Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 197
Filter
Add more filters










Publication year range
1.
Neuroscience ; 226: 441-50, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-22999972

ABSTRACT

The Reelin-signaling pathway regulates neuronal positioning during embryonic development. Reelin, the extracellular matrix protein missing in reeler mutants, is secreted by neurons in laminae I, II and V, binds to Vldl and Apoer2 receptors on nearby neurons, and tyrosine phosphorylates the adaptor protein Disabled-1 (Dab1), which activates downstream signaling. We previously reported that reeler and dab1 mutants had significantly reduced mechanical and increased heat nociception. Here we extend our analysis to chemical, visceral, and cold pain and importantly, used Fos expression to relate positioning errors in mutant mouse dorsal horn to changes in neuronal activity. We found that noxious mechanical stimulation-induced Fos expression is reduced in reeler and dab1 laminae I-II, compared to wild-type mice. Additionally, mutants had fewer Fos-immunoreactive neurons in the lateral-reticulated area of the deep dorsal horn than wild-type mice, a finding that correlates with a 50% reduction and subsequent mispositioning of the large Dab1-positive cells in the mutant lateral-reticulated area. Furthermore, several of these Dab1 cells expressed Fos in wild-type mice but rarely in reeler mutants. By contrast, paralleling the behavioral observations, noxious heat stimulation evoked significantly greater Fos expression in laminae I-II of reeler and dab1 mutants. We then used the formalin test to show that chemical nociception is reduced in reeler and dab1 mutants and that there is a corresponding decrease in formalin-induced Fos expression. Finally, neither visceral pain nor cold-pain sensitivity differed between wild-type and mutant mice. As differences in the nociceptor distribution within reeler and dab1 mutant dorsal horn were not detected, these differential effects observed on distinct pain modalities suggest that dorsal horn circuits are organized along modality-specific lines.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Nerve Tissue Proteins/physiology , Nociception/physiology , Serine Endopeptidases/physiology , Signal Transduction/physiology , Thermosensing/physiology , Touch Perception/physiology , Animals , Brain Mapping , Cell Adhesion Molecules, Neuronal/genetics , Chemoreceptor Cells/physiology , Cold Temperature , Extracellular Matrix Proteins/genetics , Formaldehyde , Gene Expression/physiology , Genes, fos/genetics , Hot Temperature , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Knockout , Nerve Tissue Proteins/genetics , Nociceptors/physiology , Pain Measurement/drug effects , Physical Stimulation , Reelin Protein , Serine Endopeptidases/genetics , Signal Transduction/genetics , Thermosensing/genetics , Touch Perception/genetics
2.
J Dent Res ; 90(6): 765-70, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21406609

ABSTRACT

Hypersensitivity to thermal and mechanical stimuli can occur in painful pulpitis. To explore the neuro-anatomical basis of heat and mechanical sensitivity, we evaluated expression of TRPV1 (heat) and TRPV2 (heat/mechanical) channels in the cell bodies and terminal arborizations of neurons that innervate the dental pulp (DP) and periodontal tissues (PDL). We report that ~50% of trigeminal ganglion (TG) neurons retrogradely labeled from the DP express TRPV2, and this was significantly greater than the general expression of this channel in the TG (15%) and slightly more than what is expressed in the PDL by retrograde labeling (40%). The TRPV1 receptor, however, was less prevalent in neurons innervating the DP than their general expression in the TG (17% vs. 26%) and was more extensively expressed in neurons innervating the PDL (26%). Co-labeling studies showed that 70% of neurons that innervate the DP are myelinated. Approximately 1/3 of the retrogradely labeled neurons from the DP were calcitonin-gene-related-peptide-positive (peptide-expressing), but very few expressed the IB4 marker of non-peptidergic unmyelinated afferents. These findings suggest that the DP has a unique neurochemical innervation with regard to TRP receptor expression, which has significant implications for the mechanisms contributing to odontogenic pain and management strategies.


Subject(s)
Dental Pulp/innervation , Nociceptors/metabolism , Periodontal Ligament/innervation , TRPV Cation Channels/biosynthesis , Trigeminal Ganglion/metabolism , Animals , Calcitonin Gene-Related Peptide/biosynthesis , Dental Pulp/metabolism , Fluorescent Dyes , Hot Temperature , Male , Mechanotransduction, Cellular/physiology , Periodontal Ligament/metabolism , Rats , Rats, Sprague-Dawley , Stilbamidines , Trigeminal Ganglion/cytology
3.
Cephalalgia ; 30(2): 170-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19489890

ABSTRACT

The association between the clinical use of nitroglycerin (NTG) and headache has led to the examination of NTG as a model trigger for migraine and related headache disorders, both in humans and laboratory animals. In this study in mice, we hypothesized that NTG could trigger behavioural and physiological responses that resemble a common manifestation of migraine in humans. We report that animals exhibit a dose-dependent and prolonged NTG-induced thermal and mechanical allodynia, starting 30-60 min after intraperitoneal injection of NTG at 5-10 mg/kg. NTG administration also induced Fos expression, an anatomical marker of neuronal activity in neurons of the trigeminal nucleus caudalis and cervical spinal cord dorsal horn, suggesting that enhanced nociceptive processing within the spinal cord contributes to the increased nociceptive behaviour. Moreover, sumatriptan, a drug with relative specificity for migraine, alleviated the NTG-induced allodynia. We also tested whether NTG reduces the threshold for cortical spreading depression (CSD), an event considered to be the physiological substrate of the migraine aura. We found that the threshold of CSD was unaffected by NTG, suggesting that NTG stimulates migraine mechanisms that are independent of the regulation of cortical excitability.


Subject(s)
Hyperalgesia/drug therapy , Nitroglycerin/toxicity , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Sumatriptan/pharmacology , Vasodilator Agents/toxicity , Animals , Brain/drug effects , Brain/metabolism , Cortical Spreading Depression/drug effects , Gene Expression/drug effects , Hot Temperature , Hyperalgesia/chemically induced , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Physical Stimulation , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism
4.
Neuroscience ; 163(4): 1220-32, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19647044

ABSTRACT

To better understand the mechanisms through which non-painful and painful stimuli evoke behavior, new resources to dissect the complex circuits engaged by subsets of primary afferent neurons are required. This is especially true to understand the consequences of injury, when reorganization of central nervous system circuits likely contributes to the persistence of pain. Here we describe a transgenic mouse line (ZWX) in which there is Cre-recombinase-dependent expression of a transneuronal tracer, wheat germ agglutinin (WGA), in primary somatic or visceral afferent neurons, but only after transection of their peripheral axons. The latter requirement allows for both regional and temporal control of tracer expression, even in the adult. Using a variety of Cre lines to target WGA transport to subpopulations of sensory neurons, here we demonstrate the extent to which myelinated and unmyelinated "pain" fibers (nociceptors) engage different spinal cord circuits. We found significant convergence (i.e., manifest as WGA-transneuronal labeling) of unmyelinated afferents, including the TRPV1-expressing subset, and myelinated afferents to NK1-receptor-expressing neurons of lamina I. By contrast, PKCgamma interneurons of inner lamina II only receive a myelinated afferent input. This differential distribution of WGA labeling in the spinal cord indicates that myelinated and unmyelinated sensory neurons target different and spatially segregated populations of postsynaptic neurons. On the other hand, we show that neurons of deeper laminae (III-V) receive direct (i.e., monosynaptic) inputs from myelinated afferents and polysynaptic input from unmyelinated afferents. Taken together, our results indicate that peripheral sensory information is transmitted to the central nervous system both through segregated and convergent pathways.


Subject(s)
Genetic Engineering/methods , Neuronal Tract-Tracers/metabolism , Sciatic Nerve/injuries , Sensory Receptor Cells/metabolism , Spinal Cord/metabolism , Synapses/metabolism , Animals , Axotomy , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Integrases/genetics , Mice , Mice, Transgenic , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Unmyelinated/metabolism , Neural Pathways/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peripheral Nerve Injuries , Protein Kinase C/metabolism , TRPV Cation Channels/metabolism , Tetanus Toxin/genetics , Tetanus Toxin/metabolism , Wheat Germ Agglutinins/genetics , Wheat Germ Agglutinins/metabolism
5.
Neuroscience ; 151(1): 164-73, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18063314

ABSTRACT

The transient receptor potential cation channel, vanilloid family, type 2 (TRPV2) is a member of the TRPV family of proteins and is a homologue of the capsaicin/vanilloid receptor (transient receptor potential cation channel, vanilloid family, type 1, TRPV1). Like TRPV1, TRPV2 is expressed in a subset of dorsal root ganglia (DRG) neurons that project to superficial laminae of the spinal cord dorsal horn. Because noxious heat (>52 degrees C) activates TRPV2 in transfected cells this channel has been implicated in the processing of high intensity thermal pain messages in vivo. In contrast to TRPV1, however, which is restricted to small diameter DRG neurons, there is significant TRPV2 immunoreactivity in a variety of CNS regions. The present report focuses on a subset of neurons in the brainstem and spinal cord of the rat including the dorsal lateral nucleus (DLN) of the spinal cord, the nucleus ambiguus, and the motor trigeminal nucleus. Double label immunocytochemistry with markers of motoneurons, combined with retrograde labeling, established that these cells are, in fact, motoneurons. With the exception of their smaller diameter, these cells did not differ from other motoneurons, which are only lightly TRPV2-immunoreactive. As for the majority of DLN neurons, the densely-labeled populations co-express androgen receptor and follow normal DLN ontogeny. The functional significance of the very intense TRPV2 expression in these three distinct spinal cord and brainstem motoneurons groups remains to be determined.


Subject(s)
Medulla Oblongata/physiology , Motor Neurons/physiology , Spinal Cord/physiology , TRPV Cation Channels/physiology , Trigeminal Nucleus, Spinal/physiology , Animals , Brain Stem/cytology , Brain Stem/physiology , Capsaicin/pharmacology , Cell Count , Cell Size , Choline O-Acetyltransferase/metabolism , Female , Immunohistochemistry , Male , Medulla Oblongata/cytology , Motor Neurons/ultrastructure , Nociceptors/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Androgen/biosynthesis , Sex Characteristics , Spinal Cord/cytology , Trigeminal Nucleus, Spinal/cytology
6.
Pain ; 126(1-3): 184-97, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16945484

ABSTRACT

Our previous recordings from dorsal root ganglion and spinal lamina V neurons from TRPV1-mutant mice showed dramatic decreases in responses to temperatures near the activation threshold of this channel (43-49 degrees C). Somewhat unexpectedly, we only observed behavioral deficits in these mice at higher temperatures (50-58 degrees C). In the present study, we tested the hypothesis that the noxious heat-evoked pain behavior that persists in TRPV1-mutant mice reflects residual responsiveness of neurons in the superficial, but not deep, dorsal horn. To this end, we performed in vivo extracellular recordings of spinal nociresponsive neurons in laminae I and V in wild type (WT) and TRPV1 mutant mice. Neurons in WT and mutant mice from both laminae did not differ in their spontaneous activity or evoked responses to mechanical or cold stimuli. By contrast, most lamina I neurons from mutant mice responded to noxious heat with significantly higher thresholds than in WT mice. In contrast, lamina V neurons from mutant mice were virtually unresponsive to noxious heat before and after topical mustard oil-induced tissue injury. Interestingly, lamina I neurons in mutant mice displayed thermal sensitization following tissue injury, comparable in magnitude, but of shorter duration, than in WT mice. We conclude that TRPV1 is necessary for noxious heat-evoked responses of lamina V neurons, both before and after tissue injury. It is also an essential contributor to the normal activation threshold of lamina I neurons to noxious heat and for the full duration of thermal sensitization of lamina I neurons following injury. Finally, our results suggest that the processing of noxious thermal messages by neurons in lamina I involves convergent inputs from a heterogeneous population of primary afferent thermal nociceptors.


Subject(s)
Cold Temperature , Hot Temperature , Pain/physiopathology , Plant Oils/pharmacology , Posterior Horn Cells , Skin/drug effects , Spinal Cord/physiopathology , TRPV Cation Channels/metabolism , Analgesics, Non-Narcotic/pharmacology , Animals , Behavior, Animal , Capsaicin/pharmacology , Electrophysiology , Mice , Mice, Knockout , Mustard Plant , Pain/etiology , Pain/psychology , Pain Threshold , Physical Stimulation , Spinal Cord/pathology , TRPV Cation Channels/genetics
7.
Neuroscience ; 139(4): 1385-96, 2006.
Article in English | MEDLINE | ID: mdl-16580148

ABSTRACT

Mutations in reeler, the gene coding for the Reelin protein, result in pronounced motor deficits associated with positioning errors (i.e. ectopic locations) in the cerebral and cerebellar cortices. In this study we provide the first evidence that the reeler mutant also has profound sensory defects. We focused on the dorsal horn of the spinal cord, which receives inputs from small diameter primary afferents and processes information about noxious, painful stimulation. We used immunocytochemistry to map the distribution of Reelin and Disabled-1 (the protein product of the reeler gene, and the intracellular adaptor protein, Dab1, involved in its signaling pathway) in adjacent regions of the developing dorsal horn, from early to late embryonic development. As high levels of Dab1 accumulate in cells that sustain positioning errors in reeler mutants, our findings of increased Dab1 immunoreactivity in reeler laminae I-III, lamina V and the lateral spinal nucleus suggest that there are incorrectly located neurons in the reeler dorsal horn. Subsequently, we identified an aberrant neuronal compaction in reeler lamina I and a reduction of neurons in the lateral spinal nucleus throughout the spinal cord. Additionally, we detected neurokinin-1 receptors expressed by Dab1-labeled neurons in reeler laminae I-III and the lateral spinal nucleus. Consistent with these anatomical abnormalities having functional consequences, we found a significant reduction in mechanical sensitivity and a pronounced thermal hyperalgesia (increased pain sensitivity) in reeler compared with control mice. As the nociceptors in control and reeler dorsal root ganglia are similar, our results indicate that Reelin signaling is an essential contributor to the normal development of central circuits that underlie nociceptive processing and pain.


Subject(s)
Cell Adhesion Molecules, Neuronal/deficiency , Extracellular Matrix Proteins/deficiency , Gene Expression Regulation, Developmental/physiology , Nerve Tissue Proteins/deficiency , Posterior Horn Cells/physiology , Receptors, Opioid/physiology , Serine Endopeptidases/deficiency , Spinal Cord/cytology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Behavior, Animal , Cell Count/methods , Embryo, Mammalian , Humans , Immunohistochemistry/methods , Male , Mice , Mice, Neurologic Mutants , Nerve Tissue Proteins/metabolism , Pain Measurement/methods , Receptors, Neurokinin-1/metabolism , Reelin Protein , Sex Factors , Spinal Cord/enzymology , Spinal Cord/growth & development , Nociceptin Receptor
8.
Neuroscience ; 128(4): 889-98, 2004.
Article in English | MEDLINE | ID: mdl-15464294

ABSTRACT

Increases in neuronal activity in response to tissue or nerve injury can lead to prolonged functional changes in the spinal cord resulting in an enhancement/sensitization of nociceptive processing. To assess the contribution of alpha-calcium-calmodulin kinase II (alpha-CaMKII) to injury-induced inflammation and pain, we evaluated nociceptive responses in mice that carry a point mutation in the alpha-CaMKII gene at position 286 (threonine to alanine). The mutated protein is unable to autophosphorylate and thus cannot function independently of calcium and calmodulin. Responses to acute noxious stimuli did not differ between alpha-CaMKII T286A mutant and wild type mice. However, the ongoing pain produced by formalin injury was significantly reduced in the mutant mice, as was formalin-evoked spinal Fos-immunoreactivity. In contrast, the decreased mechanical and thermal thresholds associated with nerve injury, Complete Freund's Adjuvant-induced inflammation or formalin-evoked tissue injury were manifest equally in wild-type and mutant mice. Double-labeling immunofluorescence studies revealed that in the mouse alpha-CaMKII is expressed in the superficial dorsal horn as well as in a population of small diameter primary afferent neurons. In summary, our results suggest that alpha-CaMKII, perhaps secondary to an N-methyl-D-aspartate-mediated calcium increase in postsynaptic dorsal horn nociresponsive neurons, is a critical contributor to the spontaneous/ongoing component of tissue-injury evoked persistent pain.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Pain/enzymology , Wounds and Injuries/complications , Animals , Behavior, Animal , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Count/methods , Edema/pathology , Freund's Adjuvant , Ganglia, Spinal/metabolism , Glycoproteins/metabolism , Immunohistochemistry/methods , Intermediate Filament Proteins/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Tissue Proteins/metabolism , Nociceptors/physiology , Oncogene Proteins v-fos/metabolism , Pain/etiology , Pain Measurement , Pain Threshold , Peripherins , Phosphorylation , Physical Stimulation/methods , Protein Kinase C/metabolism , Reaction Time/genetics , Substance P/metabolism , Time Factors , Trigeminal Ganglion/metabolism
9.
Neuroscience ; 125(3): 541-3, 2004.
Article in English | MEDLINE | ID: mdl-15099667

ABSTRACT

Several theories of opioid tolerance predict that the magnitude of micro opioid receptor (MOR) internalization in response to ligand changes in the setting of morphine tolerance. Here we show that in rats rendered tolerant to the analgesic action of morphine, cross-tolerance to the analgesic action of intrathecally administered [d-Ala2,N-MePhe4,Gly-ol5]enkephalin (DAMGO) can be produced without changes in the magnitude of DAMGO-induced internalization of the MOR in lamina II neurons of the rat spinal cord. These results suggest that opioid tolerance-related changes in signaling are located downstream from or in parallel with receptor activation and internalization and support the idea that key features of opioid signaling are maintained, rather than reduced, in the setting of morphine tolerance.


Subject(s)
Drug Tolerance/physiology , Opioid-Related Disorders/metabolism , Posterior Horn Cells/metabolism , Receptors, Opioid, mu/metabolism , Spinal Cord/metabolism , Animals , Endocytosis/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Male , Narcotics/pharmacology , Opioid-Related Disorders/physiopathology , Posterior Horn Cells/cytology , Posterior Horn Cells/drug effects , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/cytology , Spinal Cord/drug effects
10.
Neurosci Lett ; 313(1-2): 57-60, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11684339

ABSTRACT

Previous studies demonstrated that acute irritation of the lower urinary tract (LUT) induces the expression of the immediate early gene, c-fos, in lumbo-sacral spinal cord neurons "J. Neurosci. 12 (1992) 4878" "Am. J. Physiol. 265 (1993) 326" "Somatosens. Mot. Res. 15 (1998) 5". This effect was mediated in part by activation of capsaicin-sensitive bladder afferents "Am. J. Physiol. 265 (1993) 326". Here we investigate the role of preprotachykinin gene products (neurokinin A and substance P) in the response to bladder irritation in urethane-anesthetized mice. Acute irritation of the LUT (intravesical acetic acid) induced smaller numbers of Fos-positive neurons in the spinal cord of mice with a mutated preprotachykinin gene than in wild type mice. Increased Fos expression following LUT irritation or a sham operation in wild type mice was also significantly reduced by pretreatment with the NK2 antagonist, MEN 11420, but Fos expression in mutant mice was not altered by the antagonist. During cystometrograms, a significantly higher percentage (83%) of mutant mice exhibited urinary retention and overflow incontinence as compared to wild type controls. These findings suggest an involvement of tachykinins and NK2 receptors in the response to chemical irritation of the LUT in mice and also suggest that tachykinins contribute to the regulation of normal reflex bladder activity.


Subject(s)
Protein Precursors/genetics , Tachykinins/genetics , Urinary Bladder/physiopathology , Acetic Acid , Animals , Cell Count , Indicators and Reagents , Irritants , Mice , Mice, Knockout , Neurokinin A/metabolism , Neurons/chemistry , Neurons/cytology , Neurons/metabolism , Peptides, Cyclic/pharmacology , Proto-Oncogene Proteins c-fos/analysis , Receptors, Neurokinin-2/antagonists & inhibitors , Spinal Cord/cytology , Stimulation, Chemical , Substance P/metabolism , Urinary Bladder/innervation , Urinary Incontinence/genetics , Urinary Incontinence/physiopathology , Urination/physiology
11.
Nature ; 413(6852): 203-10, 2001 Sep 13.
Article in English | MEDLINE | ID: mdl-11557989

ABSTRACT

The sensation of pain alerts us to real or impending injury and triggers appropriate protective responses. Unfortunately, pain often outlives its usefulness as a warning system and instead becomes chronic and debilitating. This transition to a chronic phase involves changes within the spinal cord and brain, but there is also remarkable modulation where pain messages are initiated - at the level of the primary sensory neuron. Efforts to determine how these neurons detect pain-producing stimuli of a thermal, mechanical or chemical nature have revealed new signalling mechanisms and brought us closer to understanding the molecular events that facilitate transitions from acute to persistent pain.


Subject(s)
Neurons, Afferent/physiology , Pain , Animals , Forecasting , Humans , Signal Transduction
12.
Neuron ; 31(1): 59-73, 2001 Jul 19.
Article in English | MEDLINE | ID: mdl-11498051

ABSTRACT

Cutaneous sensory neurons that detect noxious stimuli project to the dorsal horn of the spinal cord, while those innervating muscle stretch receptors project to the ventral horn. DRG11, a paired homeodomain transcription factor, is expressed in both the developing dorsal horn and in sensory neurons, but not in the ventral spinal cord. Mouse embryos deficient in DRG11 display abnormalities in the spatio-temporal patterning of cutaneous sensory afferent fiber projections to the dorsal, but not the ventral spinal cord, as well as defects in dorsal horn morphogenesis. These early developmental abnormalities lead, in adults, to significantly attenuated sensitivity to noxious stimuli. In contrast, locomotion and sensori-motor functions appear normal. Drg11 is thus required for the formation of spatio-temporally appropriate projections from nociceptive sensory neurons to their central targets in the dorsal horn of the spinal cord.


Subject(s)
Homeodomain Proteins/metabolism , Muscle, Skeletal/innervation , Nerve Tissue Proteins , Neurons, Afferent/physiology , Pain/physiopathology , Posterior Horn Cells/physiology , Skin/innervation , Spinal Cord/physiology , Transcription Factors/metabolism , Afferent Pathways/embryology , Afferent Pathways/physiology , Amino Acid Sequence , Animals , Animals, Newborn , Body Patterning , DNA Probes , Embryo, Mammalian , Embryonic and Fetal Development , Exons , Homeodomain Proteins/genetics , Hot Temperature , Mechanoreceptors/physiology , Mice , Mice, Knockout , Molecular Sequence Data , Motor Activity , Mutagenesis, Site-Directed , Nociceptors/physiology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/deficiency , Transcription Factors/genetics
13.
Nat Med ; 7(7): 821-6, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11433347

ABSTRACT

Using a combined pharmacological and gene-deletion approach, we have delineated a novel mechanism of neurokinin-1 (NK-1) receptor-dependent hyperalgesia induced by proteinase-activated receptor-2 (PAR2), a G-protein-coupled receptor expressed on nociceptive primary afferent neurons. Injections into the paw of sub-inflammatory doses of PAR2 agonists in rats and mice induced a prolonged thermal and mechanical hyperalgesia and elevated spinal Fos protein expression. This hyperalgesia was markedly diminished or absent in mice lacking the NK-1 receptor, preprotachykinin-A or PAR2 genes, or in rats treated with a centrally acting cyclooxygenase inhibitor or treated by spinal cord injection of NK-1 antagonists. Here we identify a previously unrecognized nociceptive pathway with important therapeutic implications, and our results point to a direct role for proteinases and their receptors in pain transmission.


Subject(s)
Hyperalgesia/metabolism , Pain/metabolism , Receptors, Thrombin/metabolism , Animals , Gene Expression Regulation/drug effects , Genes, fos , Inflammation , Male , Mice , Mice, Knockout , Prostaglandins/physiology , Rats , Rats, Wistar , Receptor, PAR-2 , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-1/physiology , Receptors, Thrombin/agonists , Spinal Cord/drug effects , Spinal Cord/metabolism , Substance P/physiology
14.
J Neurosci ; 21(14): 5321-7, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11438608

ABSTRACT

In previous studies we provided evidence that the gamma isoform of protein kinase C (PKCgamma) is an important contributor to the increased pain sensitivity that occurs after injury. Here we combined electrophysiological and behavioral approaches in wild-type and PKCgamma-null mice to compare the hyperexcitability of wide dynamic range neurons in lamina V of the spinal cord dorsal horn with the behavioral hyperexcitability produced by the same injury [application of a C-fiber irritant, mustard oil (MO), to the hindpaw]. Wild-type and null mice did not differ in their response to mechanical or thermal stimuli before tissue injury, and the magnitude of the response to the MO stimuli was comparable. In wild-type mice, MO produced a dramatic and progressive enhancement of the response of lamina V neurons to innocuous mechanical and thermal stimuli. The time course of the neuronal hyperexcitability paralleled the time course of the MO-induced behavioral allodynia (nocifensive behavior in response to a previously innocuous mechanical stimulus). Neuronal hyperexcitability was also manifest in the PKCgamma-null mice, but it lasted <30 min. By contrast, the behavioral allodynia produced by MO in the PKCgamma-null mice, although reduced to approximately half that of the wild-type mice, persisted long after the lamina V hyperexcitability had subsided. Because the MO-induced behavioral allodynia was completely blocked by an NMDA receptor antagonist, we conclude that PKCgamma mediates the transition from short- to long-term hyperexcitability of lamina V nociresponsive neurons but that the persistence of injury-induced pain must involve activity within multiple NMDA-dependent spinal cord circuits.


Subject(s)
Isoenzymes/deficiency , N-Methylaspartate/metabolism , Neuralgia/physiopathology , Protein Kinase C/deficiency , Spinal Cord/physiopathology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Isoenzymes/genetics , Mice , Mice, Knockout , Mustard Plant , Pain Measurement/drug effects , Physical Stimulation , Plant Extracts/pharmacology , Plant Oils , Posterior Horn Cells/physiopathology , Protein Kinase C/genetics , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
15.
Nature ; 411(6840): 957-62, 2001 Jun 21.
Article in English | MEDLINE | ID: mdl-11418861

ABSTRACT

Tissue injury generates endogenous factors that heighten our sense of pain by increasing the response of sensory nerve endings to noxious stimuli. Bradykinin and nerve growth factor (NGF) are two such pro-algesic agents that activate G-protein-coupled (BK2) and tyrosine kinase (TrkA) receptors, respectively, to stimulate phospholipase C (PLC) signalling pathways in primary afferent neurons. How these actions produce sensitization to physical or chemical stimuli has not been elucidated at the molecular level. Here, we show that bradykinin- or NGF-mediated potentiation of thermal sensitivity in vivo requires expression of VR1, a heat-activated ion channel on sensory neurons. Diminution of plasma membrane phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2) levels through antibody sequestration or PLC-mediated hydrolysis mimics the potentiating effects of bradykinin or NGF at the cellular level. Moreover, recruitment of PLC-gamma to TrkA is essential for NGF-mediated potentiation of channel activity, and biochemical studies suggest that VR1 associates with this complex. These studies delineate a biochemical mechanism through which bradykinin and NGF produce hypersensitivity and might explain how the activation of PLC signalling systems regulates other members of the TRP channel family.


Subject(s)
Bradykinin/physiology , Nerve Growth Factor/physiology , Phosphatidylinositol 4,5-Diphosphate/physiology , Receptors, Drug/physiology , Animals , Cell Line , Electrophysiology , Enzyme Activation , Female , Hot Temperature , Male , Mice , Nociceptors/metabolism , Oocytes/physiology , Pain , Protein Kinase C/metabolism , Receptor, trkA/physiology , Receptors, Drug/genetics , Signal Transduction , Type C Phospholipases/physiology , Xenopus laevis
16.
J Neurosci ; 21(10): 3656-64, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11331395

ABSTRACT

Although the tachykinins substance P (SP) and neurokinin A (NKA) are coreleased from primary afferent nociceptors and act via neurokinin (NK) receptors, their differential effects in vivo are not known. Despite pharmacological evidence that NKA preferentially binds NK-2 receptors, this receptor is not found in spinal cord neurons. Thus, in the present studies, we compared the extent to which SP and NKA contribute to spinal nociceptive processing via the NK-1 receptor. We found that SP and NKA induce NK-1 receptor internalization with identical dose dependence and induce increases in intracellular calcium at the same concentrations, suggesting that SP and NKA equally activate the NK-1 receptor. We found, however, that the selective NK-1 receptor antagonist GR 205171 blocked NKA but not SP-induced NK-1 receptor internalization in the rat spinal cord in vivo and in embryonic day 19 rat spinal neurons in vitro. Using this selectivity of GR 205171 for NKA-induced NK-1 receptor activation, we examined the relative contribution of SP and NKA to noxious stimulus-induced activation of spinal NK-1 receptors. We estimate that NKA contributes to at least 50% of the NK-1 receptor activation in lamina I. Under inflammatory conditions, all noxious stimulus-induced NK-1 receptor internalization in deep dorsal horn neurons was blocked by GR 205171, suggesting that it is entirely NKA-mediated. Substance P-mediated NK-1 receptor internalization was focused at the site of termination of stimulated nociceptors but NKA also activated NK-1 receptors at more distant sites. We conclude that NKA not only targets the NK-1 receptor but may be a predominant pronociceptive primary afferent neurotransmitter.


Subject(s)
Neurokinin A/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction/physiology , Spinal Cord/metabolism , Substance P/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Intracellular Fluid/metabolism , Male , Neurokinin A/pharmacology , Neurokinin-1 Receptor Antagonists , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pain Measurement/drug effects , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spinal Cord/cytology , Spinal Cord/drug effects , Substance P/pharmacology , Tetrazoles/pharmacology
17.
Br J Pharmacol ; 132(8): 1827-36, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309255

ABSTRACT

There is evidence that noradrenaline contributes to the development and maintenance of neuropathic pain produced by trauma to a peripheral nerve. It is, however, unclear which subtype(s) of alpha adrenergic receptors (AR) may be involved. In addition to pro-nociceptive actions of AR stimulation, alpha(2) AR agonists produce antinociceptive effects. Here we studied the contribution of the alpha(2) AR subtypes, alpha(2A), alpha(2B) and alpha(2C) to the development of neuropathic pain. We also examined the antinociceptive effect produced by the alpha(2) AR agonist dexmedetomidine in nerve-injured mice. The studies were performed in mice that carry either a point (alpha(2A)) or a null (alpha(2B) and alpha(2C)) mutation in the gene encoding the alpha(2) AR. To induce a neuropathic pain condition, we partially ligated the sciatic nerve and measured changes in thermal and mechanical sensitivity. Baseline mechanical and thermal withdrawal thresholds were similar in all mutant and wild-type mice; and, after peripheral nerve injury, all mice developed comparable hypersensitivity (allodynia) to thermal and mechanical stimulation. Dexmedetomidine reversed the allodynia at a low dose (3 microg kg(-1), s.c.) and produced antinociceptive effects at higher doses (10 - 30 microg kg(-1)) in all groups except in alpha(2A) AR mutant mice. The effect of dexmedetomidine was reversed by intrathecal, but not systemic, injection of the alpha(2) AR antagonist RS 42206. These results suggest that neither alpha(2A), alpha(2B) nor alpha(2C) AR is required for the development of neuropathic pain after peripheral nerve injury, however, the spinal alpha(2A) AR is essential for the antinociceptive effects of dexmedetomidine.


Subject(s)
Analgesics/pharmacology , Pain/physiopathology , Peripheral Nerve Injuries , Receptors, Adrenergic, alpha-2/physiology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Dexmedetomidine/pharmacology , Female , Genotype , Guanethidine , Male , Mice , Point Mutation/genetics , Receptors, Adrenergic, alpha-2/genetics , Sympathectomy, Chemical , Sympatholytics
18.
Neuroscience ; 103(3): 725-37, 2001.
Article in English | MEDLINE | ID: mdl-11274791

ABSTRACT

Internalization of spinal cord neurokinin-1 receptors following noxious stimulation provides a reliable measure of tachykinin signaling. In the present study, we examined the contribution of GABAergic mechanisms to the control of nociceptor processing involving tachykinins. Spinal administration of the GABA(B) receptor agonist R(+)-baclofen in the rat, at antinociceptive doses, significantly reduced the magnitude of neurokinin-1 receptor internalization in neurons of lamina I in response to acute noxious mechanical or thermal stimulation. By contrast, administration of even high doses of the GABA(A) receptor agonists, muscimol or isoguvacine, were without effect. CGP55845, a selective GABA(B) receptor antagonist, completely blocked the effects of baclofen, but failed to increase the incidence of internalization when administered alone. These results provide evidence for a presynaptic control of nociceptive primary afferent neurons by GABA(B) but not GABA(A) receptors in the superficial laminae of the spinal cord, limiting tachykinin release. Because CGP5584 alone did not increase the magnitude of neurokinin-1 receptor internalization observed following noxious stimulation, there appears to be little endogenous activation of GABA(B) receptors on tachykinin-releasing nociceptors under acute stimulus conditions. The contribution of pre- and postsynaptic regulatory mechanisms to GABA(B) receptor-mediated antinociception was also investigated by comparing the effect of baclofen on Fos expression evoked by noxious stimulation to that induced by intrathecal injection of substance P. In both instances, baclofen reduced Fos expression not only in neurons that express the neurokinin-1 receptor, but also in neurons that do not. We conclude that baclofen acts at presynaptic sites to reduce transmitter release from small-diameter nociceptive afferents. Presynaptic actions on non-tachykinin-containing nociceptors could similarly account for the reduction by baclofen of noxious stimulus-induced Fos expression in neurokinin-1 receptor-negative neurons. However, the inhibition of Fos expression induced by exogenous substance P indicates that actions at sites postsynaptic to tachykinin- and/or non-tachykinin-containing primary afferent terminals must also contribute to the antinociceptive actions of GABA(B) receptor agonists.


Subject(s)
Presynaptic Terminals/physiology , Receptors, GABA-B/physiology , Signal Transduction/physiology , Spinal Cord/physiology , Tachykinins/physiology , Animals , Baclofen/pharmacology , Dose-Response Relationship, Drug , GABA-B Receptor Agonists , Male , Neurons/physiology , Nociceptors/physiology , Pain/etiology , Pain/metabolism , Physical Stimulation , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/physiology , Receptors, Neurokinin-1/metabolism , Spinal Cord/cytology
19.
Am J Physiol Regul Integr Comp Physiol ; 280(2): R345-54, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11208561

ABSTRACT

Many studies indicate that blood pressure control systems can attenuate pain (hypoalgesia) of short duration; however, we recently found exaggerated nociceptive responses (hyperalgesia) of persistent duration in the spontaneously hypertensive rat (SHR). Here, we used SHR, Dahl Salt-Sensitive (SS), and normotensive control rats to evaluate the contribution of sustained elevations in arterial pressure to nociceptive responses. Compared with Sprague-Dawley and/or Wistar-Kyoto controls, SHR were 1) hypoalgesic in the hot plate test and 2) hyperalgesic in longer latency tail and paw-withdrawal tests and in two models of inflammatory nociception. These differences were not observed between SS and salt-resistant controls fed a high-salt diet. Inflammatory hyperalgesia in SHR was correlated with neither paw edema nor the number of Fos-positive spinal cord neurons. Our results indicate that "pain" phenotype of the SHR is not restricted to hypoalgesia. This phenotype is related to genetic factors or to the autonomic systems that control blood pressure and not to sustained elevations in blood pressure, differences in spinal neuron activity, or inflammatory edema.


Subject(s)
Blood Pressure/physiology , Hyperalgesia/physiopathology , Hypertension/physiopathology , Pain/physiopathology , Animals , Edema/physiopathology , Hot Temperature , Hypertension/genetics , Inflammation , Male , Neurons/physiology , Rats , Rats, Inbred Dahl , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Spinal Cord/physiology , Spinal Cord/physiopathology , Zymosan
20.
Neurosci Lett ; 291(3): 139-42, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-10984626

ABSTRACT

Nociceptive processing is altered in individuals with inherited hypertension. Because brainstem noradrenergic (NA) neurons have been implicated in both nociceptive transmission and hypertension, we compared behavioral and cardiovascular indices of pain in spontaneously hypertensive rats (SHR) and their normotensive Wistar-Kyoto controls (WKY) after intracerebroventricular administration of an anti-DbetaH-saporin immunotoxin. In WKY rats, NA lesions decreased indices of persistent pain in the formalin test, but did not change nociceptive responses in multiple models of acute pain. In SHR rats, NA lesions did not alter persistent nociception, but decreased thresholds in the hotplate test. We conclude that coeruleospinal inhibitory pathways modulate hypoalgesia but not hyperalgesia in the SHR rat. Brainstem noradrenergic inhibition of acute nociception in the hotplate test is enhanced in the SHR rat, but brainstem noradrenergic contribution to persistent nociceptive processing in the formalin test is reduced in the SHR rat.


Subject(s)
Brain Stem/metabolism , Hypertension/metabolism , Norepinephrine/metabolism , Pain/metabolism , Rats, Inbred SHR/metabolism , Analysis of Variance , Animals , Antibodies, Monoclonal , Behavior, Animal/drug effects , Blood Pressure/drug effects , Brain Stem/drug effects , Formaldehyde/pharmacology , Hot Temperature , Hypertension/genetics , Immunotoxins/administration & dosage , Injections, Intraventricular , Male , Neurons/drug effects , Neurons/metabolism , Pain Measurement/drug effects , Rats , Rats, Inbred WKY , Reaction Time , Ribosome Inactivating Proteins, Type 1 , Saporins , Sensory Thresholds/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...