Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Digestion ; 82(3): 150-5, 2010.
Article in English | MEDLINE | ID: mdl-20588026

ABSTRACT

Afferent fibers innervating the gastrointestinal tract have major roles in consciously evoked sensations including pain. However, little is known about the molecules involved in mechanonociception from the upper gastrointestinal tract. We recently reported that activation of extracellular signal-regulated kinase 1/2 (ERK1/2), a member of the mitogen-activated protein kinase cascade in primary afferent neurons, was induced by noxious gastric distention in the rat, and that the activation of ERK1/2 in dorsal root ganglion (DRG) neurons can be implicated in acute visceral pain. Transient receptor potential (TRP) A1, a member of the TRP family of cation channels, was expressed in both DRG and nodose ganglion (NG) neurons innervating the stomach and in nerve fibers in the gastric wall. TRPA1 was coexpressed with ERK1/2 in gastric primary afferent neurons, and attenuation of TRPA1 activation using antisense peptides and a specific blocker led to suppression of both ERK1/2 activation and visceromotor responses. TRPA1 also significantly colocalized with substance P (SP) and calcitonin gene-related peptide (CGRP) in the thoracolumbar DRG, NG and stomach. These data indicate that SP and CGRP may also be released by TRPA1 activation in primary afferent neurons to elicit neurogenic inflammation and promote visceral hyperalgesia.


Subject(s)
Gastric Dilatation/metabolism , Gastric Mucosa/metabolism , Neurogenic Inflammation/metabolism , Nociceptors/metabolism , TRPC Cation Channels/metabolism , Animals , Butadienes , Denervation , Enzyme Activation , Enzyme Inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Ganglia, Spinal/metabolism , Gastric Dilatation/physiopathology , Male , Nitriles , Nodose Ganglion/metabolism , Oligodeoxyribonucleotides, Antisense , Rats , Rats, Sprague-Dawley , Stomach/innervation , Stomach/physiopathology , TRPA1 Cation Channel
2.
J Neurosci ; 28(48): 12775-87, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-19036970

ABSTRACT

Interleukin (IL)-18 is an important regulator of innate and acquired immune responses. Here we show that both the IL-18 and IL-18 receptor (IL-18R), which are induced in spinal dorsal horn, are crucial for tactile allodynia after nerve injury. Nerve injury induced a striking increase in IL-18 and IL-18R expression in the dorsal horn, and IL-18 and IL-18R were upregulated in hyperactive microglia and astrocytes, respectively. The functional inhibition of IL-18 signaling pathways suppressed injury-induced tactile allodynia and decreased the phosphorylation of nuclear factor kappaB in spinal astrocytes and the induction of astroglial markers. Conversely, intrathecal injection of IL-18 induced behavioral, morphological, and biochemical changes similar to those observed after nerve injury. Our results indicate that IL-18-mediated microglia/astrocyte interactions in the spinal cord have a substantial role in the generation of tactile allodynia. Thus, blocking IL-18 signaling in glial cells might provide a fruitful strategy for treating neuropathic pain.


Subject(s)
Astrocytes/immunology , Interleukin-18/immunology , Microglia/immunology , Peripheral Nerve Injuries , Peripheral Nervous System Diseases/immunology , Spinal Cord/immunology , Animals , Hyperalgesia/immunology , Hyperalgesia/physiopathology , Injections, Spinal , Interleukin-18/antagonists & inhibitors , MAP Kinase Signaling System/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Peripheral Nerves/physiopathology , Peripheral Nervous System Diseases/physiopathology , Posterior Horn Cells/immunology , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-18/antagonists & inhibitors , Receptors, Interleukin-18/immunology , Signal Transduction/immunology , Spinal Cord/cytology , Spinal Cord/physiopathology , Up-Regulation/immunology
3.
J Comp Neurol ; 510(2): 188-206, 2008 Sep 10.
Article in English | MEDLINE | ID: mdl-18615542

ABSTRACT

We compared the distribution of the alpha-subunit mRNAs of voltage-gated sodium channels Nav1.1-1.3 and Nav1.6-1.9 and a related channel, Nax, in histochemically identified neuronal subpopulations of the rat dorsal root ganglia (DRG). In the naïve DRG, the expression of Nav1.1 and Nav1.6 was restricted to A-fiber neurons, and they were preferentially expressed by TrkC neurons, suggesting that proprioceptive neurons possess these channels. Nav1.7, -1.8, and -1.9 mRNAs were more abundant in C-fiber neurons compared with A-fiber ones. Nax was evenly expressed in both populations. Although Nav1.8 and -1.9 were preferentially expressed by TrkA neurons, other alpha-subunits were expressed independently of TrkA expression. Actually, all IB4(+) neurons expressed both Nav1.8 and -1.9, and relatively limited subpopulations of IB4(+) neurons (3% and 12%, respectively) expressed Nav1.1 and/or Nav1.6. These findings provide useful information in interpreting the electrophysiological characteristics of some neuronal subpopulations of naïve DRG. After L5 spinal nerve ligation, Nav1.3 mRNA was up-regulated mainly in A-fiber neurons in the ipsilateral L5 DRG. Although previous studies demonstrated that nerve growth factor (NGF) and glial cell-derived neurotrophic factor (GDNF) reversed this up-regulation, the Nav1.3 induction was independent of either TrkA or GFRalpha1 expression, suggesting that the induction of Nav1.3 may be one of the common responses of axotomized DRG neurons without a direct relationship to NGF/GDNF supply.


Subject(s)
Ganglia, Spinal , Neurons/metabolism , Protein Isoforms/metabolism , Sodium Channels/metabolism , Animals , Cell Size , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Horseradish Peroxidase/metabolism , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neurons/cytology , Protein Isoforms/genetics , RNA, Messenger/metabolism , Rats , Receptor, trkA/metabolism , Receptor, trkC/metabolism , Sodium Channels/genetics
4.
Brain Nerve ; 60(5): 483-92, 2008 May.
Article in Japanese | MEDLINE | ID: mdl-18516970

ABSTRACT

Injury to the peripheral nerves often induces produces spontaneous pain, hyperalgesia (increased responsiveness to noxious stimuli), and allodynia (painful responses to normally innocuous stimuli). In contrast to inflammatory pain, the currently available therapeutics for neuropathic pain are either relatively ineffective or accompanied by considerable side effects. Numerous animal models of chronic pain following nerve injury have been introduced. All these neuropathic pain models are generated by partial nerve injury, where a few primary afferents are axotomized, while the others are spared. Among these models, the L5 spinal nerve ligation (SNL) model is unique because in this model, the L4 dorsal root ganglion (DRG) neurons are clearly separated from the axotomized L5 DRG neurons. Previous studies have focused considerable attention on the directly damaged primary afferents and their influence on the activity of the dorsal horn neurons. However, increasing evidence suggests that DRG neurons with intact axons also exhibit alterad excitability and gene expression, and these changes might play functional roles in the pathomechanisms of neuropathic pain. For example, L5 SNL increases the expression of substance P, calcitonin gene-related peptide, brain-derived neurotrophic factor, and the transient receptor potential ion channels TRPV1 and TRPA1 in the uninjured L4 DRG neurons. Furthermore, compelling evidence suggests that the glial cells in the spinal cord may also play a role in the pathogenesis of neuropathic pain. Recent studies have shown that peripheral nerve injury results in the activation of mitogen-activated protein kinases (MAPK) in spinal glial cells and that MAPK inhibitors diminish nerve injury-induced pain hypersensitivity. This review mainly focuses on the DRG neurons and spinal glial cells and will review the roles of MAPK in the nociceptive pathways for neuropathic pain.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Neuralgia/etiology , Neuroglia/physiology , Neurons, Afferent/physiology , Peripheral Nerve Injuries , Spinal Cord/cytology , Animals , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Expression , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nerve Growth Factors/metabolism , Neuralgia/drug therapy , Neuroglia/metabolism , Neuropeptides/metabolism , Transient Receptor Potential Channels/metabolism
5.
Mol Pain ; 4: 17, 2008 Apr 30.
Article in English | MEDLINE | ID: mdl-18445299

ABSTRACT

BACKGROUND: Honeybee's sting on human skin can induce ongoing pain, hyperalgesia and inflammation. Injection of bee venom (BV) into the intraplantar surface of the rat hindpaw induces an early onset of spontaneous pain followed by a lasting thermal and mechanical hypersensitivity in the affected paw. The underlying mechanisms of BV-induced thermal and mechanical hypersensitivity are, however, poorly understood. In the present study, we investigated the role of mitogen-activated protein kinase (MAPK) in the generation of BV-induced pain hypersensitivity. RESULTS: We found that BV injection resulted in a quick activation of p38, predominantly in the L4/L5 spinal dorsal horn ipsilateral to the inflammation from 1 hr to 7 d post-injection. Phosphorylated p38 (p-p38) was expressed in both neurons and microglia, but not in astrocytes. Intrathecal administration of the p38 inhibitor, SB203580, prevented BV-induced thermal hypersensitivity from 1 hr to 3 d, but had no effect on mechanical hypersensitivity. Activated ERK1/2 was observed exclusively in neurons in the L4/L5 dorsal horn from 2 min to 1 d, peaking at 2 min after BV injection. Intrathecal administration of the MEK inhibitor, U0126, prevented both mechanical and thermal hypersensitivity from 1 hr to 2 d. p-ERK1/2 and p-p38 were expressed in neurons in distinct regions of the L4/L5 dorsal horn; p-ERK1/2 was mainly in lamina I, while p-p38 was mainly in lamina II of the dorsal horn. CONCLUSION: The results indicate that differential activation of p38 and ERK1/2 in the dorsal horn may contribute to the generation and development of BV-induced pain hypersensitivity by different mechanisms.


Subject(s)
Bee Venoms/administration & dosage , Extracellular Signal-Regulated MAP Kinases/metabolism , Hyperalgesia/enzymology , Inflammation/enzymology , Spinal Cord/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Bee Venoms/adverse effects , Butadienes/pharmacology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hyperalgesia/chemically induced , Hypersensitivity/metabolism , Imidazoles/pharmacology , Immunohistochemistry , Inflammation/chemically induced , Male , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Pain Threshold/drug effects , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
6.
Gastroenterology ; 134(4): 1094-103, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18395090

ABSTRACT

BACKGROUND & AIMS: Changes in the properties of visceral sensory neurons contribute to the development of gastrointestinal pain. However, little is known about the molecules involved in mechanosensation from the gastrointestinal tract. We investigated the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), a member of the mitogen-activated protein kinase cascade, in dorsal root ganglion (DRG) and nodose ganglion (NG) neurons by noxious gastric distention (GD) and its involvement in acute visceral pain in rats. METHODS: Electromyographic responses to gastric balloon distention through gastrostomy were recorded from the acromiotrapezius muscle in rats after splanchnic nerve resection or vagotomy and in control rats. We then examined the phosphorylated-ERK1/2 (p-ERK1/2) labeling in the DRG and NG after GD using immunohistochemistry. RESULTS: Gastric distention induced p-ERK1/2 in DRG and NG neurons with a peak at 2 minutes after stimulation. We found a stimulus intensity-dependent increase in the number of activated neurons, and this activation corresponded well with the incidence of the visceromotor response. Most of these p-ERK1/2-labeled neurons were small- and medium-sized neurons that coexpressed transient receptor potential vanilloid 1 ion channel and acid-sensing ion channel 3. Splanchnic nerve resection, but not vagotomy, affected the visceromotor response, and attenuated the ERK1/2 activation in DRG neurons produced by GD. Furthermore, intrathecal administration of the mitogen-activated protein kinase kinase 1/2 inhibitor, U0126, altered the response to noxious GD. CONCLUSIONS: The activation of ERK1/2 pathways in DRG neurons by noxious GD may be correlated with functional activity, and may be involved in acute visceral pain.


Subject(s)
Abdominal Pain/enzymology , Catheterization/adverse effects , Mitogen-Activated Protein Kinase 3/metabolism , Neurons, Afferent/enzymology , Stomach/innervation , Abdominal Pain/etiology , Abdominal Pain/physiopathology , Acid Sensing Ion Channels , Acute Disease , Animals , Butadienes/pharmacology , Disease Models, Animal , Electromyography , Enzyme Activation , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/enzymology , Ganglia, Spinal/physiopathology , Gastric Emptying/drug effects , Gastric Emptying/physiology , Immunohistochemistry , Male , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurofilament Proteins/metabolism , Nitriles/pharmacology , Nodose Ganglion/enzymology , Nodose Ganglion/physiopathology , Physical Stimulation , Rats , Rats, Sprague-Dawley , Sodium Channels/metabolism , Stomach/enzymology , Stomach/physiopathology , TRPV Cation Channels/metabolism
7.
J Neurosci ; 28(11): 2892-902, 2008 Mar 12.
Article in English | MEDLINE | ID: mdl-18337420

ABSTRACT

Microglia in the spinal cord may play an important role in the development and maintenance of neuropathic pain. A metabotropic ATP receptor, P2Y(12), has been shown to be expressed in spinal microglia constitutively and be involved in chemotaxis. Activation of p38 mitogen-activated protein kinase (MAPK) occurs in spinal microglia after nerve injury and may be related to the production of cytokines and other mediators, resulting in neuropathic pain. However, it remains unknown whether any type of P2Y receptor in microglia is involved in the activation of p38 MAPK and the pain behaviors after nerve injury. Using the partial sciatic nerve ligation (PSNL) model in the rat, we found that P2Y(12) mRNA and protein increased in the spinal cord and peaked at 3 d after PSNL. Double labeling studies revealed that cells expressing increased P2Y(12) mRNA and protein after nerve injury were exclusively microglia. Both pharmacological blockades by intrathecal administration of P2Y(12) antagonist and antisense knockdown of P2Y(12) expression suppressed the development of pain behaviors and the phosphorylation of p38 MAPK in spinal microglia after PSNL. The intrathecal infusion of the P2Y(12) agonist 2-(methythio) adenosine 5'-diphosphate trisodium salt into naive rats mimicked the nerve injury-induced activation of p38 in microglia and elevated pain behaviors. These data suggest a new mechanism of neuropathic pain, in which the increased P2Y(12) works as a gateway of the following events in microglia after nerve injury. Activation of this receptor by released ATP or the hydrolyzed products activate p38 MAPK pathway and may play a crucial role in the generation of neuropathic pain.


Subject(s)
MAP Kinase Signaling System/physiology , Membrane Proteins/biosynthesis , Microglia/physiology , Neuralgia/metabolism , Receptors, Purinergic P2/biosynthesis , Up-Regulation/physiology , p38 Mitogen-Activated Protein Kinases/biosynthesis , Animals , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Neuralgia/genetics , Pain Measurement/methods , Purinergic P2 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y12 , p38 Mitogen-Activated Protein Kinases/genetics
8.
Brain ; 131(Pt 5): 1241-51, 2008 May.
Article in English | MEDLINE | ID: mdl-18356188

ABSTRACT

Bradykinin is an inflammatory mediator that plays a pivotal role in pain and hyperalgesia in inflamed tissues by exciting and/or sensitizing nociceptors. TRPA1 is an important component of the transduction machinery through which environmental irritants and endogenous proalgesic agents depolarize nociceptors to elicit inflammatory pain. Here, using electrophysiological, immunocytochemical and behavioural analyses, we showed a functional interaction of these two inflammation-related molecules in both heterologous expressing systems and primary sensory neurons. We found that bradykinin increased the TRPA1 currents evoked by allyl isothiocyanate (AITC) or cinnamaldehyde in HEK293 cells expressing TRPA1 and bradykinin receptor 2 (B2R). This potentiation was inhibited by phospholipase C (PLC) inhibitor or protein kinase A (PKA) inhibitor, and mimicked by PLC or PKA activator. The functional interaction between B2R and TRPA1, as well as the modulation mechanism, was also observed in rat dorsal root ganglia neurons. In an occlusion experiment, the PLC activator could enhance AITC-induced TRPA1 current further even in saturated PKA-mediated potentiation, indicating the additive potentiating effects of the PLC and PKA pathways. These data for the first time indicate that a cAMP-PKA signalling is involved in the downstream from B2R in dorsal root ganglia neurons in addition to PLC. Finally, subcutaneous pre-injection of a sub-inflammatory dose of bradykinin into rat hind paw enhanced AITC-induced pain behaviours, which was consistent with the observations in vitro. Collectively, these results represent a novel mechanism through which bradykinin released in response to tissue inflammation might trigger the sensation of pain by TRPA1 activation.


Subject(s)
Bradykinin/physiology , Calcium Channels/metabolism , Inflammation/metabolism , Pain/metabolism , Animals , Ankyrins , Bradykinin/pharmacology , Calcium Channels/physiology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/physiology , Dose-Response Relationship, Drug , Drug Synergism , Ganglia, Spinal/metabolism , Humans , Inflammation/physiopathology , Isothiocyanates/pharmacology , Male , Pain/physiopathology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Signal Transduction , TRPA1 Cation Channel , TRPC Cation Channels , Type C Phospholipases/physiology
9.
J Neurochem ; 105(6): 2249-59, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18363823

ABSTRACT

Toll-like receptors (TLRs) play an essential role in innate immune responses and in the initiation of adaptive immune responses. Microglia, the resident innate immune cells in the CNS, express TLRs. In this study, we show that TLR3 is crucial for spinal cord glial activation and tactile allodynia after peripheral nerve injury. Intrathecal administration of TLR3 antisense oligodeoxynucleotide suppressed nerve injury-induced tactile allodynia, and decreased the phosphorylation of p38 mitogen-activated protein kinase, but not extracellular signal-regulated protein kinases 1/2, in spinal glial cells. Antisense knockdown of TLR3 also attenuated the activation of spinal microglia, but not astrocytes, caused by nerve injury. Furthermore, down-regulation of TLR3 inhibited nerve injury-induced up-regulation of spinal pro-inflammatory cytokines, such as interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha. Conversely, intrathecal injection of the TLR3 agonist polyinosine-polycytidylic acid induced behavioral, morphological, and biochemical changes similar to those observed after nerve injury. Indeed, TLR3-deficient mice did not develop tactile allodynia after nerve injury or polyinosine-polycytidylic acid injection. Our results indicate that TLR3 has a substantial role in the activation of spinal glial cells and the development of tactile allodynia after nerve injury. Thus, blocking TLR3 in the spinal glial cells might provide a fruitful strategy for treating neuropathic pain.


Subject(s)
Hyperesthesia/metabolism , Microglia/metabolism , Spinal Nerves/injuries , Spinal Nerves/metabolism , Toll-Like Receptor 3/physiology , Touch , Animals , Down-Regulation , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 3/biosynthesis
10.
Glia ; 56(7): 723-33, 2008 May.
Article in English | MEDLINE | ID: mdl-18293403

ABSTRACT

Mitogen-activated protein kinase (MAPK) plays an important role in the induction and maintenance of neuropathic pain. Transforming growth factor-activated kinase 1 (TAK1), a member of the MAPK kinase kinase family, is indispensable for the activation of c-Jun N-terminal kinase (JNK) and p38 MAPK. We now show that TAK1 induced in spinal cord astrocytes is crucial for mechanical hypersensitivity after peripheral nerve injury. Nerve injury induced a striking increase in the expression of TAK1 in the ipsilateral dorsal horn, and TAK1 was increased in hyperactive astrocytes, but not in neurons or microglia. Intrathecal administration of TAK1 antisense oligodeoxynucleotide (AS-ODN) prevented and reversed nerve injury-induced mechanical, but not heat hypersensitivity. Furthermore, TAK1 AS-ODN suppressed the activation of JNK1, but not p38 MAPK, in spinal astrocytes. In contrast, there was no change in TAK1 expression in primary sensory neurons, and TAK1 AS-ODN did not attenuate the induction of transient receptor potential ion channel TRPV1 in sensory neurons. Taken together, these results demonstrate that TAK1 upregulation in spinal astrocytes has a substantial role in the development and maintenance of mechanical hypersensitivity through the JNK1 pathway. Thus, preventing the TAK1/JNK1 signaling cascade in astrocytes might provide a fruitful strategy for treating intractable neuropathic pain.


Subject(s)
MAP Kinase Kinase Kinases/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Spinal Cord Injuries/enzymology , Spinal Cord/enzymology , Animals , Down-Regulation , Gene Expression Regulation, Enzymologic , Hindlimb , Kinetics , Male , Oligodeoxyribonucleotides, Antisense , Rats , Rats, Sprague-Dawley , Sensory Thresholds , Spinal Nerves/physiology , Spinal Nerves/physiopathology , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Mol Pain ; 3: 18, 2007 Jul 16.
Article in English | MEDLINE | ID: mdl-17631690

ABSTRACT

The phosphorylation of extracellular signal-regulated kinase (pERK) in DRG and dorsal horn neurons is induced by the C-fiber electrical stimulation to the peripheral nerve. The present study was designed to investigate the expression and modulation of pERK in the rat dorsal horn neurons produced by repetitive electrical stimulation, and its involvement in the electrophysiological activity of dorsal horn neurons. Electrical stimulation of C-fiber intensity at different frequencies was applied to the sciatic nerve; the stimuli-induced pERK expression and the activity in dorsal horn neurons were studied by immunohistochemistry and extracellular recording, respectively. Electrical stimulation of C-fibers (3 mA) induced pERK expression in dorsal horn neurons in a frequency-dependent manner, indicating that the frequency of electrical stimulation is an important factor which activates the intracellular signal pathway in the spinal cord. To demonstrate the underlying mechanism of this frequency-dependent pERK expression, an NMDA receptor antagonist, MK-801, and a voltage sensitive calcium channel antagonist, nifedipine, were administrated intrathecally before the stimulation. We found that high frequency (0.5 Hz and 10 Hz) but not low frequent (0.05 Hz) stimulus-evoked pERK was partially inhibited by MK-801. Both high and low frequency stimulus-evoked pERK were inhibited by the nifedipine treatment. The extracellular single unit activities were recorded from the laminae I-II and V of the L4-5 dorsal horn, and we found that blockage of the intracellular ERK signal suppressed the wind-up responses in a dose-dependent manner. In contrast, any change in the mechanically evoked responses was not observed following the administration of ERK inhibitor. These observations indicate that ERK activation plays an important role in the induction of the wind-up responses in dorsal horn nociceptive neurons.


Subject(s)
Electrophysiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Posterior Horn Cells/enzymology , Sciatic Nerve/physiology , Animals , Electric Stimulation , Male , Phosphorylation , Rats , Rats, Sprague-Dawley
13.
J Clin Invest ; 117(7): 1979-87, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17571167

ABSTRACT

Proinflammatory agents trypsin and mast cell tryptase cleave and activate PAR2, which is expressed on sensory nerves to cause neurogenic inflammation. Transient receptor potential A1 (TRPA1) is an excitatory ion channel on primary sensory nerves of pain pathway. Here, we show that a functional interaction of PAR2 and TRPA1 in dorsal root ganglion (DRG) neurons could contribute to the sensation of inflammatory pain. Frequent colocalization of TRPA1 with PAR2 was found in rat DRG neurons. PAR2 activation increased the TRPA1 currents evoked by its agonists in HEK293 cells transfected with TRPA1, as well as DRG neurons. Application of phospholipase C (PLC) inhibitors or phosphatidylinositol-4,5-bisphosphate (PIP(2)) suppressed this potentiation. Decrease of plasma membrane PIP(2) levels through antibody sequestration or PLC-mediated hydrolysis mimicked the potentiating effects of PAR2 activation at the cellular level. Thus, the increased TRPA1 sensitivity may have been due to activation of PLC, which releases the inhibition of TRPA1 from plasma membrane PIP(2). These results identify for the first time to our knowledge a sensitization mechanism of TRPA1 and a novel mechanism through which trypsin or tryptase released in response to tissue inflammation might trigger the sensation of pain by TRPA1 activation.


Subject(s)
Calcium Channels/metabolism , Nerve Tissue Proteins/metabolism , Receptor, PAR-2/metabolism , Transient Receptor Potential Channels/metabolism , Acrolein/analogs & derivatives , Acrolein/pharmacology , Animals , Ankyrins , Behavior, Animal , Calcium Channels/genetics , Cell Line , Electrophysiology , Enzyme Activation , Ganglia, Spinal/metabolism , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Isothiocyanates/pharmacology , Male , Nerve Tissue Proteins/genetics , Pain/genetics , Pain/metabolism , Pain/pathology , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Receptor, PAR-2/agonists , TRPA1 Cation Channel , TRPC Cation Channels , Transient Receptor Potential Channels/genetics , Type C Phospholipases/metabolism
14.
J Neurochem ; 102(5): 1614-1624, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17573825

ABSTRACT

Heat and cold hyperalgesia is a common feature of inflammatory pain. To investigate whether activation of extracellular signal-regulated protein kinase 5 (ERK5), also known as big mitogen-activated protein kinase 1, in primary sensory neurons participates in inflammatory pain, we examined the phosphorylation of ERK5 in the dorsal root ganglion (DRG) after peripheral inflammation. Inflammation induced by complete Freund's adjuvant produced heat and cold hyperalgesia on the ipsilateral hind paw and induced an increase in the phosphorylation of ERK5, mainly in tyrosine kinase A-expressing small- and medium-size neurons. In contrast, there was no change in ERK5 phosphorylation in the spinal dorsal horn. ERK5 antisense, but not mismatch, oligodeoxynucleotide decreased the activation of ERK5 and suppressed inflammation-induced heat and cold hyperalgesia. Furthermore, the inhibition of ERK5 blocked the induction of transient receptor potential channel TRPV1 and TRPA1 expression in DRG neurons after peripheral inflammation. Our results show that ERK5 activated in DRG neurons contribute to the development of inflammatory pain. Thus, blocking ERK5 signaling in sensory neurons that has the potential for preventing pain after inflammation.


Subject(s)
Ganglia, Spinal/pathology , Hyperalgesia/pathology , Mitogen-Activated Protein Kinase 7/metabolism , Neurons, Afferent/enzymology , Animals , Behavior, Animal , Enzyme Activation/physiology , Freund's Adjuvant , Inflammation/chemically induced , Inflammation/complications , Male , Neurofilament Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Receptor, trkA/metabolism , TRPV Cation Channels/metabolism , Time Factors
15.
J Neurochem ; 102(5): 1569-1584, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17509087

ABSTRACT

Neuropathic pain that occurs after peripheral nerve injury is poorly controlled by current therapies. Increasing evidence shows that mitogen-activated protein kinase (MAPK) play an important role in the induction and maintenance of neuropathic pain. Here we show that activation of extracellular signal-regulated protein kinases 5 (ERK5), also known as big MAPK1, participates in pain hypersensitivity caused by nerve injury. Nerve injury increased ERK5 phosphorylation in spinal microglia and in both damaged and undamaged dorsal root ganglion (DRG) neurons. Antisense knockdown of ERK5 suppressed nerve injury-induced neuropathic pain and decreased microglial activation. Furthermore, inhibition of ERK5 blocked the induction of transient receptor potential channels and brain-derived neurotrophic factor expression in DRG neurons. Our results show that ERK5 activated in spinal microglia and DRG neurons contributes to the development of neuropathic pain. Thus, blocking ERK5 signaling in the spinal cord and primary afferents has potential for preventing pain after nerve damage.


Subject(s)
Ganglia, Spinal/pathology , Microglia/enzymology , Mitogen-Activated Protein Kinase 7/physiology , Neuralgia/pathology , Neurons, Afferent/enzymology , Animals , Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Functional Laterality , Hyperalgesia/physiopathology , Male , Nerve Tissue Proteins/metabolism , Neuralgia/etiology , Nitriles/pharmacology , Oligonucleotides, Antisense/pharmacology , Pain Measurement/methods , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/pathology , Rats , Rats, Sprague-Dawley , Spinal Nerves/pathology , TRPV Cation Channels/metabolism , Time Factors
16.
Eur J Neurosci ; 25(4): 1097-111, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17331206

ABSTRACT

The cell adhesion molecule L1 (L1-CAM) plays important functional roles in the developing and adult nervous systems. Here we show that peripheral nerve injury induced dynamic post-transcriptional alteration of L1-CAM in the rat dorsal root ganglia (DRGs) and spinal cord. Sciatic nerve transection (SCNT) changed the expression of L1-CAM protein but not L1-CAM mRNA. In DRGs, SCNT induced accumulation of the L1-CAM into the surface of somata, which resulted in the formation of immunoreactive ring structures in a number of unmyelinated C-fiber neurons. These neurons with L1-CAM-immunoreactive ring structures were heavily colocalized with phosphorylated p38 MAPK. Western blot analysis revealed the increase of full-length L1-CAM and decrease of fragments of L1-CAM after SCNT in DRGs. Following SCNT, L1-CAM-immunoreactive profiles in the dorsal horn showed an increase mainly in pre-synaptic areas of laminae I-II with a delayed onset and colocalized with growth-associated protein 43. In contrast to DRGs, SCNT increased the proteolytic 80-kDa fragment of L1-CAM and decreased full-length L1-CAM in the spinal cord. The intrathecal injection of L1-CAM antibody for the extracellular domain of L1-CAM inhibited activation of p38 MAPK and emergence of ring structures of L1-CAM immunoreactivity in injured DRG neurons. Moreover, inhibition of extracellular L1-CAM binding by intrathecal administration of antibody suppressed the mechanical allodynia and thermal hyperalgesia induced by partial SCNT. Collectively, these data suggest that the modification of L1-CAM in nociceptive pathways might be an important pathomechanism of neuropathic pain.


Subject(s)
Gene Expression Regulation/physiology , Neural Cell Adhesion Molecule L1/metabolism , Neurons, Afferent/metabolism , Sciatica/pathology , Sciatica/physiopathology , Animals , Cell Count , Ganglia, Spinal/pathology , Male , Nerve Tissue Proteins/metabolism , Neurons, Afferent/classification , Rats , Rats, Sprague-Dawley , Time Factors
17.
Glia ; 55(6): 595-603, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17299772

ABSTRACT

Dorsal root injury is known to induce alteration of the extracellular environment in the spinal cord and synaptic reorganization with degradation of injured primary afferent and sprouting of spared terminal. These changes affect behavioral sensitivity and sometimes lead to neuropathic pain. We have hypothesized that changes in extracellular proteolysis in the dorsal horn is involved in neuroplastic changes in the dorsal horn after nerve injury. Tissue type plasminogen activator (tPA) is a well-known extracellular serine protease and is involved in the modification of the extracellular matrix, which leads to neuroplastic changes such as long-term potentiation in the hippocampus. In the present study, we found a marked induction of tPA in activated astrocytes following L4/5 root injury and a resultant increase of proteolytic enzymatic activity in the dorsal horn. We also examined the involvement of tPA activity on mechanical hypersensitivity using a root ligation model which has been used for investigating radiculopathy pain behavior. Intrathecal and continuous administration of tPA inhibitor, tPA-STOP, suppressed root ligation-induced mechanical allodynia in a dose-dependent manner during an early stage of injury (0-4 days). In contrast, the delayed administration of tPA-STOP during the chronic stage of injury (10 days) did not affect pain behavior. These data suggest an important contribution of astrocytes in the dorsal horn to the pathophysiology of radiculopathy pain, and astrocyte-derived tPA and the proteolytic activity in the dorsal horn may be one of the essential factors involved in pain following root injury.


Subject(s)
Astrocytes/metabolism , Hyperalgesia/metabolism , Posterior Horn Cells/metabolism , Radiculopathy/metabolism , Spinal Nerve Roots/metabolism , Tissue Plasminogen Activator/metabolism , Animals , Astrocytes/cytology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins/metabolism , Hyperalgesia/physiopathology , Ligation , Male , Peptide Hydrolases/metabolism , Posterior Horn Cells/cytology , Radiculopathy/physiopathology , Rats , Rats, Sprague-Dawley , Rhizotomy , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology , Spinal Nerve Roots/injuries , Spinal Nerve Roots/physiopathology , Tissue Plasminogen Activator/antagonists & inhibitors , Up-Regulation/physiology
18.
J Pharmacol Exp Ther ; 321(1): 28-34, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17237256

ABSTRACT

Alterations in the intracellular signal transduction pathway in primary afferents may contribute to pain hypersensitivity. Recently, we have reported that the phosphorylation of extracellular signal-regulated protein kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase (MAPK) occurs in primary afferent neurons in response to noxious stimulation of the peripheral tissue, i.e., activity-dependent activation of ERK1/2 and p38 MAPK in dorsal root ganglion (DRG) neurons. In the present study, we investigated the phosphorylation of ERK5, also known as big MAPK1, in the DRG by noxious stimulation using immunohistochemistry. Capsaicin injection induced phosphorylated ERK5 (p-ERK5) in small-to-medium diameter sensory neurons with a peak at 2 min after capsaicin injection. Furthermore, we examined the p-ERK5 labeling in the DRG after noxious heat and cold stimuli and found a stimulus intensity-dependent increase in the number of activated neurons. Most of these p-ERK5-immunoreactive neurons were small- and medium-sized neurons, which coexpressed transient receptor potential (TRP) ion channel TRPV1 and TRPA1 after noxious heat and cold stimuli, respectively. In contrast, there was no change in ERK5 phosphorylation in the spinal dorsal horn. The i.t. administration of ERK5 antisense oligodeoxynucleotide reversed heat hyperalgesia, but not mechanical allodynia, produced by capsaicin injection. Taken together, these findings suggest that the in vivo activation of the ERK5 signaling pathway in sensory neurons by noxious stimulation may be, at least in part, correlated with functional activity and, further, involved in the development of pain hypersensitivity.


Subject(s)
Hyperalgesia/physiopathology , Mitogen-Activated Protein Kinase 6/metabolism , Neurons, Afferent/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Blotting, Western , Capsaicin/pharmacology , Cold Temperature , Enzyme Activation/physiology , Hot Temperature , Hyperalgesia/chemically induced , Immunohistochemistry , In Situ Hybridization , Male , Mitogen-Activated Protein Kinase 6/biosynthesis , Phosphorylation , Posterior Horn Cells/drug effects , Posterior Horn Cells/physiology , RNA, Antisense/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , TRPV Cation Channels/drug effects , TRPV Cation Channels/physiology
19.
Neuroreport ; 18(1): 17-22, 2007 Jan 08.
Article in English | MEDLINE | ID: mdl-17259854

ABSTRACT

The expression of plasma membrane Ca(2+)-ATPase, a calcium pump located in cell membrane regulating intracellular Ca(2+) levels by Ca(2+) extrusion from cells, was examined in dorsal root ganglion neurons in naive rats and after spinal nerve ligation. The mRNAs and proteins in plasma membrane Ca(2+)-ATPase 1-3 were expressed in all size neurons with intense labeling in medium to large neurons. After spinal nerve ligation, these three isoforms showed downregulation of their expression. In contrast, plasma membrane Ca(2+)-ATPase 4 was expressed mainly in small neurons, and the number and signal intensity were significantly increased after spinal nerve ligation. These data suggest that plasma membrane Ca(2+)-ATPase isoforms have a distinct pattern of expression and regulation by axotomy in dorsal root ganglion neurons in normal and pathological conditions.


Subject(s)
Axotomy , Gene Expression Regulation/physiology , Neurons, Afferent/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Animals , Ganglia, Spinal/pathology , Immunohistochemistry , In Situ Hybridization/methods , Male , Plasma Membrane Calcium-Transporting ATPases/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Spinal Cord/pathology , Time Factors
20.
J Neurosci ; 26(46): 11974-86, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108171

ABSTRACT

The p75 neurotrophin receptor (p75NTR) has been implicated in diverse neuronal responses, including survival, cell death, myelination, and inhibition of regeneration. However, the role of p75NTR in neuropathic pain, for which there is currently no effective therapy, has not been explored. Here, we report that the pharmacological blockade of p75NTR in primary sensory neurons reversed neuropathic pain after nerve injury. Nerve injury increased the expression and axonal transport of p75NTR and phosphorylation of TrkA in the uninjured primary afferents. Functional inhibition of p75NTR suppressed injury-induced neuropathic pain and decreased the phosphorylation of TrkA and p38 mitogen-activated protein kinase, and the induction of transient receptor potential channels in dorsal root ganglion (DRG) neurons. Our results show that p75NTR induced in undamaged DRG neurons facilitates TrkA signaling and contributes to heat and cold hyperalgesia.


Subject(s)
Hyperalgesia/drug therapy , Neuralgia/drug therapy , Neurons, Afferent/drug effects , Peripheral Nervous System Diseases/drug therapy , Receptor, Nerve Growth Factor/antagonists & inhibitors , Spinal Nerves/drug effects , Animals , Axonal Transport/drug effects , Axonal Transport/physiology , Denervation , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Ligation , Male , Neuralgia/metabolism , Neuralgia/physiopathology , Neurons, Afferent/metabolism , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/physiopathology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Spinal Nerves/injuries , Spinal Nerves/physiopathology , TRPV Cation Channels/drug effects , TRPV Cation Channels/metabolism , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...