDLINE | ID: mdl-28537967

ABSTRACT

BACKGROUND: Combination drug therapy is commonly used to treat chronic pain conditions such as neuropathic pain, and antidepressant is often used together with opioid analgesics. While rewarding is an intrinsic property of opioid analgesics, it is unknown whether the use of an antidepressant would influence opioid reward, which may contribute to opioid addiction. In this study, we examined whether nortriptyline (a tricyclic antidepressant and a first-line medication for neuropathic pain) would enhance the morphine rewarding property in both naive and chronic constriction sciatic nerve injury (CCI) rats. METHODS: The rewarding effect of these drugs was assessed using conditioned place preference (CPP). The real-time polymerase chain reaction, western blot, and enzyme-linked immunosorbent assay analysis were used to investigate the function of central noradrenergic system. RESULTS: In naive rats, coadministration of nortriptyline with morphine did not change the acquisition of morphine-induced CPP. However, nortriptyline enhanced the acquisition, delayed the extinction, and augmented the reinstatement of morphine-induced CPP in CCI rats. In CCI rats treated with both nortriptyline and morphine, the expression of α2A-adrenergic receptors, norepinephrine transporter, and tyrosine hydroxylase was markedly decreased in the locus coeruleus, whereas the norepinephrine concentration in the nucleus accumbens was remarkably increased. CONCLUSIONS: These results demonstrate that nortriptyline enhanced morphine reward when both drugs were used to treat neuropathic pain in rats and that this behavioral phenotype is likely to be mediated by upregulation of the central noradrenergic system. These findings may have implications in opioid therapy commonly used for chronic pain management.


Subject(s)
Conditioning, Operant/drug effects , Morphine/administration & dosage , Neuralgia/drug therapy , Neuralgia/metabolism , Norepinephrine/metabolism , Nortriptyline/administration & dosage , Adrenergic Neurons/drug effects , Adrenergic Neurons/metabolism , Analgesics, Opioid/administration & dosage , Animals , Conditioning, Operant/physiology , Drug Therapy, Combination , Male , Rats
10.
J Pain ; 18(8): 899-907, 2017 08.
Article in English | MEDLINE | ID: mdl-28238950

ABSTRACT

The number of studies on trigeminal nerve injury using animal models remains limited. A rodent model of trigeminal neuropathic pain was first developed in 1994, in which chronic constriction injury (CCI) is induced by ligation of the infraorbital nerve (IoN). This animal model has served as a major tool to study trigeminal neuropathic pain. Unfortunately, the surgical procedure in this model is complicated and far more difficult than ligation of peripheral nerves (eg, sciatic nerve). The aim of this study was to improve on the current surgical procedure of IoN ligation to induce trigeminal neuropathic pain in rats. We show that the IoN can be readily accessed through a small facial incision. CCI can be induced by ligation of a segment at the distal IoN (dIoN). This dIoN-CCI procedure is simple, minimally invasive, and time-saving. Our data show that the dIoN-CCI procedure consistently induced acute as well as chronic nociceptive behaviors in rats. Daily gabapentin treatment attenuated mechanical allodynia and reduced face-grooming episodes in dIoN-CCI rats. PERSPECTIVE: The orofacial pain caused by trigeminal nerve damage is severe and perhaps more debilitating than other types of neuropathic pain. However, studies on trigeminal neuropathic pain remain limited. This is largely because of the lack of proper animal models because of the complexity of the existing surgical procedures required to induce trigeminal nerve injury. Our improved dIoN-CCI model is likely to make it more accessible to study the cellular and molecular mechanisms of neuropathic pain caused by trigeminal nerve damage.


Subject(s)
Amines/therapeutic use , Analgesics/therapeutic use , Cyclohexanecarboxylic Acids/therapeutic use , Functional Laterality/physiology , Neuralgia/drug therapy , Neuralgia/etiology , Pain/etiology , Trigeminal Neuralgia/etiology , gamma-Aminobutyric Acid/therapeutic use , Animals , Disease Models, Animal , Gabapentin , Grooming , Hyperalgesia/etiology , Hyperalgesia/therapy , Male , Pain Measurement , Rats , Rats, Sprague-Dawley , Time Factors
11.
J Neurosci ; 34(29): 9779-88, 2014 Jul 16.
Article in English | MEDLINE | ID: mdl-25031415

ABSTRACT

Opioid analgesics are commonly used in chronic pain management despite a potential risk of rewarding. However, it remains unclear whether opioid analgesia would enhance the opioid rewarding effect thereby contributing to opioid rewarding. Utilizing a rat paradigm of conditioned place preference (CPP) combined with ankle monoarthritis as a condition of persistent nociception, we showed that analgesia induced by either morphine or the nonsteroid anti-inflammatory drug ibuprofen increased CPP scores in arthritic rats, suggesting that analgesia itself had a rewarding effect. However, arthritic rats exhibited a significantly higher CPP score in response to morphine than ibuprofen. Thus, the rewarding effect of morphine was enhanced in the presence of persistent nociception, producing a phenomenon of analgesia-enhanced opioid reward. At the cellular level, administration of morphine activated a cascade of leptin expression, glial activation, and dopamine receptor upregulation in the nucleus accumbens (NAc), while administration of ibuprofen decreased glial activation with no effect on leptin expression in the NAc. Furthermore, the morphine rewarding effect was blocked in leptin deficient ob/ob mice or by neutralizing leptin or interleukin-1ß in the NAc without diminishing morphine analgesia. The data indicate that systemic opioid can activate a leptin-mediated central mechanism in the NAc that led to the enhanced opioid rewarding effect. These findings provide evidence for an interaction between opioid analgesia and opioid rewarding, which may have implications in clinical opioid dose escalation in chronic pain management.


Subject(s)
Analgesics, Opioid/administration & dosage , Leptin/physiology , Morphine/administration & dosage , Pain/drug therapy , Reward , Animals , Animals, Newborn , Arthritis, Experimental/complications , Astrocytes/drug effects , Cerebral Cortex/cytology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Leptin/genetics , Leptin/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Pain/etiology , Pain Measurement/drug effects , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley
12.
Pain ; 155(8): 1504-1515, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24793908

ABSTRACT

Anxiety disorder is a comorbid condition of chronic pain. Analgesics and anxiolytics, subject to addiction and abuse, are currently used to manage pain and anxiety symptoms. However, the cellular mechanism underlying chronic pain and anxiety interaction remains to be elucidated. We report that persistent nociception following peripheral nerve injury induced anxiety-like behavior in rodents. Brain expression and release of neuropeptide S (NPS), a proposed endogenous anxiolytic peptide, was diminished in rodents with coexisting nociceptive and anxiety-like behaviors. Intracerebroventricular administration of exogenous NPS concurrently improved both nociceptive and anxiety-like behaviors. At the cellular level, NPS enhanced intra-amygdaloidal inhibitory transmission by increasing presynaptic gamma-aminobutyric acid (GABA) release from interneurons. These findings indicate that the interaction between nociceptive and anxiety-like behaviors in rodents may be regulated by the altered NPS-mediated intra-amygdaloidal GABAergic inhibition. The data suggest that enhancing the brain NPS function may be a new strategy to manage comorbid pain and anxiety.


Subject(s)
Amygdala/metabolism , Anxiety/metabolism , Behavior, Animal/physiology , Neuropeptides/metabolism , Nociception/physiology , Amygdala/drug effects , Amygdala/physiopathology , Animals , Anxiety/physiopathology , Disease Models, Animal , Exploratory Behavior/drug effects , Male , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neuropeptides/pharmacology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism
13.
Brain Res ; 1535: 137-47, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-23998984

ABSTRACT

Previous studies have shown that the morphology and number of cells in the spinal cord dorsal horn could change following peripheral nerve injury and that the Hippo signaling pathway plays an important role in cell growth, proliferation, apoptosis, and dendritic remolding. In the present study, we examined whether the expression of YAP and TAZ, two critical components regulated by Hippo signaling, in the spinal cord dorsal horn would be altered by chronic constriction sciatic nerve injury (CCI). We found that (1) YAP was mainly expressed on CGRP- and IB4-immunoreactive primary afferent nerve terminals without noticeable expression on glial cells, whereas TAZ was mainly expressed on spinal cord second order neurons as well as microglia; (2) upregulation of YAP and TAZ expression followed two distinct temporal patterns after CCI, such that the highest expression of YAP and TAZ was on day 14 and day 1 after CCI, respectively; (3) there were also unique topographic patterns of YAP and TAZ distribution in the spinal cord dorsal horn consistent with their distinctive association with primary afferents and second order neurons; (4) changes in the YAP expression were selectively induced by CCI but not CFA-induced hindpaw inflammation; and (5) the number of nuclear profiles of TAZ expression was significantly increased after CCI, indicating translocation of TAZ from the cytoplasma to nucleus. These findings indicate that peripheral nerve injury induced time-dependent and region-specific changes in the spinal YAP and TAZ expression. A role for Hippo signaling in synaptic and structural plasticity is discussed in relation to the cellular mechanism of neuropathic pain.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Peripheral Nerve Injuries/metabolism , Signal Transduction/physiology , Spinal Cord/metabolism , Transcription Factors/metabolism , Acyltransferases , Animals , Calcitonin Gene-Related Peptide/metabolism , Hyperalgesia/metabolism , Lectins/metabolism , Male , Neuralgia/metabolism , Neurons/metabolism , Pain Measurement , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Up-Regulation , YAP-Signaling Proteins
14.
Pain Med ; 13(12): 1590-600, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23110394

ABSTRACT

OBJECTIVE: To examine the hypothesis that glial activation would regulate the expression of the N-methyl-D-aspartate receptor subunit 1 (NR1) in the trigeminal subnucleus caudalis (Sp5C) after temporomandibular joint (TMJ) inflammation. METHODS: Inflammation of TMJ was produced in rats by injecting 50 µL complete Freund's adjuvant (CFA) into unilateral TMJ space. Sham control rats received incomplete Freund's adjuvant injection. Mechanical nociception in the affected and non-affected TMJ site was tested by using a digital algometer. Fractalkine, fluorocitrate, and/or MK801 were intracisternally administrated to examine the relationship between astroglial activation and NR1 upregulation. RESULTS: CFA TMJ injection resulted in persistent ipsilateral mechanical hyperalgesia 1, 3, and 5 days after CFA injection. The inflammation also induced significant upregulation of CX3C chemokine receptor 1 and glial fibrillary acidic protein (GFAP) beginning on day 1 and of NR1 beginning on day 3 within the ipsilateral Sp5C. Intracisternal administration of fluorocitrate for 5 days blocked the development of mechanical hyperalgesia as well as the upregulation of GFAP and NR1 in the Sp5C. Conversely, intracisternal injection of fractalkine for 5 days exacerbated the expression of NR1 in Sp5C and mechanical hyperalgesia induced by TMJ inflammation. Moreover, once daily intracisternal fractalkine administration for 5 days in naïve rats induced the upregulation of NR1 and mechanical hyperalgesia. CONCLUSIONS: These results suggest that astroglial activation contributes to the mechanism of TMJ pain through the regulation of NR1 expression in Sp5C.


Subject(s)
Astrocytes/metabolism , Hyperalgesia/metabolism , Inflammation/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Temporomandibular Joint Disorders/metabolism , Trigeminal Caudal Nucleus/metabolism , Adjuvants, Immunologic , Animals , CX3C Chemokine Receptor 1 , Disease Models, Animal , Freund's Adjuvant , Glial Fibrillary Acidic Protein/metabolism , Hyperalgesia/physiopathology , Inflammation/chemically induced , Inflammation/physiopathology , Male , Pain Measurement , Physical Stimulation , Rats , Rats, Sprague-Dawley , Receptors, Chemokine/metabolism , Temporomandibular Joint Disorders/chemically induced , Temporomandibular Joint Disorders/physiopathology , Trigeminal Caudal Nucleus/physiopathology , Up-Regulation
15.
Pain ; 153(12): 2448-2457, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23046768

ABSTRACT

A connection between pain and depression has long been recognized in the clinical setting; however, its mechanism remains unclear. This study showed that mechanical hyperalgesia induced by unilateral temporomandibular joint (TMJ) inflammation was exacerbated in Wistar-Kyoto (WKY) rats with genetically predisposed depressive behavior. Reciprocally, TMJ inflammation enhanced depressive behavior such that a lower nociceptive threshold correlated with a higher score of depressive behavior in the same WKY rats. As compared with Wistar rats, WKY rats showed a lower plasma melatonin level, downregulation of the melatonin MT1 receptor, but upregulation of the NR1 subunit of the NMDA receptor in the ipsilateral trigeminal subnucleus caudalis (Sp5C). Intracisternal administration of 6-chloromelatonin (250 µg, twice daily for 7 days) concurrently attenuated mechanical hyperalgesia and depressive behavior in WKY rats as well as downregulated the NR1 expression in the ipsilateral Sp5C. In patch-clamp recordings, melatonin dose-dependently decreased NMDA-induced currents in spinal cord dorsal horn substantia gelatinosa neurons. These results demonstrate a reciprocal relationship between TMJ inflammation-induced mechanical hyperalgesia and depressive behavior and suggest that the central melatoninergic system, through modulation of the NMDA receptor expression and activity, may play a role in the mechanisms of the comorbidity between pain and depression.


Subject(s)
Depressive Disorder/complications , Depressive Disorder/physiopathology , Genetic Predisposition to Disease/genetics , Hyperalgesia/complications , Hyperalgesia/physiopathology , Receptors, Melatonin/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Male , Rats , Rats, Inbred WKY , Touch
16.
J Clin Invest ; 122(8): 2940-54, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22751107

ABSTRACT

Pain and depression are frequently comorbid disorders, but the mechanism underlying this association is unknown. Here, we report that brain indoleamine 2,3-dioxygenase 1 (IDO1), a rate-limiting enzyme in tryptophan metabolism, plays a key role in this comorbidity. We found that chronic pain in rats induced depressive behavior and IDO1 upregulation in the bilateral hippocampus. Upregulation of IDO1 resulted in the increased kynurenine/tryptophan ratio and decreased serotonin/tryptophan ratio in the bilateral hippocampus. We observed elevated plasma IDO activity in patients with both pain and depression, as well as in rats with anhedonia induced by chronic social stress. Intra-hippocampal administration of IL-6 in rats, in addition to in vitro experiments, demonstrated that IL-6 induces IDO1 expression through the JAK/STAT pathway. Further, either Ido1 gene knockout or pharmacological inhibition of hippocampal IDO1 activity attenuated both nociceptive and depressive behavior. These results reveal an IDO1-mediated regulatory mechanism underlying the comorbidity of pain and depression and suggest a new strategy for the concurrent treatment of both conditions via modulation of brain IDO1 activity.


Subject(s)
Brain/enzymology , Brain/physiopathology , Chronic Pain/enzymology , Chronic Pain/physiopathology , Depression/enzymology , Depression/physiopathology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Adolescent , Adult , Aged , Animals , Chronic Pain/complications , Comorbidity , Depression/etiology , Female , Gene Knockout Techniques , Hippocampus/drug effects , Hippocampus/enzymology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/blood , Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interleukin-6/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Rats , Rats, Wistar , Signal Transduction/drug effects , Up-Regulation , Young Adult
17.
Pain ; 152(6): 1263-1271, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21376468

ABSTRACT

Recent studies have shown that leptin (an adipocytokine) played an important role in nociceptive behavior induced by nerve injury, but the cellular mechanism of this action remains unclear. Using the whole-cell patch-clamp recording from rat's spinal cord slices, we showed that superfusion of leptin onto spinal cord slices dose-dependently enhanced N-methyl-d-aspartate (NMDA) receptor-mediated currents in spinal cord lamina II neurons. At the cellular level, the effect of leptin on spinal NMDA-induced currents was mediated through the leptin receptor and the JAK2/STAT3 (but not PI3K or MAPK) pathway, as the leptin effect was abolished in leptin receptor-deficient (db/db) mice and inhibited by a JAK/STAT inhibitor. Moreover, we demonstrated in naïve rats that a single intrathecal administration of leptin enhanced spontaneous biting, scratching, and licking behavior induced by intrathecal NMDA and that repeated intrathecal administration of leptin elicited thermal hyperalgesia and mechanical allodynia, which was attenuated by the noncompetitive NMDA receptor antagonist MK-801. Intrathecal leptin also upregulated the expression of NMDA receptors and pSTAT3 within the rat's spinal cord dorsal horn, and intrathecal MK-801 attenuated this leptin effect as well. Our data demonstrate a relationship between leptin and NMDA receptor-mediated spinal neuronal excitation and its functional role in nociceptive behavior. Since leptin contributes to nociceptive behavior induced by nerve injury, the present findings suggest an important cellular link between the leptin's spinal effect and the NMDA receptor-mediated cellular mechanism of neuropathic pain. A functional link is demonstrated between leptin, an adipocytokine, and the cellular mechanisms of neuropathic pain via enhancement of function and expression of spinal N-methyl-d-aspartate receptors.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Leptin/pharmacology , N-Methylaspartate/pharmacology , Sensory Receptor Cells/drug effects , Spinal Cord/drug effects , Analysis of Variance , Animals , Behavior, Animal/drug effects , Dizocilpine Maleate/therapeutic use , Drug Interactions , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/therapeutic use , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Knockout , Neuralgia/chemically induced , Neuralgia/physiopathology , Pain Measurement , Patch-Clamp Techniques/methods , Physical Stimulation/adverse effects , Rats , Rats, Sprague-Dawley , Receptors, Leptin/deficiency , Receptors, N-Methyl-D-Aspartate/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Spinal Cord/cytology , Up-Regulation/drug effects , Up-Regulation/genetics
18.
Brain Res ; 1378: 66-71, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21223950

ABSTRACT

The hippocampus plays an important role in learning and memory and possibly contributes to the formation of pain-related memory and emotional responses. However, there is currently little data linking the hippocampus to neuropathic pain. It has been reported that NF-κB is an important regulatory factor in memory consolidation within the hippocampus. This study aims to examine a possible relationship between the hippocampal NF-κB expression and nerve injury-induced thermal hyperalgesia using a rat model of constriction sciatic nerve injury (CCI). Immunofluorescence and Western blot analysis were performed to detect and quantify the hippocampal NF-κB expression. Thermal hyperalgesia was examined on day 0 and postoperative days 1, 7 and 14. The nuclear portion of the p65 NF-κB expression was significantly increased on the contralateral side on days 7 and 14 as well as significantly increased on the ipsilateral side on day 14 as compared to the sham control group. Intraperitoneal administration of MK-801, an N-methyl-d-aspartate (NMDA) receptor antagonist, reduced hyperalgesia and modulated the NF-κB expression in the contralateral side of hippocampus. These results suggest an association between the hippocampal NF-κB expression and the behavioral manifestation of thermal hyperalgesia, which is likely to be mediated through activation of the NMDA receptor.


Subject(s)
Hippocampus/metabolism , Hyperalgesia/metabolism , NF-kappa B/biosynthesis , Neuralgia/metabolism , Animals , Blotting, Western , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Fluorescent Antibody Technique , Functional Laterality/physiology , Hot Temperature , Ligation , Male , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , Sciatic Nerve/injuries
19.
J Pain ; 12(1): 13-21, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20537956

ABSTRACT

UNLABELLED: The expression of NF-κB in the spinal cord is associated with neuropathic pain. However, little is known about its expression beyond the spinal cord. Here we examined a spatial and temporal pattern of the NF-κB expression in both spinal and supraspinal regions. After chronic constriction injury (CCI) of the sciatic nerve, NF-κB (p65) expression was significantly increased in the ipsilateral spinal cord. In contrast, the NF-κB expression in the contralateral primary somatosensory cortex was decreased with no significant differences seen in the thalamus. In the contralateral anterior cingulate cortex, the NF-κB expression was increased significantly on day 14 as compared with the sham group. In the contralateral amygdala, the NF-κB expression showed a time-dependent downregulation after CCI, which became significant on day 14. MK-801 reduced nociceptive behaviors and reversed the direction of NF-κB expression. These results indicate that the CCI-induced expression of p65 NF-κB is both time-dependent and region-specific, in areas that process both sensory-discriminative and motivational-affective dimensions of pain. PERSPECTIVE: This article presents a spatiotemporal mapping of the NF-κB expression in spinal and supraspinal regions after peripheral nerve injury. These findings point to an involvement of NF-κB beyond the spinal cord in both the sensory discriminative and emotional affective aspects of neuropathic pain processing.


Subject(s)
Gene Expression Regulation/physiology , NF-kappa B/metabolism , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Somatosensory Cortex/metabolism , Spinal Cord/metabolism , Animals , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Dizocilpine Maleate/therapeutic use , Functional Laterality/physiology , Gene Expression Regulation/drug effects , Male , Neuralgia/drug therapy , Neuralgia/etiology , Neuralgia/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Pain Measurement , Peripheral Nervous System Diseases/complications , Rats , Synaptotagmin I/metabolism , Thalamus/metabolism , Thalamus/pathology
20.
Brain Res ; 1288: 42-9, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19595681

ABSTRACT

Previous study has shown that administration of melatonin into the anterior cingulate cortex contralateral to peripheral nerve injury prevented exacerbation of mechanical allodynia with a concurrent improvement of depression-like behavior in Wistar-Kyoto (WKY) rats, a genetic variation of Wistar rats. In the present study, we examined the effect of the individual versus combined treatment of melatonin and/or dextromethorphan (DM), a clinically available N-methyl-d-aspartate (NMDA) receptor antagonist, on pain behaviors in WKY rats with chronic constriction sciatic nerve injury (CCI). Pain behaviors (thermal hyperalgesia and mechanical allodynia) were established at one week after CCI. WKY rats were then treated intraperitoneally with various doses of melatonin, DM or their combination once daily for the following week. At the end of this one-week treatment, behavioral tests were repeated in these same rats. While DM alone was effective in reducing thermal hyperalgesia at three tested doses (15, 30 or 60 mg/kg), it reduced mechanical allodynia only at high doses (30 or 60 mg/kg). By comparison, administration of melatonin alone was effective in reducing thermal hyperalgesia only at the highest dose (120 mg/kg, but not 30 or 60 mg/kg) tested in this experiment. Melatonin alone failed to reverse allodynia at all three tested doses (30, 60 and 120 mg/kg). However, the combined intraperitoneal administration of melatonin (30 mg/kg) and DM (15 mg/kg) effectively reversed both thermal hyperalgesia and mechanical allodynia although each individual dose alone did not reduce pain behaviors. These results suggest that a combination of melatonin with a clinically available NMDA receptor antagonist might be more effective than either drug alone for the treatment of neuropathic pain.


Subject(s)
Chronic Disease/drug therapy , Dextromethorphan/therapeutic use , Melatonin/therapeutic use , Neuralgia/drug therapy , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Drug Therapy, Combination , Hyperalgesia/drug therapy , Injections, Intraperitoneal , Male , Pain Measurement , Pain Threshold/drug effects , Rats , Rats, Inbred WKY , Sciatic Nerve/injuries
SELECTION OF CITATIONS
SEARCH DETAIL
...