Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Phys Chem B ; 128(19): 4846-4852, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38700512

ABSTRACT

Epoxy resins are essential for various applications, and their properties depend on the curing reactions during which epoxy and amine compounds form the network structure. We here focus on how the presence or absence of two methyl groups in common epoxy bases, diglycidyl ether of bisphenol A and F (4,4'-DGEBA and 4,4'-DGEBF), affects the curing kinetics. The chemical reactions of both 4,4'-DGEBA and 4,4'-DGEBF, when cured with the same amine, were monitored by Fourier-transform infrared (FT-IR) spectroscopy and differential scanning calorimetry (DSC). Despite no difference in the reactivity of epoxy groups between 4,4'-DGEBA and 4,4'-DGEBF, the initial curing reaction was slower for the latter. This delay for the 4,4'-DGEBF system was attributed to intermolecular stacking, which hindered the approach of unreacted epoxy groups to amino groups and vice versa. This conclusion was drawn from the results obtained through ultraviolet (UV) spectroscopy, wide-angle X-ray scattering (WAXS), density functional theory (DFT) calculation, and all-atom molecular dynamics (MD) simulation.

2.
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
3.
Eur J Pharmacol ; 553(1-3): 120-8, 2006 Dec 28.
Article in English | MEDLINE | ID: mdl-17064685

ABSTRACT

The physiological and pharmacological properties of gamma-aminobutyric acid (GABA)ergic miniature inhibitory postsynaptic currents (mIPSCs) were investigated in substantia gelatinosa neurons of mouse spinal cord using whole-cell patch clamp recordings. Two cell populations were pharmacologically identified based on the effect of propofol (10 muM) on the mIPSC decay kinetics: those exhibiting propofol-sensitive mIPSCs, with a slow decay kinetic (mIPSC(SLOW)), and those exhibiting propofol-resistant mIPSCs, with a fast decay kinetic (mIPSC(FAST)) (decay time constants of 14.2+/-0.7 and 7.4+/-0.8 ms, respectively). The frequency and amplitude of both types of mIPSCs were not affected by propofol. Miniature IPSC(FAST) showed midazolam insensitivity, while midazolam prolonged the decay phase of mIPSC(SLOW) without modulation of the frequency and amplitude. Exogenous GABA-evoked responses in the neurons with mIPSC(SLOW) were potentiated by propofol, while those in neurons with mIPSC(FAST) were unaffected by propofol. Furthermore, non-stationary noise analysis of the two kinetically and pharmacologically distinct mIPSCs revealed different conductance of GABA(A) receptor channels underlying the synaptic events. Pharmacological responses to propofol and midazolam suggested that mIPSC(FAST) and mIPSC(SLOW) in substantia gelatinosa neurons can be mediated by GABA(A) receptors with different subunit compositions.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Neurons/physiology , Substantia Gelatinosa/physiology , gamma-Aminobutyric Acid/physiology , Animals , Drug Resistance , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , GABA Modulators/pharmacology , Hypnotics and Sedatives/pharmacology , In Vitro Techniques , Male , Mice , Midazolam/pharmacology , Neurons/drug effects , Neurons/ultrastructure , Patch-Clamp Techniques , Propofol/pharmacology , Receptors, GABA-A/drug effects , Spinal Cord/drug effects , Substantia Gelatinosa/drug effects
4.
J Comp Neurol ; 498(4): 443-54, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16874807

ABSTRACT

We examined the precise distribution of mRNAs for six cloned rat P2Y receptor subtypes, P2Y1, P2Y2, P2Y4, P2Y6, P2Y12, and P2Y14, in the dorsal root ganglion (DRG) and spinal cord by in situ hybridization histochemistry (ISHH) with 35S-labeled riboprobes. In the DRG, P2Y1 and P2Y2 mRNAs were expressed by 15% and 24% of all neurons, respectively. Although each receptor was evenly distributed between neurofilament-positive and -negative neurons, P2Y2 was rather selectively expressed by TrkA-positive neurons. Schwann cells expressed P2Y2 mRNA, and the nonneuronal cells around the DRG neurons, perhaps the satellite cells, expressed P2Y12 and P2Y14 mRNAs. No ISHH signals for P2Y4 or P2Y6 were seen in any cellular components of the DRG. In the spinal cord, P2Y1 and P2Y4 mRNAs were expressed by some of the dorsal horn neurons, whereas the motor neurons in the ventral horn had P2Y4 and P2Y6 mRNAs. In addition, astrocytes in the gray matter had P2Y1 mRNA, and the microglia throughout the spinal cord expressed P2Y12 mRNA. P2Y14 mRNA was weakly expressed by putative microglia. These findings should provide useful information in interpreting pharmacological and electrophysiological studies in this field given the lack of highly selective antagonists for each P2Y receptor subtype.


Subject(s)
Ganglia, Spinal/metabolism , Neuroglia/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Receptors, Purinergic P2/metabolism , Spinal Cord/metabolism , Animals , Ganglia, Spinal/cytology , Immunohistochemistry , In Situ Hybridization , Male , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/classification , Receptors, Purinergic P2/genetics , Spinal Cord/cytology , Tissue Distribution
5.
Pain ; 123(1-2): 193-203, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16616417

ABSTRACT

Pain during inflammatory joint diseases is enhanced by the generation of hypersensitivity in nociceptive neurons in the peripheral nervous system. To explore the signaling mechanisms of mechanical hypersensitivity during joint inflammation, experimental arthritis was induced by injection of complete Freund's adjuvant (CFA) into the synovial cavity of rat knee joints. As a pain index, the struggle threshold of the knee extension angle was measured. In rats with arthritis, the phosphorylation of extracellular signal-regulated kinase (ERK), induced by passive joint movement, increased significantly in dorsal root ganglion (DRG) neurons innervating the knee joint compared to the naïve rats that received the same movement. The intrathecal injection of a MEK inhibitor, U0126, reduced the phosphorylation of ERK in DRG neurons and alleviated the struggle behavior elicited by the passive movement of the joint. In addition, the injection of U0126 into the joint also reduced the struggle behavior. These findings indicate that the ERK signaling is activated in both cell bodies in DRG neurons and peripheral nerve fibers and may be involved in the mechanical sensitivity of the inflamed joint. Furthermore, the phosphorylated ERK-positive neurons co-expressed the P2X3 receptor, and the injection of TNP-ATP, which antagonizes P2X receptors, into the inflamed joint reduced the phosphorylated ERK and the struggle behavior. Thus, it is suggested that the activation of the P2X3 receptor is involved in the phosphorylation of ERK in DRG neurons and the mechanical hypersensitivity of the inflamed knee joint.


Subject(s)
Arthritis, Experimental/physiopathology , Extracellular Signal-Regulated MAP Kinases/physiology , Osteoarthritis, Knee/physiopathology , Pain/physiopathology , Protein Processing, Post-Translational , Range of Motion, Articular , Receptors, Purinergic P2/physiology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/therapeutic use , Animals , Axonal Transport , Butadienes/therapeutic use , Disease Models, Animal , Freund's Adjuvant/toxicity , Ganglia, Spinal/pathology , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Injections, Intra-Articular , Injections, Spinal , Male , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Nitriles/therapeutic use , Pain/etiology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Purinergic P2 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X3 , Signal Transduction , Stifle/innervation , Stress, Mechanical
6.
Exp Neurol ; 200(1): 112-23, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16546170

ABSTRACT

Patients with neuropathic pain frequently experience hypersensitivity to cold stimulation. However, the underlying mechanisms of this enhanced sensitivity to cold are not well understood. After partial nerve injury, the transient receptor potential ion channel TRPV1 increases in the intact small dorsal root ganglion (DRG) neurons in several neuropathic pain models. In the present study, we precisely examined the incidence of cold hyperalgesia and the changes of TRPA1 and TRPM8 expression in the L4 and L5 DRG following L5 spinal nerve ligation (SNL), because it is likely that the activation of two distinct populations of TRPA1- and TRPM8-expressing small neurons underlie the sensation of cold. We first confirmed that L5 SNL rats developed cold hyperalgesia for more than 14 days after surgery. In the nearby uninjured L4 DRG, TRPA1 mRNA expression increased in trkA-expressing small-to-medium diameter neurons from the 1st to 14th day after the L5 SNL. This upregulation corresponded well with the development and maintenance of nerve injury-induced cold hyperalgesia of the hind paw. In contrast, there was no change in the expression of the TRPM8 mRNA/protein in the L4 DRG throughout the 2-week time course of the experiment. In the injured L5 DRG, on the other hand, both TRPA1 and TRPM8 expression decreased over 2 weeks after ligation. Furthermore, intrathecal administration of TRPA1, but not TRPM8, antisense oligodeoxynucleotide suppressed the L5 SNL-induced cold hyperalgesia. Our data suggest that increased TRPA1 in uninjured primary afferent neurons may contribute to the exaggerated response to cold observed in the neuropathic pain model.


Subject(s)
Calcium Channels/genetics , Cold Temperature , Hyperalgesia/metabolism , Oligonucleotides, Antisense/therapeutic use , Spinal Nerves , TRPM Cation Channels/genetics , Animals , Ankyrins , Calcium Channel Blockers/therapeutic use , Calcium Channels/biosynthesis , Calcium Channels/physiology , Gene Targeting , Hindlimb , Hyperalgesia/genetics , Hyperalgesia/physiopathology , Hyperalgesia/therapy , Ligation , Male , Oligonucleotides, Antisense/genetics , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Spinal Nerves/cytology , Spinal Nerves/metabolism , TRPA1 Cation Channel , TRPC Cation Channels , TRPM Cation Channels/biosynthesis , TRPM Cation Channels/physiology , Time Factors
7.
J Comp Neurol ; 493(4): 596-606, 2005 Dec 26.
Article in English | MEDLINE | ID: mdl-16304633

ABSTRACT

The transient receptor potential (TRP) superfamily of cation channels contains four temperature-sensitive channels, named TRPV1-4, that are activated by heat stimuli from warm to that in the noxious range. Recently, two other members of this superfamily, TRPA1 and TRPM8, have been cloned and characterized as possible candidates for cold transducers in primary afferent neurons. Using in situ hybridization histochemistry and immunohistochemistry, we characterized the precise distribution of TRPA1, TRPM8, and TRPV1 mRNAs in the rat dorsal root ganglion (DRG) and trigeminal ganglion (TG) neurons. In the DRG, TRPM8 mRNA was not expressed in the TRPV1-expressing neuronal population, whereas TRPA1 mRNA was only seen in some neurons in this population. Both A-fiber and C-fiber neurons expressed TRPM8, whereas TRPV1 was almost exclusively seen in C-fiber neurons. All TRPM8-expressing neurons also expressed TrkA, whereas the expression of TRPV1 and TRPA1 was independent of TrkA expression. None of these three TRP channels were coexpressed with TrkB or TrkC. The TRPM8-expressing neurons were more abundant in the TG compared with the DRG, especially in the mandibular nerve region innervating the tongue. Our data suggest heterogeneity of TRPM8 and TRPA1 expression by subpopulations of primary afferent neurons, which may result in the difference of cold-sensitive primary afferent neurons in sensitivity to chemicals such as menthol and capsaicin and nerve growth factor.


Subject(s)
Calcium Channels/metabolism , Ganglia, Spinal/metabolism , Neurons, Afferent/metabolism , TRPM Cation Channels/metabolism , TRPV Cation Channels/metabolism , Trigeminal Ganglion/metabolism , Animals , Ankyrins , Calcium Channels/genetics , Ganglia, Spinal/cytology , Gene Expression , Immunohistochemistry , Male , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Unmyelinated/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, trkA/metabolism , TRPA1 Cation Channel , TRPC Cation Channels , TRPM Cation Channels/genetics , TRPV Cation Channels/genetics , Thermosensing/physiology , Tissue Distribution , Trigeminal Ganglion/cytology
8.
J Clin Invest ; 115(9): 2393-401, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16110328

ABSTRACT

Cold hyperalgesia is a well-documented symptom of inflammatory and neuropathic pain; however, the underlying mechanisms of this enhanced sensitivity to cold are poorly understood. A subset of transient receptor potential (TRP) channels mediates thermosensation and is expressed in sensory tissues, such as nociceptors and skin. Here we report that the pharmacological blockade of TRPA1 in primary sensory neurons reversed cold hyperalgesia caused by inflammation and nerve injury. Inflammation and nerve injury increased TRPA1, but not TRPM8, expression in tyrosine kinase A-expressing dorsal root ganglion (DRG) neurons. Intrathecal administration of anti-nerve growth factor (anti-NGF), p38 MAPK inhibitor, or TRPA1 antisense oligodeoxynucleotide decreased the induction of TRPA1 and suppressed inflammation- and nerve injury-induced cold hyperalgesia. Conversely, intrathecal injection of NGF, but not glial cell line-derived neurotrophic factor, increased TRPA1 in DRG neurons through the p38 MAPK pathway. Together, these results demonstrate that an NGF-induced TRPA1 increase in sensory neurons via p38 activation is necessary for cold hyperalgesia. Thus, blocking TRPA1 in sensory neurons might provide a fruitful strategy for treating cold hyperalgesia caused by inflammation and nerve damage.


Subject(s)
Calcium Channels/metabolism , Cold Temperature , Hyperalgesia , Inflammation , Neurons, Afferent/metabolism , Neurons, Afferent/pathology , Animals , Ankyrins , Calcium Channels/genetics , Ganglia, Spinal/cytology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , In Situ Hybridization , MAP Kinase Signaling System/physiology , Male , Nerve Growth Factor/metabolism , Neurons, Afferent/cytology , Pain Measurement , Rats , Rats, Sprague-Dawley , TRPA1 Cation Channel , TRPC Cation Channels , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
9.
Eur J Neurosci ; 21(9): 2467-74, 2005 May.
Article in English | MEDLINE | ID: mdl-15932604

ABSTRACT

Ca2+/calmodulin-dependent protein kinase II (CaMKII) is known to subserve activity-dependent neuronal plasticity in the central nervous system. To examine in vivo the implication of spinal CaMKII activity in the generation and development of neuropathic pain after peripheral nerve injury, we used an animal model of mononeuropathy, the chronic constriction injury (CCI) model, in the rat. We found that, 3 days after CCI, the total CaMKII (tCaMKII) immunoreactivity increased in the superficial laminae of the spinal cord and this increase continued for up to 14 days. The immunoreactivity of phosphorylated CaMKII showed an increase from 1 day after CCI, which preceded the up-regulation of tCaMKII. A non-selective N-methyl-d-aspartate receptor antagonist, MK801, significantly attenuated the increase of tCaMKII and phosphorylated CaMKII. Moreover, intrathecal administration of an inhibitor of CaMKII, KN93, before the CCI surgery attenuated the development of thermal hyperalgesia and mechanical allodynia. In addition, KN93 significantly reduced the nociceptive behavior in phase II of the formalin test. These findings demonstrate that the activity of CaMKII in spinal neurons is elevated after peripheral nerve injury and may be involved in central sensitization. The alteration of CaMKII is considered to be a neuroplastic change that occurs in spinal neurons that contributes to neuropathic pain, suggesting the potential for the development of novel therapeutics for neuropathic pain that target CaMKII.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mononeuropathies/metabolism , Neuralgia/metabolism , Posterior Horn Cells/enzymology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Chronic Disease , Disease Models, Animal , Male , Nerve Compression Syndromes/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
10.
Brain Res ; 1041(2): 205-11, 2005 Apr 18.
Article in English | MEDLINE | ID: mdl-15829229

ABSTRACT

Proteinase-activated receptors (PARs) are members of the superfamily of G-protein coupled receptors that initiate intracellular signaling by the proteolytic activity of extracellular serine proteases. Three member of this family (PAR-1, PAR-3, and PAR-4) are considered thrombin receptors, whereas PAR-2 is activated by trypsin and tryptase. Recently, activation of PAR-2 signal was identified as a pro-inflammatory factor that mediates peripheral sensitization of nociceptors. Activation of PAR-1 in the periphery is also considered to be a neurogenic mediator of inflammation that is involved in peptide release. Here, we investigated the expression of these four members of PARs in the adult rat dorsal root ganglia (DRG) using radioisotope-labeled in situ hybridization histochemistry. We detected mRNA for all subtypes of PARs in the DRG. Histological analysis revealed the specific expression patterns of the PARs. PAR-1, PAR-2, and PAR-3 mRNA was expressed in 29.0+/-4.0%, 16.0+/-3.2%, and 40.9+/-1.3% of DRG neurons, respectively. In contrast, PAR-4 mRNA was mainly observed in non-neuronal cells. A double-labeling study of PARs with NF-200 and alpha calcitonin gene-related peptide (CGRP) also revealed the distinctive expression of PARs mRNA in myelinated or nociceptive neurons. This study shows the precise expression pattern of PARs mRNA in the DRG and indicates that the cells in DRG can receive modulation with different types of proteinase-activated receptors.


Subject(s)
Ganglia, Spinal/metabolism , Neurons, Afferent/metabolism , Peptide Hydrolases/metabolism , RNA, Messenger/metabolism , Receptor, PAR-1/genetics , Animals , Apoptosis Regulatory Proteins , Calcitonin Gene-Related Peptide/genetics , Carrier Proteins/genetics , Ganglia, Spinal/cytology , Gene Expression/physiology , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Male , Nerve Tissue Proteins , Neurofilament Proteins/genetics , Nociceptors/metabolism , Rats , Rats, Sprague-Dawley , Receptor, PAR-2/genetics
11.
J Comp Neurol ; 481(4): 377-90, 2005 Jan 24.
Article in English | MEDLINE | ID: mdl-15593340

ABSTRACT

The ionotropic purine receptors, P2X receptors, are composed of an assembly of multiple P2X subunits. At present, seven subunits have been cloned and named "P2X1-7." We examined the precise distribution of mRNAs for these subunits in the rat lumbar dorsal root ganglion (DRG) by in situ hybridization histochemistry (ISHH) using riboprobes and characterized their expression among some neuronal subpopulations by ISHH and immunohistochemistry. P2X1 was not expressed by DRG neurons. P2X2 mRNA was preferentially expressed by neurofilament (NF)-200 negative small-sized neurons expressing Ret, but not TrkA or TrkC mRNAs. P2X3 mRNA was mainly expressed by NF-200-negative neurons. Most P2X3-positive neurons had Ret mRNA, and about a half of them coexpressed TrkA and TRPV1 mRNAs. P2X4 was the most ubiquitous subunit, evenly distributing among all examined neuronal subpopulations. P2X5 and P2X6 were expressed by about half of the neurons, and most of these neurons were NF-200-positive. P2X7 mRNA-expressing neurons were quite rare. We further examined the coexpression of all pairs of P2X2-P2X6 mRNAs in DRG neurons and found that: 1) P2X4 was always present in combination with the other subunits. 2) All TrkC neurons had three subunits, P2X4, P2X5, and P2X6, and made up 32% of the total neurons. 3) 12.5% of the total neurons had both P2X2 and P2X3. 4) 12.9% of the neurons had both P2X3 and P2X5. We determined the neuronal subpopulation-specific distribution of P2X subunits in the DRG. These findings suggest possible combinations of subunits of native P2X receptor in DRG neurons.


Subject(s)
Ganglia, Spinal/metabolism , Gene Expression Profiling , Neurons/metabolism , RNA, Messenger/metabolism , Receptors, Purinergic P2/metabolism , Animals , Ganglia, Spinal/cytology , In Situ Hybridization , Ion Channels/metabolism , Lumbar Vertebrae , Male , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Unmyelinated/metabolism , Neurons/cytology , Protein Subunits/classification , Protein Subunits/genetics , Protein Subunits/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Purinergic P2/classification , Receptors, Purinergic P2/genetics , TRPV Cation Channels , Tissue Distribution
12.
Pain ; 113(1-2): 51-60, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15621364

ABSTRACT

Alterations in the intracellular signal transduction pathway in primary afferents may contribute to pain hypersensitivity. We demonstrated that very rapid phosphorylation of p38 mitogen-activated protein kinase occurred in dorsal root ganglion (DRG) neurons that were participating in the transmission of noxious signals. Capsaicin injection induced phosphorylated-p38 (p-p38) in small-to-medium diameter sensory neurons with a peak at 2 min after capsaicin injection. Furthermore, we examined the p-p38 labeling in the DRG after noxious thermal stimuli and found a stimulus intensity-dependent increase in labeled cell size and the number of activated neurons. Most of these p-p38-immunoreactive (IR) neurons were small- and medium-sized neurons, which coexpressed transient receptor potential ion channel TRPV1 and phosphorylated-extracellular signal-regulated protein kinase. Intrathecal administration of the p38 inhibitor, FR167653, reversed the thermal hyperalgesia produced by the capsaicin injection. Inhibition of p38 activation was confirmed by the decrease in the number of p-p38-IR neurons in the DRG following capsaicin injection. Taken together, these findings suggest that the activation of p38 pathways in primary afferents by noxious stimulation in vivo may be, at least in part, correlated with functional activity, and further, involved in the development of thermal hyperalgesia.


Subject(s)
Enzyme Activation/physiology , Ganglia, Spinal/cytology , Hyperalgesia/metabolism , Neurons/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Capsaicin/pharmacology , Cell Count/methods , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Interactions , Hot Temperature , Hyperalgesia/etiology , Immunohistochemistry/methods , Ion Channels/metabolism , Male , Mitogen-Activated Protein Kinase 6/metabolism , Neurofilament Proteins/metabolism , Neurons/drug effects , Pain Measurement/drug effects , Pain Measurement/radiation effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , TRPV Cation Channels , Time Factors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
13.
Eur J Neurosci ; 20(11): 2881-95, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15579142

ABSTRACT

To investigate the intracellular signal transduction pathways involved in the pathophysiological mechanisms of neuropathic pain after partial nerve injury, we examined the activation of extracellular signal-regulated protein kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) in the dorsal root ganglion (DRG) in the chronic constriction injury (CCI) model. The CCI induced an increase in the phosphorylation of ERK in predominantly injured medium-sized and large-sized DRG neurons and in satellite glial cells. Treatment with the MAPK kinase 1/2 inhibitor, U0126, suppressed CCI-induced mechanical allodynia and partially reversed the increase in neuropeptide Y (NPY) expression in damaged DRG neurons. In contrast, the CCI induced the activation of p38, mainly in uninjured small-to-medium-diameter DRG neurons and in satellite glial cells. The p38 inhibitor, SB203580, reversed the CCI-induced heat hyperalgesia and also the increase in brain-derived neurotrophic factor (BDNF) expression in intact DRG neurons. On the other hand, the nerve growth factor (NGF)-induced increase in BDNF expression in small-to-medium-diameter neurons was reversed by SB203580, whereas the anti-NGF-induced increase in NPY in medium-sized and large-sized neurons was partially blocked by U0126. Taken together, our results demonstrate that the activation of ERK and p38 and also the changes in NPY and BDNF expression may occur in different populations of DRG neurons after CCI, partially through alterations in the target-derived NGF. These changes in injured and intact primary afferents are likely to have a substantial role in pathological states, and MAPK pathways in nociceptors may be potential targets for the development of novel analgesics.


Subject(s)
Ganglia, Spinal/cytology , Neurons/enzymology , Sciatic Neuropathy/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Activating Transcription Factor 3 , Animals , Antibodies/pharmacology , Behavior, Animal , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Butadienes/pharmacology , Cell Count/methods , Constriction , Drug Interactions , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Functional Laterality/physiology , Gene Expression Regulation/drug effects , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Imidazoles/pharmacology , Immunohistochemistry/methods , In Situ Hybridization , Male , Nerve Growth Factor/immunology , Nerve Growth Factor/pharmacology , Neurons/cytology , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Nitriles/pharmacology , Pain Measurement/drug effects , Phosphorylation , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors , Transcription Factors/metabolism
14.
Brain Res Mol Brain Res ; 131(1-2): 26-32, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15530649

ABSTRACT

Plasma membrane Ca2+-ATPase (PMCA) is a calcium pump that exists on the plasma membrane and has a role in keeping the intracellular Ca2+ concentration low. In the current study, the expression of PMCA isoforms in spinal cord tissues was investigated in detail and the changes of the expression was examined after contusion injury. Rats received a weight drop on the thoracic spinal cord as the injury or they received a sham surgery as a control. Three or twenty-four hours after spinal cord injury (SCI), the spinal cord was removed and processed for in situ hybridization, reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry. PMCA1-4 mRNAs were expressed in neurons in the control spinal cord. Each isoform of the PMCA proteins showed distinct expression patterns in the spinal cord. PMCA1 and PMCA3 were expressed in all of the layers of gray matter. PMCA2 was also abundant in gray matter, except laminae I and II, while PMCA4 expression was restricted to the superficial layers of the dorsal horn. Distinct expression patterns of the PMCA isoforms suggest differential functions of each isoform in the spinal cord. After spinal cord injury, the expression of PMCA2 was decreased; however, the change in expression of other isoforms showed a tendency of decrease but did not reach a statistically significant level. The decrease in PMCA expression may contribute to the increase in intracellular Ca2+ concentration and PMCA may have a role in secondary injury following spinal cord injury.


Subject(s)
Calcium-Transporting ATPases/genetics , Cell Membrane/enzymology , Spinal Cord Injuries/physiopathology , Spinal Cord/physiopathology , Animals , Calcium-Transporting ATPases/metabolism , Gene Expression Regulation, Enzymologic , In Situ Hybridization , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/enzymology , Spinal Cord Injuries/metabolism , Thoracic Vertebrae
15.
J Neurosci ; 24(45): 10211-22, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15537893

ABSTRACT

To investigate whether activation of mitogen-activated protein kinase (MAPK) in damaged and/or undamaged primary afferents participates in neuropathic pain after partial nerve injury, we examined the phosphorylation of extracellular signal-regulated protein kinase (ERK), p38 MAPK, and c-Jun N-terminal kinase (JNK) in the L4 and L5 dorsal root ganglion (DRG) in the L5 spinal nerve ligation (SNL) model. We first confirmed, using activating transcription factor 3 and neuropeptide Y immunoreactivity, that virtually all L4 DRG neurons are spared from axotomy in this model. In the injured L5 DRG, the L5 SNL induced the activation of ERK, p38, and JNK in different populations of DRG neurons. In contrast, in the uninjured L4 DRG, the L5 SNL induced only p38 activation in tyrosine kinase A-expressing small- to medium-diameter neurons. Intrathecal ERK, p38, and JNK inhibitor infusions reversed SNL-induced mechanical allodynia, whereas only p38 inhibitor application attenuated SNL-induced thermal hyperalgesia. Furthermore, the L5 dorsal rhizotomy did not prevent SNL-induced thermal hyperalgesia. We therefore hypothesized that p38 activation in the uninjured L4 DRG might be involved in the development of heat hypersensitivity in the L5 SNL model. In fact, the treatment of the p38 inhibitor and also anti-nerve growth factor reduced SNL-induced upregulation of brain-derived neurotrophic factor and transient receptor potential vanilloid type 1 expression in the L4 DRG. Together, our results demonstrate that the L5 SNL induces differential activation of MAPK in injured and uninjured DRG neurons and, furthermore, that MAPK activation in the primary afferents may participate in generating pain hypersensitivity after partial nerve injury.


Subject(s)
Ganglia, Spinal/cytology , Hyperalgesia/physiopathology , MAP Kinase Signaling System/physiology , Neurons, Afferent/physiology , Spinal Nerves/injuries , Animals , Anthracenes/pharmacology , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/physiology , Butadienes/pharmacology , Enzyme Activation , Ganglia, Spinal/enzymology , Gene Expression Regulation/physiology , Hot Temperature/adverse effects , Hyperalgesia/etiology , Imidazoles/pharmacology , Ion Channels/biosynthesis , Ion Channels/genetics , Ion Channels/physiology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/physiology , Ligation , Lumbar Vertebrae , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/physiology , Nitriles/pharmacology , Physical Stimulation/adverse effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Rhizotomy , Stress, Mechanical , TRPV Cation Channels , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/physiology
16.
Exp Neurol ; 188(1): 149-60, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15191811

ABSTRACT

To elucidate the role of the degeneration of motor and sensory fibers in neuropathic pain, we examined the pain-related behaviors and the changes of brain-derived neurotrophic factor (BDNF) in the L4/5 dorsal root ganglion (DRG) and the spinal cord after L5 ventral rhizotomy. L5 ventral rhizotomy, producing a selective lesion of motor fibers, produced thermal hyperalgesia and increased BDNF expression in tyrosine kinase A-containing small- and medium-sized neurons in the L5 DRG and their central terminations within the spinal cord, but not in the L4 DRG. Furthermore, L5 ventral rhizotomy up-regulated nerve growth factor (NGF) protein in small to medium diameter neurons in the L5 DRG and also in ED-1-positive cells in the L5 spinal nerve, suggesting that NGF synthesized in the degenerative fibers is transported to the L5 DRG and increases BDNF synthesis. On the other hand, L5 ganglionectomy, producing a selective lesion of sensory fibers, produced heat hypersensitivity and an increase in BDNF and NGF in the L4 DRG. These data indicate that degeneration of L5 sensory fibers distal to the DRG, but not motor fibers, might influence the neighboring L4 nerve fibers and induce neurotrophin changes in the L4 DRG. We suggest that these changes of neurotrophins in the intact primary afferents of neighboring nerves may be one of many complex mechanisms, which can explain the abnormal pain behaviors after nerve injury. The ventral rhizotomy and ganglionectomy models may be useful to investigate the pathophysiological mechanisms of neuropathic pain after Wallerian degeneration in motor or sensory or mixed nerve.


Subject(s)
Ganglia, Spinal/metabolism , Motor Neurons/pathology , Nerve Growth Factors/metabolism , Neuralgia/physiopathology , Neurons, Afferent/pathology , Peripheral Nervous System Diseases/physiopathology , Receptor, trkA , Animals , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/metabolism , Carrier Proteins/metabolism , Cell Size/physiology , Denervation , Disease Models, Animal , Ganglia, Spinal/pathology , Hyperalgesia/metabolism , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Lumbosacral Region , Male , Membrane Proteins/metabolism , Motor Neurons/metabolism , Nerve Growth Factor/metabolism , Neuralgia/metabolism , Neuralgia/pathology , Neurons, Afferent/metabolism , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Rhizotomy , Spinal Nerve Roots/metabolism , Spinal Nerve Roots/pathology , Up-Regulation/physiology , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , Wallerian Degeneration/physiopathology
17.
Pain ; 109(1-2): 64-72, 2004 May.
Article in English | MEDLINE | ID: mdl-15082127

ABSTRACT

Extracellular signal-regulated protein kinase (ERK) is a mitogen-activated protein kinase (MAPK) that mediates several cellular responses to mitogenic and differentiation signals, and activation of ERK in dorsal horn neurons by noxious stimulation is known to contribute to pain hypersensitivity. In order to elucidate the pathophysiological mechanisms of the cauda equina syndrome, secondary to spinal canal stenosis, we evaluated walking dysfunction triggered by forced exercise and activation of ERK in the dorsal horn using a rat model of neuropathic intermittent claudication. Rats in the lumbar canal stenosis (LCS) group showed a shorter running distance from 1 to 14 days after surgery. Two minutes after running on the treadmill apparatus, phosphorylation of ERK was induced in neurons in the superficial laminae in the LCS group but not in the sham group, whereas there was no change in the deeper laminae. Intrathecal administration of the MAPK kinase inhibitor, U0126, 30 min before running, clearly increased the running distance, whereas there was no significant change in the vehicle control group 3 days after surgery. In addition, a prostaglandin E1 analog, OP-1206 alpha-CD, administered orally, improved the walking dysfunction, and further, inhibited activation of ERK following running 7 days after surgery. These findings suggest that intermittent claudication triggered by forced walking might affect the phosphorylation of ERK in the superficial laminae, possibly via transient (partial) ischemia of the spinal cord. ERK activation in the dorsal horn neurons may be involved in the transient pain in the neuropathic intermittent claudication model.


Subject(s)
Alprostadil/analogs & derivatives , Intermittent Claudication/enzymology , Mitogen-Activated Protein Kinases/metabolism , Posterior Horn Cells/enzymology , Alprostadil/administration & dosage , Analysis of Variance , Animals , Butadienes/administration & dosage , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Immunohistochemistry/methods , Intermittent Claudication/drug therapy , Intermittent Claudication/physiopathology , MAP Kinase Signaling System/physiology , Male , Nitriles/administration & dosage , Rats , Rats, Sprague-Dawley , Spinal Stenosis/physiopathology , Walking/physiology
18.
Pain ; 108(3): 258-266, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15030945

ABSTRACT

The mechanism of mechanical hyperalgesia in inflammation might involve a 'mechanochemical' process whereby stretch evokes the release of adenosine 5'-triphosphate (ATP) from the damaged tissue that then excites nearby primary sensory nerve terminals. In the present study, phosphorylated extracellular signal-regulated protein kinase (pERK) immunoreactivity was used as a marker indicating functional activation of primary afferent neurons to examine the P2X receptor-mediated noxious response in DRG neurons in a rat model of peripheral inflammation. We found that very few pERK-labeled DRG neurons were detected in normal rats after alpha, beta methylene-ATP (alphabetame-ATP) intraplantar injection. However, a number of DRG neurons were labeled for pERK after alphabetame-ATP injection to the complete Freund's adjuvant (CFA) induced inflamed paw. Seventy-three percent of pERK-labeled DRG neurons co-expressed the P2X3 receptor. After mechanical noxious stimulation to the hind paw of CFA-inflamed rats, we found many more pERK-labeled neurons compared to those in the normal rats. Administration of the P2X3 receptor antagonists, pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid or 2'- (or 3')-O-(trinitrophenyl)adenosine 5'-triphosphate (TNP-ATP), significantly decreased the mechanical stimulation-evoked pERK labeling in CFA-inflamed rats, but not in normal rats. We also found the recruitment of neurons with myelinated A fibers labeled for pERK in CFA-inflamed rats, which was reversed by P2X3 receptor antagonists. Moreover, TNP-ATP dose dependently reduced the mechanical hypersensitivity of CFA rats. These data suggest that the P2X receptors in primary afferent neurons increase their activity with enhanced sensitivity of the intracellular ERK signaling pathway during inflammation and then contribute to the hypersensitivity to mechanical noxious stimulation in the inflammatory state.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Ganglia, Spinal/cytology , Inflammation/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , Pyridoxal Phosphate/analogs & derivatives , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Count , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Freund's Adjuvant , Functional Laterality/drug effects , Immunohistochemistry/methods , Inflammation/chemically induced , Male , Neurofilament Proteins/metabolism , Phosphorylation , Physical Stimulation , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2 Receptor Agonists , Purinergic P2 Receptor Antagonists , Pyridoxal Phosphate/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X
19.
Eur J Neurosci ; 19(4): 884-90, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15009135

ABSTRACT

It has been suggested that low-threshold sensory pathways have an important role in the formation and maintenance of sensory abnormalities which are observed after peripheral nerve injury. In the present study, we examined the involvement of these pathways in the development of hyperexcitability after sciatic nerve injury (SNI) by detecting the intracellular signal molecule. The rats that received a transection of the sciatic nerve 7 days before were electrically stimulated at 0.1 mA and 3 mA in the proximal region of the nerve injury site. We found a small number of phosphorylated extracellular signal-regulated kinase (pERK)-labelled neurons in laminae I-II and III-IV of the spinal dorsal horn in the control rats after 0.1 mA stimulation. By contrast, there was a marked increased of pERK-labelled neurons both in the superficial laminae and laminae III-IV after the same stimulation in the SNI rats. Enhancement of ERK activation induced by 3 mA stimulation was also observed. Immunoreactivity of pERK in gracile nucleus neurons was also dramatically increased after 0.1 mA stimulation to the injured nerve. These data suggest that the rats with peripheral nerve injury had an increased responsiveness to the low- or high-threshold peripheral stimuli in I-II, III-IV and gracile nucleus neurons. Furthermore, SNI rats that received neonatal capsaicin treatment showed a decreased number of pERK neurons after 0.1 mA stimulation in the dorsal horn and gracile nucleus neurons compared to the control rats. Thus, C-fibres may contribute to the enhanced excitability of the low-threshold sensory neurons after peripheral nerve injury.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Neurons/enzymology , Posterior Horn Cells/enzymology , Sciatic Neuropathy/enzymology , Animals , Capsaicin/pharmacology , Electric Stimulation/methods , Male , Neurons/drug effects , Peripheral Nerve Injuries , Peripheral Nerves/drug effects , Peripheral Nerves/enzymology , Phosphorylation , Posterior Horn Cells/drug effects , Rats , Rats, Sprague-Dawley
20.
Eur J Neurosci ; 19(1): 93-102, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14750967

ABSTRACT

The extracellular protease cascade of plasminogen activators and plasminogen are known to regulate neuronal plasticity and extracellular matrix modification, and to be important factors involved in producing long-term potentiation in the CNS. The purpose of this study is to examine the expression of plasminogen activators in primary afferents and its role in nociceptive pathways after peripheral nerve injury. We found the induction of mRNAs for tissue type plasminogen activator (tPA) and urokinase plasminogen activator (uPA) in the rat dorsal root ganglia following sciatic nerve transection. Immunoreactivity for tPA was increased in laminae I and II of the dorsal horn and, importantly, the increase in proteolytic activity mediated by tPA was observed in the same area. As neither immunoreactivity for uPA nor uPA-mediated proteolysis was observed, we further examined the effects of tPA on dorsal horn excitability and neuropathic pain behaviour. Intrathecal injection of a specific inhibitor of tPA decreased electrical stimulation-induced Fos expression in dorsal horn neurons following axotomy, and also prevented the development of thermal hyperalgesia following partial sciatic nerve ligation. These findings suggest that the increased tPA in the dorsal horn due to mRNA expression in the dorsal root ganglia increases the dorsal horn excitability and has an important role in pain behaviour after peripheral nerve injury. The tPA-mediated hypersensitivity in dorsal horn neurons may be a novel molecular mechanism of neuropathic pain.


Subject(s)
Afferent Pathways/metabolism , Ganglia, Spinal/metabolism , Neuralgia/metabolism , Peripheral Nervous System Diseases/metabolism , Posterior Horn Cells/metabolism , Tissue Plasminogen Activator/genetics , Afferent Pathways/cytology , Afferent Pathways/physiopathology , Animals , Axotomy , Electric Stimulation , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/physiopathology , Hyperalgesia/genetics , Hyperalgesia/metabolism , Immunohistochemistry , Ligation , Male , Neuralgia/genetics , Neuralgia/physiopathology , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/physiopathology , Posterior Horn Cells/cytology , Posterior Horn Cells/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/genetics , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology , Spinal Nerve Roots/cytology , Spinal Nerve Roots/metabolism , Spinal Nerve Roots/physiopathology , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/metabolism , Up-Regulation/genetics , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...