Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Osteoarthritis Cartilage ; 30(12): 1616-1630, 2022 12.
Article in English | MEDLINE | ID: mdl-36075514

ABSTRACT

OBJECTIVE: To investigate whether Ccr2 inactivation in aggrecan-expressing cells induced before post-traumatic OA (PTOA) onset or during progression, improves joint structures, synovial thickness and pain. DESIGN: We induced a Ccr2 deletion in aggrecan-expressing cells (CCR2-AggKO) in skeletally mature mice using a tamoxifen-inducible Ccr2 inactivation. We stimulated PTOA changes (destabilization of medial meniscus, DMM) in CCR2-AggKO and CCR2+/+ mice, inducing recombination before DMM or 4 wks after DMM (early-vs late-inactivation). Joint damage was evaluated 2, 4, 8, 12 wks post-DMM using multiple scores: articular-cartilage structure (ACS), Safranin-O, histomorphometry, osteophyte size/maturity, subchondral bone thickness and synovial hyperplasia. Spontaneous (incapacitance meter) and evoked pain (von-Frey filaments) were assessed up to 20 wks. RESULTS: Early aggrecan-Ccr2 inactivation in CCR2-AggKO mice (N=8) resulted in improved ACS score (8-12wk, P=0.002), AC area (4-12wk, P<0.05) and Saf-O score (2wks P=0.004, 4wks P=0.02, 8-12wks P=0.002) compared to CCR2+/+. Increased subchondral bone thickness was delayed only at 2 wks and exclusively following early recombination. Osteophyte size was not affected, but osteophyte maturation (cartilage-to-bone) was delayed (4wks P=0.04; 8 wks P=0.03). Although late aggrecan-Ccr2 deletion led to some cartilage improvement, most data did not reach statistical significance; osteophyte maturity was delayed at 12wks. Early aggrecan-Ccr2 deletion led to improved pain measures of weight bearing compared to CCR2+/+ mice (N = 9, 12wks diff 0.13 [0.01, 0.26], 16wks diff 0.15 [0.05, 0.26], 20wks diff 0.23 [0.14, 0.31]). Improved mechanosensitivity in evoked pain, although less noticeable, was detected. CONCLUSIONS: We demonstrated that deletion of Ccr2 in aggrecan expressing cells reduces the initiation but not progression of OA.


Subject(s)
Cartilage, Articular , Knee Injuries , Osteoarthritis , Osteophyte , Mice , Animals , Aggrecans/genetics , Disease Models, Animal , Osteoarthritis/genetics , Cartilage, Articular/injuries , Pain/genetics , Receptors, CCR2/genetics
2.
Osteoarthritis Cartilage ; 25(6): 914-925, 2017 06.
Article in English | MEDLINE | ID: mdl-27856294

ABSTRACT

OBJECTIVE: We previously found in our embryonic studies that proper regulation of the chemokine CCL12 through its sole receptor CCR2, is critical for joint and growth plate development. In the present study, we examined the role of CCR2 in injury-induced-osteoarthritis (OA). METHOD: We used a murine model of injury-induced-OA (destabilization of medial meniscus, DMM), and systemically blocked CCR2 using a specific antagonist (RS504393) at different times during disease progression. We examined joint degeneration by assessing cartilage (cartilage loss, chondrocyte hypertrophy, MMP-13 expression) and bone lesions (bone sclerosis, osteophytes formation) with or without the CCR2 antagonist. We also performed pain behavioral studies by assessing the weight distribution between the normal and arthritic hind paws using the IITS incapacitance meter. RESULTS: Testing early vs delayed administration of the CCR2 antagonist demonstrated differential effects on joint damage. We found that OA changes in articular cartilage and bone were ameliorated by pharmacological CCR2 blockade, if given early in OA development: specifically, pharmacological targeting of CCR2 during the first 4 weeks (wks) following injury, reduced OA cartilage and bone damage, with less effectiveness with later treatments. Importantly, our pain-related behavioral studies showed that blockade of CCR2 signaling during early, 1-4 wks post-surgery or moderate, 4-8 wks post-surgery, OA was sufficient to decrease pain measures, with sustained improvement at later stages, after treatment was stopped. CONCLUSIONS: Our data highlight the potential efficacy of antagonizing CCR2 at early stages to slow the progression of post-injury OA and, in addition, improve pain symptoms.


Subject(s)
Benzoxazines/pharmacology , Bone and Bones/drug effects , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Menisci, Tibial/drug effects , Osteoarthritis/pathology , Receptors, CCR2/antagonists & inhibitors , Spiro Compounds/pharmacology , Animals , Bone and Bones/pathology , Disease Models, Animal , Disease Progression , Hypertrophy , Matrix Metalloproteinase 13/drug effects , Matrix Metalloproteinase 13/metabolism , Menisci, Tibial/surgery , Mice , Osteoarthritis/metabolism , Osteophyte , Receptors, CCR2/physiology , Sclerosis , Tibial Meniscus Injuries
3.
Br J Pharmacol ; 168(4): 822-34, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22946657

ABSTRACT

BACKGROUND AND PURPOSE: Although it is established that the receptor activity modifying proteins (RAMPs) can interact with a number of GPCRs, little is known about the consequences of these interactions. Here the interaction of RAMPs with the glucagon-like peptide 1 receptor (GLP-1 receptor), the human vasoactive intestinal polypeptide/pituitary AC-activating peptide 2 receptor (VPAC(2)) and the type 1 corticotrophin releasing factor receptor (CRF(1)) has been examined. EXPERIMENTAL APPROACH: GPCRs were co-transfected with RAMPs in HEK 293S and CHO-K1 cells. Cell surface expression of RAMPs and GPCRs was examined by ELISA. Where there was evidence for interactions, agonist-stimulated cAMP production, Ca(2+) mobilization and GTPγS binding to G(s), G(i), G(12) and G(q) were examined. The ability of CRF to stimulate adrenal corticotrophic hormone release in Ramp2(+/-) mice was assessed. KEY RESULTS: The GLP-1 receptor failed to enhance the cell surface expression of any RAMP. VPAC(2) enhanced the cell surface expression of all three RAMPs. CRF(1) enhanced the cell surface expression of RAMP2; the cell surface expression of CRF(1) was also increased. There was no effect on agonist-stimulated cAMP production. However, there was enhanced G-protein coupling in a receptor and agonist-dependent manner. The CRF(1) : RAMP2 complex resulted in enhanced elevation of intracellular calcium to CRF and urocortin 1 but not sauvagine. In Ramp2(+/-) mice, there was a loss of responsiveness to CRF. CONCLUSIONS AND IMPLICATIONS: The VPAC(2) and CRF(1) receptors interact with RAMPs. This modulates G-protein coupling in an agonist-specific manner. For CRF(1), coupling to RAMP2 may be of physiological significance.


Subject(s)
Receptor Activity-Modifying Proteins/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Adrenocorticotropic Hormone/blood , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Enzyme-Linked Immunosorbent Assay , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , Protein Binding , Real-Time Polymerase Chain Reaction , Receptor Activity-Modifying Protein 2/genetics , Receptor Activity-Modifying Protein 2/metabolism , Transfection
4.
Osteoarthritis Cartilage ; 17(2): 244-51, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18684647

ABSTRACT

OBJECTIVE: The vanilloid receptor transient receptor potential vanilloid 1 (TRPV1), expressed by sensory neurons that innervate joints, is implicated in arthritis but the mechanisms are not fully understood. One possibility is that downstream effects of activation of TRPV1 are mediated by the extracellularly-regulated kinase (ERK). ERK is phosphorylated (p-ERK) in sensory neurons in response to noxious stimuli and its inhibition has been found to be antinociceptive in several pain models. We here wanted to ascertain whether TRPV1 may contribute to the pain hypersensitivity and inflammation of arthritis via an ERK-mediated pathway. METHODS: We used a model of adjuvant-induced arthritis (AIA) of the ankle and investigated the changes in expression of p-ERK in sensory afferent neurons in dorsal root ganglia (DRG) and spinal dorsal horn of TRPV1-knockout (KO) mice, compared to wild-type (WT) mice of the same genetic background, using multiple immunofluorescence. RESULTS: Two to three weeks after inducing AIA in mice, the number of neurons in DRG and spinal cord that expressed p-ERK was significantly higher on the side of AIA than on the contralateral, vehicle-injected side. The fraction of p-ERK-positive neurons in the DRG that also expressed TRPV1 was increased, indicating that activation of ERK occurred preferentially in TRPV1-positive neurons. Moreover, TRPV1-KO mice had reduced activation of ERK in sensory neurons, compared to WT mice. These changes in expression of p-ERK correlated with changes in pain behavior and joint histopathology: TRPV1-KO mice had reduced nociceptive behavior and severity of arthritis, compared to WT mice. CONCLUSION: Our results support the idea that activation of ERK in primary afferent neurons is mediated, at least in part, by TRPV1. In the absence of TRPV1, the signs of arthralgia and histopathology in the mouse model of AIA are reduced. We conclude that TRPV1, expressed by neurons in the articular afferent pathway, contributes to the pathogenesis of arthritis via an ERK-mediated pathway.


Subject(s)
Arthritis, Experimental/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , TRPV Cation Channels/physiology , Animals , Arthritis, Experimental/complications , Arthritis, Experimental/pathology , Arthritis, Experimental/physiopathology , Cartilage, Articular/pathology , Enzyme Activation/physiology , Ganglia, Spinal/enzymology , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/physiology , Sensory Receptor Cells/enzymology , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
5.
Neurosci Lett ; 297(2): 117-20, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11121884

ABSTRACT

In our laboratory, preliminary whole-cell, tight seal recordings of rat spinal substantia gelatinosa neurons including biocytin in the patch pipette yielded a significantly smaller proportion of neurons hyperpolarized by selective opioid agonists compared with recordings without biocytin. Therefore, we investigated the effects of biocytin inclusion on opioid responses and other membrane properties during whole-cell, tight seal recordings of these neurons. The percentage of neurons hyperpolarized by mu-, delta(1)-, and kappa-selective opioids was significantly reduced when 1% but not < or =0.2% biocytin was included in the recording pipette, compared with neurons recorded without biocytin. However, a significantly higher proportion of neurons fired spontaneous action potentials with either 0.05-0.2 or 1% biocytin compared to no biocytin. Resting membrane potential, input impedance and the proportion of neurons displaying transient outward rectification were each significantly altered for neurons recorded with 1% but not 0.05-0.2% biocytin. These effects may be due to a relatively specific blockade of diverse potassium channel types. Because efficient labeling can be achieved with 0.1% biocytin with whole-cell recording, higher concentrations are contraindicated.


Subject(s)
Analgesics, Opioid/pharmacology , Benzeneacetamides , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Lysine/pharmacology , Substantia Gelatinosa/cytology , Substantia Gelatinosa/drug effects , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Drug Interactions , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Lysine/analogs & derivatives , Membrane Potentials/drug effects , Organ Culture Techniques , Patch-Clamp Techniques , Pyrrolidines/pharmacology , Rats , Receptors, Opioid, delta/physiology , Receptors, Opioid, kappa/physiology , Receptors, Opioid, mu/physiology , Substantia Gelatinosa/physiology
6.
J Neurophysiol ; 82(6): 3316-26, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10601463

ABSTRACT

Using the in vivo whole cell recording procedure described previously, we recorded 73 neurons in laminae I and II in the lumbar spinal cord of the rat. Input impedances averaged 332 MOmega, which indicated that prior sharp electrode recordings contained a significant current shunt. Characterization of the adequate stimuli from the excitatory hindlimb receptive field indicated that 39 of 73 neurons were nociceptive, 6 were innocuous cooling cells, 20 responded maximally to brush, and 8 cells were not excited by stimulation of the skin of the hindlimb. The locations of 15 neurons were marked with biocytin. Nociceptive neurons were mostly found in lamina I and outer II, cooling cells in lamina I, and innocuous mechanoreceptive cells were mostly found in inner II or in the overlying white matter. The mu-opioid agonist [D-Ala(2), N-Me-Phe(4), Gly(5)-ol]-Enkephalin (DAMGO) hyperpolarized 7 of 19 tested neurons with a conductance increase. This hyperpolarization was reversed by naloxone in the neurons in which it was applied. DAMGO also decreased the frequency of spontaneous PSPs in 13 neurons, 7 of which were also hyperpolarized by DAMGO. Five of the seven hyperpolarized neurons were nociceptive, responding to both heat and mechanically noxious stimuli, whereas two responded to slow, innocuous brush. These results indicate that whole cell, tight seal recordings sample a similar population of lamina I and II neurons in the rat as those found with sharp electrode recordings in cat and monkey. They further indicate that DAMGO hyperpolarizes a subset of the nociceptive neurons that have input from both heat and mechanical nociceptors and that presynaptic DAMGO effects can be observed in nociceptive neurons that are not hyperpolarized by DAMGO.


Subject(s)
Narcotics/pharmacology , Neurons/physiology , Spinal Cord/physiology , Animals , Cold Temperature , Electric Stimulation , Electrophysiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Female , Histocytochemistry , Hot Temperature , Lysine/analogs & derivatives , Membrane Potentials/physiology , Microelectrodes , Neurons/drug effects , Pain/physiopathology , Patch-Clamp Techniques , Rats , Rats, Long-Evans , Receptors, Opioid, mu/agonists , Receptors, Presynaptic/drug effects , Spinal Cord/cytology , Spinal Cord/drug effects , Substantia Gelatinosa/cytology , Substantia Gelatinosa/drug effects , Substantia Gelatinosa/physiology , Synaptic Membranes/physiology
7.
Somatosens Mot Res ; 12(2): 151-61, 1995.
Article in English | MEDLINE | ID: mdl-7502605

ABSTRACT

Stimulation of peripheral nerves activates the proto-oncogene c-fos, which in turn generates its gene product, Fos. Fos and Fos-like proteins are produced in the central nervous system in response to chemical, mechanical, thermal, and electrical manipulation. The present study demonstrated a relationship between the number of Fos-like-immunoreactive nuclei in the spinal dorsal horn and graded intensities of electrical stimulation applied to the hindpaws of anesthetized and unanesthetized rats. Stimulation levels within the range of 0.1 to 1.0 mA were chosen on the basis of parmeters previously determined in behavioral investigations of escape reactions. Focal stimulation at these intensities activates peripheral axons directly, but does not injure or traumatize peripheral tissues. There was no evidence of inflammation or edema as a result of the focal electrical stimulation. As the stimulation intensity increased, the number and distribution of Fos-like-labeled nuclei increased with respect to rostral-caudal and laminar orientation. The threshold for expression of Fos-like immunoreactivity was different for anesthetized and unanesthetized animals. For anesthetized animals, the number of labeled nuclei increased significantly from the control level only when 1.0 mA was applied. However, in unanesthetized animals, the pattern of labeling was statistically significant at 0.2 mA. The present study demonstrates that electrical stimulation can evoke the expression of Fos-like immunoreactivity by activating nociceptors in the absence of tissue injury, and that the use of anesthetics can modulate this expression.


Subject(s)
Ganglia, Spinal/physiology , Hindlimb/innervation , Peripheral Nerves/physiology , Proto-Oncogene Proteins c-fos/genetics , Skin/innervation , Synaptic Transmission/genetics , Afferent Pathways/physiology , Animals , Electric Stimulation , Female , Gene Expression/physiology , Immunoenzyme Techniques , Neuronal Plasticity/genetics , Nociceptors/physiology , Pain Threshold/physiology , Rats
8.
Somatosens Mot Res ; 12(2): 163-74, 1995.
Article in English | MEDLINE | ID: mdl-7502606

ABSTRACT

In order to determine the effects of spinal cord lesions on nociceptive sensitivity of rodents, methods were developed to assess the speed of operant escape responses to electrocutaneous stimulation (ES). ES was delivered across the dorsal and ventral surfaces of either hindpaw, producing a current path through deep tissues. In order to guide establishment of a range of stimulus intensities for this manner of stimulation, a preliminary human psychophysical experiment was conducted with stimulation between the dorsal and ventral surfaces of a finger. For the human subjects, detection thresholds averaged 0.13 mA, and thresholds for a sharp (but nonpainful) sensation were 0.42 mA. Levels of stimulation between these thresholds for detection and a sharp quality elicited sensations of tingle or itch. Thresholds for reports of pain averaged 0.67 mA. On the basis of these results, intensities of ES ranging from 0.05 to 1.0 mA were presented to the feet of rats that were trained to perform an escape response with one forelimb. Thresholds for escape averaged slightly less than 0.1 mA; responding was consistent at 0.4 mA; and response probability and speed were maximal at approximately 0.8 mA. Thus, the rats responded aversively at intensities below those rated as sharp or painful by the human subjects, but the speed of escape reached a plateau at intensities that were above pain threshold for the human subjects. Unilateral thoracic lesions of the lateral spinal column of rats produced a contralateral hypalgesia. Escape thresholds were elevated, and the speed of escape responses to all intensities was reduced. This effect depended upon interruption of axons in the middle and anterior portions of one lateral column, corresponding to the location of long ascending pathways for nociception, including the spinothalamic tract. The speed of escape responding increased over 20 weeks of postoperative testing of animals with the largest lesions. This confirms results obtained previously from monkeys (by means of a similar paradigm), and corresponds to clinical reports of humans who have received spinal lesions for control of intractable pain. Thus, the location and organization of nociceptive pathways in the spinal cord of rodents appear to be similar to those of primates, and similar adaptations occur following interruption of these pathways.


Subject(s)
Escape Reaction/physiology , Hindlimb/innervation , Nociceptors/physiology , Pain Threshold/physiology , Spinal Cord/physiology , Adult , Animals , Electric Stimulation , Female , Functional Laterality/physiology , Humans , Male , Middle Aged , Rats , Reaction Time/physiology , Skin/innervation
9.
Brain Res ; 580(1-2): 172-9, 1992 May 15.
Article in English | MEDLINE | ID: mdl-1504797

ABSTRACT

C-fos is a proto-oncogene that is expressed within some neurons following depolarization. The protein product, fos, has been proposed as an anatomical marker for neuronal activity following noxious peripheral stimulation. However, the literature on noxious-stimulus induced fos expression contains several puzzling observations on the time course and laminar distribution of neuronal labeling within the spinal cord. This study has analyzed the effect of stimulus duration on the expression of fos-like immunoreactivity (FLI) within the spinal cord of anesthetized rats. In order to examine the time course of fos expression following brief periods of stimulation, we required a type of stimulus that was intense enough to activate nociceptors but that did not produce tissue damage. We have therefore employed pulsed, high intensity electrical stimulation, with stimulus durations ranging from 3 s to 24 h. The results indicate that stimulus duration has a profound effect upon the number of labeled cells, the intensity of neuronal labeling, the laminar pattern of FLI, and the time course of fos expression. Brief stimulation periods induce relatively few and relatively lightly labeled neurons, located predominantly within the most superficial laminae of the dorsal horn. Maximal immunoreactivity appears approximately 2 h after stimulation has ceased, and disappears within hours. Continuous stimulation produces many more labeled cells, darker labeling, and FLI within both dorsal and ventral laminar regions. Maximal FLI is seen after approximately 4.5 h of continuous stimulation, with reduction in the number of labeled cells thereafter. These data indicate that the results of any study employing c-fos as a marker for neuronal activity may be affected by the duration of the exciting stimulus.


Subject(s)
Gene Expression/physiology , Genes, fos/genetics , Spinal Cord/physiology , Animals , Electric Stimulation , Female , Male , Rats , Time Factors
10.
J Comp Neurol ; 295(2): 197-207, 1990 May 08.
Article in English | MEDLINE | ID: mdl-2358512

ABSTRACT

In the present study, serotoninergic and noradrenergic varicosities were identified in the ventral posterolateral nucleus of the macaque monkey. Monoaminergic neurons projecting to the ventral posterolateral nucleus of the thalamus were identified by using retrograde labeling with horseradish peroxidase combined with immunocytochemical staining for serotonin or dopamine-beta-hydroxylase. The midbrain nucleus raphe dorsalis was the major site of origin for neurons providing a serotoninergic projection to the ventral posterolateral nucleus. A few retrogradely labeled serotonin-containing neurons were also observed in the central superior and the raphe pontis nuclei. Noradrenergic cells with projections to the thalamus were primarily located in the nucleus locus coeruleus with some projection neurons in the nucleus subcoeruleus, and the A5 catecholamine cell group of the pons.


Subject(s)
Locus Coeruleus/metabolism , Macaca fascicularis/metabolism , Macaca/metabolism , Norepinephrine/metabolism , Raphe Nuclei/metabolism , Serotonin/metabolism , Thalamic Nuclei/metabolism , Animals , Dopamine beta-Hydroxylase , Horseradish Peroxidase , Immunohistochemistry , Locus Coeruleus/cytology , Macaca fascicularis/anatomy & histology , Neural Pathways/anatomy & histology , Neural Pathways/metabolism , Raphe Nuclei/cytology , Thalamic Nuclei/cytology
11.
J Comp Neurol ; 261(4): 467-80, 1987 Jul 22.
Article in English | MEDLINE | ID: mdl-3301921

ABSTRACT

Several lines of evidence indicate that the processing of somatosensory information in the dorsal column nuclei (DCN) is subject to descending controls. Anatomical experiments have demonstrated projections to the DCN from the sensorimotor cerebral cortex and the reticular formation. Physiological studies have shown that the activity of DCN neurons can be altered following stimulation of the cerebral cortex, reticular formation, periaqueductal gray, or raphe nuclei. Recent biochemical and electrophysiological evidence suggests a serotoninergic modulation of DCN neurons. The present study identifies serotonin-containing contacts on cells in the DCN that project to the thalamus in the rat. Retrograde labeling of brainstem neurons by horseradish peroxidase demonstrated projections to the DCN from the nucleus reticularis paragigantocellularis lateralis and from several raphe nuclei, including nuclei raphe obscurus (RO), pallidus (RP), and magnus (RM). Double labeling with horseradish peroxidase and antibody for serotonin indicated that the RO, RP and RM are likely to be the sources of the serotoninergic projections to the DCN. Thus, the role of the serotoninergic output from the raphe nuclei includes modulation of activity in the DCN.


Subject(s)
Medulla Oblongata/anatomy & histology , Rats, Inbred Strains/anatomy & histology , Serotonin/metabolism , Thalamus/anatomy & histology , Afferent Pathways/anatomy & histology , Animals , Immunoenzyme Techniques , Medulla Oblongata/cytology , Medulla Oblongata/metabolism , Raphe Nuclei/anatomy & histology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL