Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J Allergy Clin Immunol ; 141(2): 608-619.e7, 2018 02.
Article in English | MEDLINE | ID: mdl-28807414

ABSTRACT

BACKGROUND: Chronic itch is a highly debilitating symptom that underlies many medical disorders with no universally effective treatments. Although unique neuronal signaling cascades in the sensory ganglia and spinal cord have been shown to critically promote the pathogenesis of chronic itch, the role of skin-associated cells remains poorly understood. OBJECTIVE: We sought to examine the cutaneous mechanisms underlying transient receptor potential vanilloid 4 (TRPV4)-mediated allergic and nonallergic chronic itch. METHODS: Expression of TRPV4 in chronic itch and healthy control skin preparations was examined by using real-time RT-PCR. Trpv4eGFP mice were used to study the expression and function of TRPV4 in the skin by means of immunofluorescence staining, flow cytometry, calcium imaging, and patch-clamp recordings. Genetic and pharmacologic approaches were used to examine the role and underlying mechanisms of TRPV4 in mouse models of dry skin-associated chronic itch and spontaneous scratching associated with squaric acid dibutylester-induced allergic contact dermatitis. RESULTS: TRPV4 is selectively expressed by dermal macrophages and epidermal keratinocytes in mice. Lineage-specific deletion of TRPV4 in macrophages and keratinocytes reduces allergic and nonallergic chronic itch in mice, respectively. Importantly, TRPV4 expression is significantly increased in skin biopsy specimens from patients with chronic idiopathic pruritus in comparison with skin from healthy control subjects. Moreover, TRPV4-dependent chronic itch requires 5-hydroxytryptamine (5-HT) signaling secondary to activation of distinct 5-HT receptors in mice with allergic and those with nonallergic chronic itch conditions. CONCLUSION: Our study reveals previously unrecognized mechanisms by which TRPV4-expressing epithelial and immune cells in the skin critically and dynamically mediate chronic itch and unravels novel targets for therapeutics in the setting of chronic itch.


Subject(s)
Dermatitis, Allergic Contact/immunology , Dermis/immunology , Gene Expression Regulation/immunology , Keratinocytes/immunology , Macrophages/immunology , Pruritus/immunology , TRPV Cation Channels/immunology , Animals , Chronic Disease , Dermatitis, Allergic Contact/genetics , Dermatitis, Allergic Contact/pathology , Dermis/pathology , Female , Gene Expression Regulation/genetics , Humans , Keratinocytes/pathology , Macrophages/pathology , Male , Mice , Mice, Knockout , Pruritus/genetics , Pruritus/pathology , TRPV Cation Channels/genetics
2.
Sci Rep ; 7(1): 1842, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28500286

ABSTRACT

Mechanical pain serves as a base clinical symptom for many of the world's most debilitating syndromes. Ion channels expressed by peripheral sensory neurons largely contribute to mechanical hypersensitivity. Transient Receptor Potential A 1 (TRPA1) is a ligand-gated ion channel that contributes to inflammatory mechanical hypersensitivity, yet little is known as to the post-translational mechanism behind its somatosensitization. Here, we utilize biochemical, electrophysiological, and behavioral measures to demonstrate that metabotropic glutamate receptor-induced sensitization of TRPA1 nociceptors stimulates targeted modification of the receptor. Type 1 mGluR5 activation increases TRPA1 receptor agonist sensitivity in an AKA-dependent manner. As a scaffolding protein for Protein Kinases A and C (PKA and PKC, respectively), AKAP facilitates phosphorylation and sensitization of TRPA1 in ex vivo sensory neuronal preparations. Furthermore, hyperalgesic priming of mechanical hypersensitivity requires both TRPA1 and AKAP. Collectively, these results identify a novel AKAP-mediated biochemical mechanism that increases TRPA1 sensitivity in peripheral sensory neurons, and likely contributes to persistent mechanical hypersensitivity.


Subject(s)
A Kinase Anchor Proteins/metabolism , Receptors, Metabotropic Glutamate/metabolism , TRPA1 Cation Channel/metabolism , A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/genetics , Animals , CHO Cells , Calcium/metabolism , Chromatography, Liquid , Cricetulus , Male , Mice , Mice, Knockout , Molecular Imaging , Phosphorylation , Rats , Receptors, Metabotropic Glutamate/chemistry , TRPA1 Cation Channel/chemistry , TRPA1 Cation Channel/genetics , Tandem Mass Spectrometry
3.
J Neurotrauma ; 34(6): 1260-1270, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28073317

ABSTRACT

A majority of people who have sustained spinal cord injury (SCI) experience chronic pain after injury, and this pain is highly resistant to available treatments. Contusive SCI in rats at T10 results in hyperexcitability of primary sensory neurons, which contributes to chronic pain. KCNQ channels are widely expressed in nociceptive dorsal root ganglion (DRG) neurons, are important for controlling their excitability, and their activation has proven effective in reducing pain in peripheral nerve injury and inflammation models. The possibility that activators of KCNQ channels could be useful for treating SCI-induced chronic pain is strongly supported by the following findings. First, SCI, unlike peripheral nerve injury, failed to decrease the functional or biochemical expression of KCNQ channels in DRG as revealed by electrophysiology, real-time quantitative polymerase chain reaction, and Western blot; therefore, these channels remain available for pharmacological targeting of SCI pain. Second, treatment with retigabine, a specific KCNQ channel opener, profoundly decreased spontaneous activity in primary sensory neurons of SCI animals both in vitro and in vivo without changing the peripheral mechanical threshold. Third, retigabine reversed SCI-induced reflex hypersensitivity, adding to our previous demonstration that retigabine supports the conditioning of place preference after SCI (an operant measure of spontaneous pain). In contrast to SCI animals, naïve animals showed no effects of retigabine on reflex sensitivity or conditioned place preference by pairing with retigabine, indicating that a dose that blocks chronic pain-related behavior has no effect on normal pain sensitivity or motivational state. These results encourage the further exploration of U.S. Food and Drug Administration-approved KCNQ activators for treating SCI pain, as well as efforts to develop a new generation of KCNQ activators that lack central side effects.


Subject(s)
Behavior, Animal/drug effects , Carbamates/pharmacology , Chronic Pain/metabolism , Ganglia, Spinal/metabolism , KCNQ Potassium Channels/metabolism , Membrane Transport Modulators/pharmacology , Phenylenediamines/pharmacology , Spinal Cord Injuries/metabolism , Animals , Carbamates/administration & dosage , Chronic Pain/drug therapy , Disease Models, Animal , Ganglia, Spinal/drug effects , KCNQ Potassium Channels/drug effects , Male , Membrane Transport Modulators/administration & dosage , Phenylenediamines/administration & dosage , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/drug therapy
4.
J Neurosci ; 35(35): 12217-31, 2015 Sep 02.
Article in English | MEDLINE | ID: mdl-26338332

ABSTRACT

Pain management in opioid abusers engenders ethical and practical difficulties for clinicians, often resulting in pain mismanagement. Although chronic opioid administration may alter pain states, the presence of pain itself may alter the propensity to self-administer opioids, and previous history of drug abuse comorbid with chronic pain promotes higher rates of opioid misuse. Here, we tested the hypothesis that inflammatory pain leads to increased heroin self-administration resulting from altered mu opioid receptor (MOR) regulation of mesolimbic dopamine (DA) transmission. To this end, the complete Freund's adjuvant (CFA) model of inflammation was used to assess the neurochemical and functional changes induced by inflammatory pain on MOR-mediated mesolimbic DA transmission and on rat intravenous heroin self-administration under fixed ratio (FR) and progressive ratio (PR) schedules of reinforcement. In the presence of inflammatory pain, heroin intake under an FR schedule was increased for high, but attenuated for low, heroin doses with concomitant alterations in mesolimbic MOR function suggested by DA microdialysis. Consistent with the reduction in low dose FR heroin self-administration, inflammatory pain reduced motivation for a low dose of heroin, as measured by responding under a PR schedule of reinforcement, an effect dissociable from high heroin dose PR responding. Together, these results identify a connection between inflammatory pain and loss of MOR function in the mesolimbic dopaminergic pathway that increases intake of high doses of heroin. These findings suggest that pain-induced loss of MOR function in the mesolimbic pathway may promote opioid dose escalation and contribute to opioid abuse-associated phenotypes. SIGNIFICANCE STATEMENT: This study provides critical new insights that show that inflammatory pain alters heroin intake through a desensitization of MORs located within the VTA. These findings expand our knowledge of the interactions between inflammatory pain and opioid abuse liability, and should help to facilitate the development of novel and safer opioid-based strategies for treating chronic pain.


Subject(s)
Analgesics, Opioid/administration & dosage , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , Pain , Receptors, Opioid, mu/metabolism , Ventral Tegmental Area/metabolism , Action Potentials/drug effects , Animals , Conditioning, Operant/drug effects , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Glycine Agents/pharmacology , Heroin/administration & dosage , Hyperalgesia/drug therapy , Inflammation/chemically induced , Inflammation/complications , Inhibitory Postsynaptic Potentials/drug effects , Male , Neurons/drug effects , Pain/drug therapy , Pain/pathology , Pain/psychology , Pain Threshold/drug effects , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Strychnine/pharmacology , Sucrose/administration & dosage , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/pathology
5.
Pain ; 156(11): 2364-2372, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26172554

ABSTRACT

Glutamate serves as the primary excitatory neurotransmitter in the nervous system. Previous studies have identified a role for glutamate and group I metabotropic receptors as targets for study in peripheral inflammatory pain. However, the coordination of signaling events that transpire from receptor activation to afferent neuronal sensitization has not been explored. Herein, we identify that scaffolding protein A-kinase anchoring protein 79/150 (AKAP150) coordinates increased peripheral thermal sensitivity after group I metabotropic receptor (mGluR5) activation. In both acute and persistent models of thermal somatosensory behavior, we report that mGluR5 sensitization requires AKAP150 expression. Furthermore, electrophysiological approaches designed to record afferent neuronal activity reveal that mGluR5 sensitization also requires functional AKAP150 expression. In dissociated primary afferent neurons, mGluR5 activation increases TRPV1 responses in an AKAP-dependent manner through a mechanism that induces AKAP association with TRPV1. Experimental results presented herein identify a mechanism of receptor-driven scaffolding association with ion channel targets. Importantly, this mechanism could prove significant in the search for therapeutic targets that repress episodes of acute pain from becoming chronic in nature.


Subject(s)
A Kinase Anchor Proteins/metabolism , Gene Expression Regulation/genetics , Receptors, Metabotropic Glutamate/metabolism , Sensory Receptor Cells/metabolism , A Kinase Anchor Proteins/genetics , Animals , Cells, Cultured , Estrenes/pharmacology , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Conduction/physiology , Neurotransmitter Agents/pharmacology , Pain Threshold/physiology , Peripheral Nerves/physiology , Phosphodiesterase Inhibitors/pharmacology , Physical Stimulation/adverse effects , Pyrrolidinones/pharmacology , Rats , Sensory Receptor Cells/drug effects , Skin/innervation , Trigeminal Ganglion/cytology
6.
Pain ; 156(5): 849-858, 2015 May.
Article in English | MEDLINE | ID: mdl-25734988

ABSTRACT

N-methyl-D-aspartate receptor (NMDAR) antagonists have been shown to reduce mechanical hypersensitivity in animal models of inflammatory pain. However, their clinical use is associated with significant dose-limiting side effects. Small-conductance Ca-activated K channels (SK) have been shown to modulate NMDAR activity in the brain. We demonstrate that in vivo activation of SK channels in the spinal cord can alleviate mechanical hypersensitivity in a rat model of inflammatory pain. Intrathecal (i.t.) administration of the SK channel activator, 6,7-dichloro-1H-indole-2,3-dione 3-oxime (NS309), attenuates complete Freund adjuvant (CFA)-induced mechanical hypersensitivity in a dose-dependent manner. Postsynaptic expression of the SK channel subunit, SK3, and apamin-sensitive SK channel-mediated currents recorded from superficial laminae are significantly reduced in the dorsal horn (DH) after CFA. Complete Freund adjuvant-induced decrease in SK-mediated currents can be reversed in vitro by bath application of NS309. In addition, immunostaining for the SK3 subunit indicates that SK3-containing channels within DH neurons can have both somatic and dendritic localization. Double immunostaining shows coexpression of SK3 and NMDAR subunit, NR1, compatible with functional interaction. Moreover, we demonstrate that i.t. coadministration of NS309 with an NMDAR antagonist reduces the dose of NMDAR antagonist, DL-2-amino-5-phosphonopentanoic acid (DL-AP5), required to produce antinociceptive effects in the CFA model. This reduction could attenuate the unwanted side effects associated with NMDAR antagonists, giving this combination potential clinical implications.


Subject(s)
Indoles/pharmacology , Inflammation/chemically induced , Oximes/pharmacology , Pain/drug therapy , Posterior Horn Cells/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Animals , Disease Models, Animal , Freund's Adjuvant/toxicity , Indoles/administration & dosage , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/physiopathology , Injections, Spinal , Male , Oximes/administration & dosage , Pain/chemically induced , Pain/metabolism , Pain/physiopathology , Pain Threshold , Posterior Horn Cells/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , Treatment Outcome
7.
J Physiol ; 592(16): 3403-11, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24879874

ABSTRACT

Nociceptive primary afferents have three surprising properties: they are highly complex in their expression of neurotransmitters and receptors and most probably participate in autocrine and paracrine interactions; they are capable of exerting tonic and activity-dependent inhibitory control over incoming nociceptive input; they can generate signals in the form of dorsal root reflexes that are transmitted antidromically out to the periphery and these signals can result in neurogenic inflammation in the innervated tissue. Thus, nociceptive primary afferents are highly complicated structures, capable of modifying input before it is ever transmitted to the central nervous system and capable of altering the tissue they innervate.


Subject(s)
Ganglia, Spinal/metabolism , Nociceptors/metabolism , Synaptic Transmission , Animals , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Humans , Neurotransmitter Agents/metabolism , Nociceptors/physiology , Reflex
8.
Ann Neurol ; 75(6): 837-50, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24633867

ABSTRACT

OBJECTIVE: Chronic pain is a common neurological comorbidity of human immunodeficiency virus (HIV)-1 infection, but the etiological cause remains elusive. The objective of this study was to identify the HIV-1 causal factor that critically contributes to the pathogenesis of HIV-associated pain. METHODS: We first compared the levels of HIV-1 proteins in postmortem tissues of the spinal cord dorsal horn (SDH) from HIV-1/acquired immunodeficiency syndrome patients who developed chronic pain (pain-positive HIV-1 patients) and HIV-1 patients who did not develop chronic pain (pain-negative HIV-1 patients). Then we used the HIV-1 protein that was specifically increased in the pain-positive patients to generate mouse models. Finally, we performed comparative analyses on the pathological changes in the models and the HIV-1 patients. RESULTS: We found that HIV-1 gp120 was significantly higher in pain-positive HIV-1 patients (vs pain-negative HIV-1 patients). This finding suggested that gp120 was a potential causal factor of the HIV-associated pain. To test this hypothesis, we used a mouse model generated by intrathecal injection of gp120 and compared the pathologies of the model and the pain-positive human HIV-1 patients. The results showed that the mouse model and pain-positive human HIV-1 patients developed extensive similarities in their pathological phenotypes, including pain behaviors, peripheral neuropathy, glial reactivation, synapse degeneration, and aberrant activation of pain-related signaling pathways in the SDH. INTERPRETATION: Our findings suggest that gp120 may critically contribute to the pathogenesis of HIV-associated pain.


Subject(s)
HIV Envelope Protein gp120/metabolism , HIV Infections/complications , Pain/etiology , Pain/metabolism , Adult , Animals , Case-Control Studies , Disease Models, Animal , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , HIV Envelope Protein gp120/genetics , Humans , Hyperalgesia/virology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pain/virology , Pain Threshold , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/virology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Spinal Cord/pathology , Viral Load
9.
Transl Res ; 164(1): 1-12, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24561013

ABSTRACT

Autophagy is essential to neuronal homeostasis, and its impairment is implicated in the development of neurodegenerative pathology. However, the underlying mechanisms and consequences of this phenomenon remain a matter of conjecture. We show that misexpression of human tau in Drosophila induces accumulation of autophagic intermediates with a preponderance of large vacuoles, which we term giant autophagic bodies (GABs), which are reminiscent of dysfunctional autophagic entities. Lowering basal autophagy reduces GABs, whereas increasing autophagy decreases mature autolysosomes. Induction of autophagy is also associated with rescue of the tauopathy phenotype, suggesting that formation of GABs may be a compensatory mechanism rather than a trigger of neurodegeneration. Last, we show that the peculiar Biondi bodies observed in the choroid epithelium of both elderly and Alzheimer's disease human brains express immunoreactive markers similar to those of GABs. Collectively, these data indicate that autophagic gridlock contributes to the development of pathology in aging and neurodegeneration.


Subject(s)
Aging/physiology , Autophagy/physiology , Neurodegenerative Diseases/metabolism , tau Proteins/metabolism , Aging/pathology , Animals , Animals, Genetically Modified , Biomarkers , Disease Models, Animal , Drosophila/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Genotype , Humans , Neurodegenerative Diseases/pathology , Retina/cytology , Retina/metabolism , Sirolimus/pharmacology , tau Proteins/genetics
10.
J Clin Invest ; 123(9): 3941-51, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23925292

ABSTRACT

Retinoids are structurally related derivatives of vitamin A and are required for normal vision as well as cell proliferation and differentiation. Clinically, retinoids are effective in treating many skin disorders and cancers. Application of retinoids evokes substantial irritating side effects, including pain and inflammation; however, the precise mechanisms accounting for the sensory hypersensitivity are not understood. Here we show that both naturally occurring and synthetic retinoids activate recombinant or native transient receptor potential channel vanilloid subtype 1 (TRPV1), an irritant receptor for capsaicin, the pungent ingredient of chili peppers. In vivo, retinoids produced pain-related behaviors that were either eliminated or significantly reduced by genetic or pharmacological inhibition of TRPV1 function. These findings identify TRPV1 as an ionotropic receptor for retinoids and provide cellular and molecular insights into retinoid-evoked hypersensitivity. These findings also suggest that selective TRPV1 antagonists are potential therapeutic drugs for treating retinoid-induced sensory hypersensitivity.


Subject(s)
Acitretin/pharmacology , Nicotinic Acids/pharmacology , Nociception/drug effects , TRPV Cation Channels/agonists , Tetrahydronaphthalenes/pharmacology , Action Potentials , Animals , Benzoates/pharmacology , Bexarotene , Binding Sites , Calcitonin Gene-Related Peptide/metabolism , Edema/physiopathology , Ganglia, Spinal/cytology , HEK293 Cells , Hindlimb/drug effects , Hindlimb/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Nociceptors/drug effects , Nociceptors/physiology , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/metabolism , Retinoids/pharmacology , Retinoids/physiology , Signal Transduction , TRPV Cation Channels/chemistry , TRPV Cation Channels/metabolism
11.
Pain ; 154(11): 2297-2309, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23711478

ABSTRACT

Patients receiving opioids for pain may experience decreased effectiveness of the drug and even abnormal pain sensitivity-hyperalgesia and/or allodynia. We hypothesized that peripheral nociceptor hyperexcitability contributes to opioid-induced hyperalgesia and tested this using an in vitro mouse glabrous skin-nerve preparation. Mice were injected intraperitoneally with escalating doses of morphine (5, 8, 10, 15 mg/kg) or saline every 12 hours for 48 hours and killed approximately 12 hours after the last injection. Receptive fields of nociceptors were tested for mechanical, heat, and cold sensitivity. Activity was also measured during an initial 2-minute period and during 5-minute periods between stimuli. Aberrant activity was common in fibers from morphine-treated mice but rare in saline-treated mice. Resting background activity was elevated in C-fibers from morphine-treated mice. Both C- and Aδ-fibers had afterdischarge in response to mechanical, heat, and/or cold stimulation of the skin as well as spontaneous, unevoked activity. Compared to saline, morphine treatment increased the proportion of fibers displaying polymodal rather than mechanical-only responses. A significant increase in Aδ-mechanoreceptive fibers responding to cold accounted for most of this change. In agreement with this, morphine-treated mice showed increased sensitivity in the cold tail flick test. In morphine-treated mice, aberrant activity and hyperexcitability of nociceptors could contribute to increased pain sensitivity. Importantly, this activity is likely driving central sensitization, a phenomenon contributing to abnormal sensory processing and chronic pain. If similar changes occur in human patients, aberrant nociceptor activity is likely to be interpreted as pain and could contribute to opioid-induced hyperalgesia.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Nerve Fibers/drug effects , Neurons, Afferent/drug effects , Nociceptors/drug effects , Skin/innervation , Analgesics, Opioid/administration & dosage , Animals , Cold Temperature , Electrophysiological Phenomena/physiology , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Unmyelinated/drug effects , Physical Stimulation , Reaction Time/physiology , Signal Transduction/drug effects , Skin/drug effects
12.
Neuropsychopharmacology ; 38(8): 1472-84, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23403695

ABSTRACT

Withdrawal from prescribed opioids results in increased pain sensitivity, which prolongs the treatment. This pain sensitivity is attributed to neuroplastic changes that converge at the spinal cord dorsal horn. We have recently reported that repeated morphine administration triggers an insertion of GluA2-lacking (Ca(2+)-permeable) α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPAR) in the hippocampus. This finding together with the reported involvement of AMPAR in the mechanisms underlying inflammatory pain led us to hypothesize a role for spinal AMPAR in opioid-induced pain behavior. Mice treated with escalating doses of morphine showed hypersensitivity to mechanical stimulation. Intrathecal administration of a Ca(2+)-permeable AMPAR selective blocker disrupted morphine-induced mechanical sensitivity. Analysis of the expression and phosphorylation levels of AMPAR subunits (GluA1/2/3/4) in homogenates and in postsynaptic density fractions from spinal cord dorsal horns showed an increase in GluA4 expression and phosphorylation in the postsynaptic density after morphine. Co-immunoprecipitation analyses suggested an increase in GluA4 homomers (Ca(2+)-permeable AMPAR) and immunohistochemical staining localized the increase in GluA4 levels in laminae III-V. The excitatory postsynaptic currents (EPSCs) recorded in laminae III-V showed enhanced sensitivity to Ca(2+)-permeable AMPAR blockers in morphine-treated mice. Furthermore, current-voltage relationships of AMPAR-mediated EPSCs showed that rectification index (an indicator of Ca(2+)-permeable AMPAR contribution) is increased in morphine-treated but not in saline-treated mice. These effects could be reversed by infusion of GluA4 antibody through patch pipette. This is the first direct evidence for a role of GluA4-containing AMPAR in morphine-induced pain and highlights spinal GluA4-containing AMPAR as targets to prevent the morphine-induced pain sensitivity.


Subject(s)
Morphine/administration & dosage , Pain/metabolism , Posterior Horn Cells/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Pain/pathology , Pain Measurement/methods , Posterior Horn Cells/pathology , Synapses/pathology , Treatment Outcome
13.
Curr Neuropharmacol ; 11(6): 652-63, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24396340

ABSTRACT

Chronic pain affects billions of lives globally and is a major public health problem in the United States. However, pain management is still a challenging task due to a lack of understanding of the fundamental mechanisms of pain. In the past decades transient receptor potential (TRP) channels have been identified as molecular sensors of tissue damage and inflammation. Activation/sensitization of TRP channels in peripheral nociceptors produces neurogenic inflammation and contributes to both somatic and visceral pain. Pharmacological and genetic studies have affirmed the role of TRP channels in multiple forms of inflammatory and neuropathic pain. Thus pain-evoking TRP channels emerge as promising therapeutic targets for a wide variety of pain and inflammatory conditions.

14.
Mol Pain ; 8: 47, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22713358

ABSTRACT

BACKGROUND: Central sensitization-associated synaptic plasticity in the spinal cord dorsal horn (SCDH) critically contributes to the development of chronic pain, but understanding of the underlying molecular pathways is still incomplete. Emerging evidence suggests that Wnt signaling plays a crucial role in regulation of synaptic plasticity. Little is known about the potential function of the Wnt signaling cascades in chronic pain development. RESULTS: Fluorescent immunostaining results indicate that ß-catenin, an essential protein in the canonical Wnt signaling pathway, is expressed in the superficial layers of the mouse SCDH with enrichment at synapses in lamina II. In addition, Wnt3a, a prototypic Wnt ligand that activates the canonical pathway, is also enriched in the superficial layers. Immunoblotting analysis indicates that both Wnt3a a ß-catenin are up-regulated in the SCDH of various mouse pain models created by hind-paw injection of capsaicin, intrathecal (i.t.) injection of HIV-gp120 protein or spinal nerve ligation (SNL). Furthermore, Wnt5a, a prototypic Wnt ligand for non-canonical pathways, and its receptor Ror2 are also up-regulated in the SCDH of these models. CONCLUSION: Our results suggest that Wnt signaling pathways are regulated by nociceptive input. The activation of Wnt signaling may regulate the expression of spinal central sensitization during the development of acute and chronic pain.


Subject(s)
Nociception , Wnt Signaling Pathway , Animals , Capsaicin/administration & dosage , Disease Models, Animal , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , HIV Envelope Protein gp120/administration & dosage , Male , Mice , Mice, Inbred C57BL , Neuralgia/metabolism , Neuralgia/pathology , Posterior Horn Cells/metabolism , Posterior Horn Cells/pathology , Protein Transport , Up-Regulation , Wnt Proteins/metabolism , Wnt-5a Protein , Wnt3A Protein/metabolism , beta Catenin/metabolism
15.
J Neurosci ; 31(36): 12727-37, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21900552

ABSTRACT

There is pharmacological evidence that group II and III metabotropic glutamate receptors (mGluRs) function as activity-dependent autoreceptors, inhibiting transmission in supraspinal sites. These receptors are expressed by peripheral nociceptors. We investigated whether mGluRs function as activity-dependent autoreceptors inhibiting pain transmission to the rat CNS, particularly transient receptor potential vanilloid 1 (TRPV1)-induced activity. Blocking peripheral mGluR activity by intraplantar injection of antagonists LY341495 [(2S)-2-amino-2-[(1S,2S)-2-carboxycycloprop-1-yl]-3-(xanth-9-yl) propanoic acid] (LY) (20, 100 µm, group II/III), APICA [(RS)-1-amino-5-phosphonoindan-1-carboxylic acid] (100 µm, group II), or UBP1112 (α-methyl-3-methyl-4-phosphonophenylglycine) (30 µm, group III) increased capsaicin (CAP)-induced nociceptive behaviors and nociceptor activity. In contrast, group II agonist APDC [(2R,4R)-4-aminopyrrolidine-2,4-dicarboxylate] (0.1 µm) or group III agonist l-(+)-2-amino-4-phosphonobutyric acid (l-AP-4) (10 µm) blocked the LY-induced increase. Ca(2+) imaging in dorsal root ganglion (DRG) cells confirmed LY enhanced CAP-induced Ca(2+) mobilization, which was blocked by APDC and l-AP-4. We hypothesized that excess glutamate (GLU) released by high intensity and/or prolonged stimulation endogenously activated group II/III, dampening nociceptor activation. In support of this, intraplantar GLU + LY produced heat hyperalgesia, and exogenous GLU + LY applied to nociceptors produced enhanced nociceptor activity and thermal sensitization. Intraplantar Formalin, known to elevate extracellular GLU, enhanced pain behaviors in the presence of LY. LY alone produced no pain behaviors, no change in nociceptor discharge rate or heat-evoked responses, and no change in cytosolic Ca(2+) in DRG cells, demonstrating a lack of tonic inhibitory control. Group II/III mGluRs maintain an activity-dependent autoinhibition, capable of significantly reducing TRPV1-induced activity. They are endogenously activated after high-frequency and/or prolonged nociceptor stimulation, acting as built-in negative modulators of TRPV1 and nociceptor function, reducing pain transmission to the CNS.


Subject(s)
Nociceptors/physiology , Peripheral Nervous System/physiology , Receptors, Metabotropic Glutamate/physiology , TRPV Cation Channels/physiology , Amino Acids/pharmacology , Animals , Calcium/metabolism , Capsaicin , Cells, Cultured , Cytosol/metabolism , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Formaldehyde , Glutamic Acid/metabolism , Hot Temperature , Male , Pain/chemically induced , Pain Measurement/drug effects , Proline/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Skin/innervation , TRPV Cation Channels/drug effects , Xanthenes/pharmacology
16.
J Neurosci ; 30(44): 14870-82, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-21048146

ABSTRACT

Mechanisms underlying chronic pain that develops after spinal cord injury (SCI) are incompletely understood. Most research on SCI pain mechanisms has focused on neuronal alterations within pain pathways at spinal and supraspinal levels associated with inflammation and glial activation. These events might also impact central processes of primary sensory neurons, triggering in nociceptors a hyperexcitable state and spontaneous activity (SA) that drive behavioral hypersensitivity and pain. SCI can sensitize peripheral fibers of nociceptors and promote peripheral SA, but whether these effects are driven by extrinsic alterations in surrounding tissue or are intrinsic to the nociceptor, and whether similar SA occurs in nociceptors in vivo are unknown. We show that small DRG neurons from rats (Rattus norvegicus) receiving thoracic spinal injury 3 d to 8 months earlier and recorded 1 d after dissociation exhibit an elevated incidence of SA coupled with soma hyperexcitability compared with untreated and sham-treated groups. SA incidence was greatest in lumbar DRG neurons (57%) and least in cervical neurons (28%), and failed to decline over 8 months. Many sampled SA neurons were capsaicin sensitive and/or bound the nociceptive marker, isolectin B4. This intrinsic SA state was correlated with increased behavioral responsiveness to mechanical and thermal stimulation of sites below and above the injury level. Recordings from C- and Aδ-fibers revealed SCI-induced SA generated in or near the somata of the neurons in vivo. SCI promotes the entry of primary nociceptors into a chronic hyperexcitable-SA state that may provide a useful therapeutic target in some forms of persistent pain.


Subject(s)
Action Potentials/physiology , Ganglia, Spinal/physiology , Nociceptors/physiology , Pain/physiopathology , Sensory Receptor Cells/physiology , Spinal Cord Injuries/physiopathology , Animals , Behavior, Animal/physiology , Cells, Cultured , Chronic Disease , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Male , Nociceptors/cytology , Pain/etiology , Pain Measurement/methods , Physical Stimulation/adverse effects , Physical Stimulation/methods , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/cytology , Spinal Cord Injuries/complications
17.
Mol Pain ; 6: 15, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20205720

ABSTRACT

BACKGROUND: Cisplatin is primarily used for treatment of ovarian and testicular cancer. Oxaliplatin is the only effective treatment for metastatic colorectal cancer. Both are known to cause dose related, cumulative toxic effects on the peripheral nervous system and thirty to forty percent of cancer patients receiving these agents experience painful peripheral neuropathy. The mechanisms underlying painful platinum-induced neuropathy remain poorly understood. Previous studies have demonstrated important roles for TRPV1, TRPM8, and TRPA1 in inflammation and nerve injury induced pain. RESULTS: In this study, using real-time, reverse transcriptase, polymerase chain reaction (RT-PCR), we analyzed the expression of TRPV1, TRPM8, and TRPA1 induced by cisplatin or oxaliplatin in vitro and in vivo. For in vitro studies, cultured E15 rat dorsal root ganglion (DRG) neurons were treated for up to 48 hours with cisplatin or oxaliplatin. For in vivo studies, trigeminal ganglia (TG) were isolated from mice treated with platinum drugs for three weeks. We show that cisplatin and oxaliplatin-treated DRG neurons had significantly increased in TRPV1, TRPA1, and TRPM8 mRNA expression. TG neurons from cisplatin treated mice had significant increases in TRPV1 and TRPA1 mRNA expression while oxaliplatin strongly induced only TRPA1. Furthermore, compared to the cisplatin-treated wild-type mice, cisplatin-treated TRPV1-null mice developed mechanical allodynia but did not exhibit enhancement of noxious heat- evoked pain responses. Immunohistochemistry studies showed that cisplatin-treated mice had no change in the proportion of the TRPV1 immunopositive TG neurons. CONCLUSION: These results indicate that TRPV1 and TRPA1 could contribute to the development of thermal hyperalgesia and mechanical allodynia following cisplatin-induced painful neuropathy but that TRPV1 has a crucial role in cisplatin-induced thermal hyperalgesia in vivo.


Subject(s)
Cisplatin/toxicity , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/metabolism , TRPV Cation Channels/metabolism , Animals , Antineoplastic Agents/toxicity , Cells, Cultured , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/physiopathology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociceptors/drug effects , Nociceptors/metabolism , Organoplatinum Compounds/toxicity , Oxaliplatin , Peripheral Nervous System Diseases/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , TRPA1 Cation Channel , TRPM Cation Channels/drug effects , TRPM Cation Channels/metabolism , TRPV Cation Channels/drug effects , TRPV Cation Channels/genetics , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/metabolism
18.
Pain ; 147(1-3): 265-76, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19853381

ABSTRACT

Central neuropathic pain (CNP) developing after spinal cord injury (SCI) is described by the region affected: above-level, at-level and below-level pain occurs in dermatomes rostral, at/near, or below the SCI level, respectively. People with SCI and rodent models of SCI develop above-level pain characterized by mechanical allodynia and thermal hyperalgesia. Mechanisms underlying this pain are unknown and the goals of this study were to elucidate components contributing to the generation of above-level CNP. Following a thoracic (T10) contusion, forelimb nociceptors had enhanced spontaneous activity and were sensitized to mechanical and thermal stimulation of the forepaws 35 days post-injury. Cervical dorsal horn neurons showed enhanced responses to non-noxious and noxious mechanical stimulation as well as thermal stimulation of receptive fields. Immunostaining dorsal root ganglion (DRG) cells and cord segments with activating transcription factor 3 (ATF3, a marker for neuronal injury) ruled out neuronal damage as a cause for above-level sensitization since few C8 DRG cells expressed AFT3 and cervical cord segments had few to no ATF3-labeled cells. Finally, activated microglia and astrocytes were present in thoracic and cervical cord at 35 days post-SCI, indicating a rostral spread of glial activation from the injury site. Based on these data, we conclude that peripheral and central sensitization as well as reactive glia in the uninjured cervical cord contribute to CNP. We hypothesize that reactive glia in the cervical cord release pro-inflammatory substances which drive chronic CNP. Thus a complex cascade of events spanning many cord segments underlies above-level CNP.


Subject(s)
Neuralgia/etiology , Pain Threshold/physiology , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology , Spinal Cord/pathology , Spinal Cord/physiopathology , Action Potentials/physiology , Activating Transcription Factor 3/metabolism , Animals , Behavior, Animal , Cell Count/methods , Disease Models, Animal , Forelimb/physiopathology , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , Hyperalgesia/physiopathology , In Vitro Techniques , Male , Nociceptors/pathology , Nociceptors/physiology , Physical Stimulation/methods , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/physiology , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Statistics, Nonparametric
20.
Brain Res Rev ; 60(1): 202-13, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19154757

ABSTRACT

Not all spinal contusions result in mechanical allodynia, in which non-noxious stimuli become noxious. The studies presented use the NYU impactor at 12.5 mm drop or the Infinite Horizons Impactor (150 kdyn, 1 s dwell) devices to model spinal cord injury (SCI). Both of these devices and injury parameters, if done correctly, will result in animals with above level (forelimb), at level (trunk) and below level (hindlimb) mechanical allodynia that model the changes in evoked somatosensation experienced by the majority of people with SCI. The sections are as follows: 1) Mechanisms of remote microglial activation and pain signaling in "below-level" central pain 2) Intracellular signaling mechanisms in central sensitization in "at-level" pain 3) Peripheral sensitization contributes to "above level" injury pain following spinal cord injury and 4) Role of reactive oxygen species in central sensitization in regional neuropathic pain following SCI. To summarize, differential regional mechanisms contribute to the regional chronic pain states. We propose the importance of understanding the mechanisms in the differential regional pain syndromes after SCI in the chronic condition. Targeting regional mechanisms will be of enormous benefit to the SCI population that suffer chronic pain, and will contribute to better treatment strategies for other chronic pain syndromes.


Subject(s)
Hyperalgesia/physiopathology , Pain, Intractable/physiopathology , Spinal Cord Injuries/physiopathology , Spinal Cord/physiopathology , Chemokine CCL21/metabolism , Gliosis/etiology , Gliosis/physiopathology , Hyperalgesia/etiology , Inflammation/etiology , Inflammation/physiopathology , Microglia/metabolism , Oxidative Stress/physiology , Pain, Intractable/etiology , Reactive Oxygen Species/metabolism , Spinal Cord Injuries/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...