Calcitonin gene-related peptide promotes cellular changes in trigeminal neurons and glia implicated in peripheral and central sensitization

Ryan J Cady, Joseph R Glenn, Kael M Smith, Paul L Durham, Ryan J Cady, Joseph R Glenn, Kael M Smith, Paul L Durham

Abstract

Background: Calcitonin gene-related peptide (CGRP), a neuropeptide released from trigeminal nerves, is implicated in the underlying pathology of temporomandibular joint disorder (TMD). Elevated levels of CGRP in the joint capsule correlate with inflammation and pain. CGRP mediates neurogenic inflammation in peripheral tissues by increasing blood flow, recruiting immune cells, and activating sensory neurons. The goal of this study was to investigate the capability of CGRP to promote peripheral and central sensitization in a model of TMD.

Results: Temporal changes in protein expression in trigeminal ganglia and spinal trigeminal nucleus were determined by immunohistochemistry following injection of CGRP in the temporomandibular joint (TMJ) capsule of male Sprague-Dawley rats. CGRP stimulated expression of the active forms of the MAP kinases p38 and ERK, and PKA in trigeminal ganglia at 2 and 24 hours. CGRP also caused a sustained increase in the expression of c-Fos neurons in the spinal trigeminal nucleus. In contrast, levels of P2X3 in spinal neurons were only significantly elevated at 2 hours in response to CGRP. In addition, CGRP stimulated expression of GFAP in astrocytes and OX-42 in microglia at 2 and 24 hours post injection.

Conclusions: Our results demonstrate that an elevated level of CGRP in the joint, which is associated with TMD, stimulate neuronal and glial expression of proteins implicated in the development of peripheral and central sensitization. Based on our findings, we propose that inhibition of CGRP-mediated activation of trigeminal neurons and glial cells with selective non-peptide CGRP receptor antagonists would be beneficial in the treatment of TMD.

Figures

Figure 1
Figure 1
Increased number of trigeminal ganglia neurons expressing P-p38 in response to CGRP injection into the TMJ capsule. Sections of the posterolateral portion of the ganglion (V3) were obtained from untreated animals (CON), and animals receiving bilateral injections of CGRP after 2 and 24 hours. (A) Images of neuron-satellite glial regions stained for P-p38 are shown in the top panels. Magnification bar = 50 μm. The bottom panels are the same sections co-stained for P-p38 and DAPI. (B) The number of P-p38 positive cells ± SEM for each condition is reported (n = 3 independent experiments) #P < 0.01 when compared to control levels, while † P < 0.01 when compared to CGRP stimulated levels at 2 hours.
Figure 2
Figure 2
CGRP stimulates increased P-ERK expression in trigeminal ganglia neurons and satellite glia. Sections of the posterolateral portion of the ganglion (V3) were obtained from untreated animals (CON), or animals injected with CGRP in each TMJ capsule. (A) Images of neuron-satellite glial regions stained for P-ERK are shown in the top panels. Magnification bar = 50 μm. The bottom panels are the same sections co-stained for P-ERK and DAPI. (B) The average fold change ± SEM of P-ERK staining intensity from control values, whose mean was made equal to one, is reported (n = 3 independent experiments) # P < 0.01 when compared to control levels, while † P < 0.01 when compared to CGRP stimulated levels at 2 hours.
Figure 3
Figure 3
Peripheral injection of CGRP induces a sustained increase in expression of neuronal c-Fos in spinal trigeminal nucleus. (A) An image (40×) of a section of spinal cord 4 mm from the obex stained with the nuclear dye DAPI is shown. The white box represents the area encompassing the spinomedullary junction (Vc/C1-C2) transition zone. (B) Sections of spinal cord within the Vc/C1-2 region of the spinal trigeminal nucleus were obtained from control animals (CON), animals 2 hours post CGRP injections, or animals 24 hours post CGRP injection. Images of spinal cord tissues stained for c-Fos are shown in the top panels. Magnification bar = 50 μm. The same sections costained for c-Fos and DAPI are displayed in the bottom panels. (C) The average fold change ± SEM of c-Fos staining intensity from control values is reported (n = 3 independent experiments). * P < 0.05 when compared to control values.
Figure 4
Figure 4
PKA levels are elevated in the spinal trigeminal nucleus in response to CGRP. Spinal cord sections were obtained from control animals (CON), animals 2 hours post CGRP injections, or animals 24 hours post CGRP injection. (A) Images of spinal cord tissues stained for PKA are shown in the top panels. Magnification bar = 50 μm. The same sections costained for PKA and DAPI are displayed in the bottom panels. (B) The average fold change ± SEM of PKA staining intensity from control values is reported (n = 3). * P < 0.05 when compared to control values.
Figure 5
Figure 5
CGRP causes a transient increase in P2X3 expression in the spinal trigeminal nucleus at 2 and 24 hours. Sections of spinal cord were obtained from control animals (CON), animals 2 hours post CGRP injections or animals 24 hours post CGRP injection. (A) Images of spinal cord tissues stained for P2X3 are shown in the top panels. Magnification bar = 50 μm. The same sections costained for P2X3 and DAPI are displayed in the bottom panels. (B) The average fold change ± SEM of P2X3 staining intensity from control values is reported (n = 3). * P < 0.05 when compared to control values.
Figure 6
Figure 6
Elevated expression of OX-42 in microglia. Sections of spinal cord were obtained from control animals (CON), animals 2 hours post CGRP injections, or animals 24 hours post CGRP injection. (A) Images of spinal cord tissues stained for OX-42 are shown in the top panels. Magnification bar = 50 μm. The same sections costained for OX-42 and DAPI are shown in the bottom panels. (B) The average fold change ± SEM of OX-42 staining intensity from control values is reported (n = 3). * P < 0.05 when compared to control values.
Figure 7
Figure 7
CGRP induces a prolonged increase in expression of GFAP in astrocytes. Spinal cord sections were obtained from control animals (CON), animals 2 hours post CGRP injections, or animals 24 hours post CGRP injection. (A) Images of spinal cord tissues stained for GFAP are shown in the top panels. Magnification bar = 50 μm. The same sections costained for GFAP and DAPI are displayed in the bottom panels. (B) The average fold change ± SEM of GFAP staining intensity from control values is reported (n = 3). # P < 0.01 when compared to control values.

References

    1. Sessle BJ. Neural mechanisms and pathways in craniofacial pain. Can J Neurol Sci. 1999;26(Suppl 3):S7–11.
    1. Sessle BJ. Peripheral and central mechanisms of orofacial pain and their clinical correlates. Minerva Anestesiol. 2005;71:117–136.
    1. Okeson J. Management of Temporomandibular Disorders and Occlusion. 6. St. Louis, MO; 2008.
    1. Bonjardim LR, Lopes-Filho RJ, Amado G, Albuquerque RL, Goncalves SR. Association between symptoms of temporomandibular disorders and gender, morphological occlusion, and psychological factors in a group of university students. Indian J Dent Res. 2009;20:190–194.
    1. Shankland W. The trigeminal nerve. Part IV: the mandibular division. Journal of Craniomandibular Practice. 2001;19:153–161.
    1. Ji RR. Peripheral and central mechanisms of inflammatory pain, with emphasis on MAP kinases. Curr Drug Targets Inflamm Allergy. 2004;3:299–303.
    1. Guo W, Wang H, Watanabe M, Shimizu K, Zou S, LaGraize SC, Wei F, Dubner R, Ren K. Glial-cytokine-neuronal interactions underlying the mechanisms of persistent pain. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2007;27:6006–6018.
    1. Kopp S. Neuroendocrine, immune, and local responses related to temporomandibular disorders. J Orofac Pain. 2001;15:9–28.
    1. Sessle B. New insights into peripheral chemical mediators of temporomandibular pain and inflammation. J Orofac Pain. 2001;15:5.
    1. Kido M, Kiyoshima T, Kondo T, Ayasaka N, Moroi R, Terada Y, Tanaka T. Distribution of substance P and calcitonin gene-related peptide-like immunoreactive nerve fibers in the rat temporomandibular joint. J Dent Res. 1993;72:592–598.
    1. Uddman R, Grunditz T, Kato J, Sundler F. Distribution and origin of nerve fibers in the rat temporomandibular joint capsule. Anat Embryol. 1998;197:273–282.
    1. Appelgren A, Appelgren B, Kopp S, Lundeberg T, Theodorsson E. Neuropeptides in the arthritic TMJ and symptoms and signs from the stomatognathic system with special considerations to rheumatoid arthritis. J Orofac Pain. 1995;9:215–225.
    1. Appelgren A, Appelgren B, Kopp S, Lundeberg T, Theodorsson E. Relation between intra-articular temperature of the arthritic temporomandibular joint and presence of calcitonin gene-related peptide in the joint fluid. A clinical study. Acta Odontol Scand. 1993;51:285–291.
    1. Ottosson A, Edvinsson L. Release of histamine from dural mast cells by substance P and calcitonin gene-related peptide. Cephalalgia. 1997;17:166–174.
    1. Seybold V. The Role of Peptides in Central Sensitization. Handbook of Exp Pharma. 2009;194:451–491.
    1. Seybold V, McCarson K, Mermelstein P, Groth R, Abrahams J. Calcitonin gene-related peptide regulates expression of neurokinin1 receptors by rat spinal neurons. The Journal of Neuroscience. 2003;23:1816–1824.
    1. Hucho T, Levine JD. Signaling pathways in sensitization: toward a nociceptor cell biology. Neuron. 2007;55:365–376.
    1. Li D, Ren Y, Xu X, Zou X, Fang L, Lin Q. Sensitization of primary afferent nociceptors induced by intradermal capsaicin involves the peripheral release of calcitonin gene-related Peptide driven by dorsal root reflexes. J Pain. 2008;9:1155–1168.
    1. Lennerz JK, Ruhle V, Ceppa EP, Neuhuber WL, Bunnett NW, Grady EF, Messlinger K. Calcitonin receptor-like receptor (CLR), receptor activity-modifying protein 1 (RAMP1), and calcitonin gene-related peptide (CGRP) immunoreactivity in the rat trigeminovascular system: differences between peripheral and central CGRP receptor distribution. J Comp Neurol. 2008;507:1277–1299.
    1. Thalakoti S, Patil VV, Damodaram S, Vause CV, Langford LE, Freeman SE, Durham PL. Neuron-glia signaling in trigeminal ganglion: implications for migraine pathology. Headache. 2007;47:1008–1023.
    1. Van Rossum D, Hanisch U, Quirion R. Neuroanatomical localization, pharmacological characterization and functions of CGRP, related peptides and their receptors. Neurosci Biobehav Rev. 1997;21:649–678.
    1. Hughes SR, Brain SD. A calcitonin gene-related peptide (CGRP) antagonist (CGRP8-37) inhibits microvascular responses induced by CGRP and capsaicin in skin. Br J Pharmacol. 1991;104:738–742.
    1. Edvinsson L, Nilsson E, Jansen-Olesen I. Inhibitory effect of BIBN4096BS, CGRP8-37, a CGRP antibody and an RNA-Spiegelmer on CGRP induced vasodilatation in the perfused and non-perfused rat middle cerebral artery. Br J Pharmacol. 2007;150:633–640.
    1. Jang JH, Nam TS, Paik KS, Leem JW. Involvement of peripherally released substance P and calcitonin gene-related peptide in mediating mechanical hyperalgesia in a traumatic neuropathy model of the rat. Neurosci Lett. 2004;360:129–132.
    1. Zhang L, Hoff A, Wimalawansa S, Cote G, Gagel R, Westlund K. Arthritic calcitonin/α calcitonin gene-related peptide knockout mice have reduced nociceptive hypersensitivity. Pain. 2001;89:265–273.
    1. Cady RJ, Hirst JJ, Durham PL. Dietary grape seed polyphenols repress neuron and glia activation in trigeminal ganglion and trigeminal nucleus caudalis. Mol Pain. 2010;6:91.
    1. Shinoda M, Ozaki N, Asai H, Nagamine K, Sugiura Y. Changes in P2X3 receptor expression in the trigeminal ganglion following monoarthritis of the temporomandibular joint in rats. Pain. 2005;116:42–51.
    1. Ji RR, Gereau RWt, Malcangio M, Strichartz GR. MAP kinase and pain. Brain Res Rev. 2009;60:135–148.
    1. Sluka KA, Price MP, Breese NM, Stucky CL, Wemmie JA, Welsh MJ. Chronic hyperalgesia induced by repeated acid injections in muscle is abolished by the loss of ASIC3, but not ASIC1. Pain. 2003;106:229–239.
    1. Ji R, Gereau RW IV, Malcangio M, Strichartz GR. MAP kinase and pain. Brain Res Rev. 2009;60:135–148.
    1. Alstergren P, Appelgren A, Appelgren B, Kopp S, Lundeberg T, Theodorsson E. Co-variation of neuropeptide Y, calcitonin gene-related peptide, substance P and neurokinin A in joint fluid from patients with temporomandibular joint arthritis. Arch Oral Biol. 1995;40:127–135.
    1. Holmlund A, Ekblom A, Hansson P, Lind J, Lundenberg T, Theodorsson E. Concentration of neuropeptide substance - P, Neurokinin A, calcitonin gene related peptide, neuropeptide Y and vasoactive intestinal polypeptide in the synovial fluid of the -human temporomandibular joint. Int J Oral Maxillofac Surg. 1991;20:228–231.
    1. Geppetti P, Del Bianco E, Patacchini R, Santicoli P, Maggi C, Tramontana M. Low pH-induced release of calcitonin gene-related peptide from capsaicin-sensitive sensory nerves; mechanism of action and biological response. Neurosci. 1991;41:295–301.
    1. Cheng J, Ji R. Intracellular signaling in primary sensory neurons and persistent pain. Neurochem Res. 2008;33:1970–1978.
    1. Takeda M, Takahashi M, Matsumoto S. Contribution of the activation of satellite glia in sensory ganglia to pathological pain. Neurosci Biobehav Rev. 2009;33:784–792.
    1. Cheng JK, Ji RR. Intracellular signaling in primary sensory neurons and persistent pain. Neurochem Res. 2008;33:1970–1978.
    1. Crown ED, Ye Z, Johnson KM, Xu GY, McAdoo DJ, Hulsebosch CE. Increases in the activated forms of ERK 1/2, p38 MAPK, and CREB are correlated with the expression of at-level mechanical allodynia following spinal cord injury. Exp Neurol. 2006;199:397–407.
    1. Milligan ED, Twining C, Chacur M, Biedenkapp J, O'Connor K, Poole S, Tracey K, Martin D, Maier SF, Watkins LR. Spinal glia and proinflammatory cytokines mediate mirror-image neuropathic pain in rats. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2003;23:1026–1040.
    1. Tsuda M, Mizokoshi A, Shigemoto-Mogami Y, Koizumi S, Inoue K. Activation of p38 mitogen-activated protein kinase in spinal hyperactive microglia contributes to pain hypersensitivity following peripheral nerve injury. Glia. 2004;45:89–95.
    1. Suter MR, Wen YR, Decosterd I, Ji RR. Do glial cells control pain? Neuron Glia Biol. 2007;3:255–268.
    1. Ren K. Emerging role of astroglia in pain hypersensitivity. Jpn Dent Sci Rev. 2010;46:86.
    1. Watkins LR, Maier SF. Beyond neurons: evidence that immune and glial cells contribute to pathological pain states. Physiol Rev. 2002;82:981–1011.
    1. Han JS, Li W, Neugebauer V. Critical role of calcitonin gene-related peptide 1 receptors in the amygdala in synaptic plasticity and pain behavior. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2005;25:10717–10728.
    1. Fitzgerald EM, Okuse K, Wood JN, Dolphin AC, Moss SJ. cAMP-dependent phosphorylation of the tetrodotoxin-resistant voltage-dependent sodium channel SNS. J Physiol. 1999;516(Pt 2):433–446.
    1. Bhave G, Zhu W, Wang H, Brasier DJ, Oxford GS, Gereau RWt. cAMP-dependent protein kinase regulates desensitization of the capsaicin receptor (VR1) by direct phosphorylation. Neuron. 2002;35:721–731.
    1. Aley KO, Levine JD. Role of protein kinase A in the maintenance of inflammatory pain. The Journal of neuroscience: the official journal of the Society for Neuroscience. 1999;19:2181–2186.
    1. Malmberg AB, Brandon EP, Idzerda RL, Liu H, McKnight GS, Basbaum AI. Diminished inflammation and nociceptive pain with preservation of neuropathic pain in mice with a targeted mutation of the type I regulatory subunit of cAMP-dependent protein kinase. The Journal of neuroscience: the official journal of the Society for Neuroscience. 1997;17:7462–7470.
    1. Burnstock G. Purinergic signalling--an overview. Novartis Found Symp. 2006;276:26–48. discussion 48-57, 275-281.
    1. Burnstock G, Wood JN. Purinergic receptors: their role in nociception and primary afferent neurotransmission. Curr Opin Neurobiol. 1996;6:526–532.
    1. Hamilton SG, McMahon SB. ATP as a peripheral mediator of pain. J Auton Nerv Syst. 2000;81:187–194.
    1. Staikopoulos V, Sessle BJ, Furness JB, Jennings EA. Localization of P2X2 and P2X3 receptors in rat trigeminal ganglion neurons. Neuroscience. 2007;144:208–216.
    1. Jarvis MF. Contributions of P2X3 homomeric and heteromeric channels to acute and chronic pain. Expert Opin Ther Targets. 2003;7:513–522.
    1. Burnstock G. P2X receptors in sensory neurones. Br J Anaesth. 2000;84:476–488.
    1. Jarvis MF, Burgard EC, McGaraughty S, Honore P, Lynch K, Brennan TJ, Subieta A, Van Biesen T, Cartmell J, Bianchi B. et al.A-317491, a novel potent and selective non-nucleotide antagonist of P2X3 and P2X2/3 receptors, reduces chronic inflammatory and neuropathic pain in the rat. Proc Natl Acad Sci USA. 2002;99:17179–17184.
    1. Oliveira MC, Pelegrini-da-Silva A, Tambeli CH, Parada CA. Peripheral mechanisms underlying the essential role of P2X3,2/3 receptors in the development of inflammatory hyperalgesia. Pain. 2009;141:127–134.
    1. Ho TW, Ferrari MD, Dodick DW, Galet V, Kost J, Fan X, Leibensperger H, Froman S, Assaid C, Lines C. et al.Efficacy and tolerability of MK-0974 (telcagepant), a new oral antagonist of calcitonin gene-related peptide receptor, compared with zolmitriptan for acute migraine: a randomised, placebo-controlled, parallel-treatment trial. Lancet. 2008;372:2115–2123.
    1. Cady RJ, Durham PL. Cocoa-enriched diets enhance expression of phosphatases and decrease expression of inflammatory molecules in trigeminal ganglion neurons. Brain Res. 2010.
    1. Garrett FG, Durham PL. Differential expression of connexins in trigeminal ganglion neurons and satellite glial cells in response to chronic or acute joint inflammation. Neuron Glia Biol. 2008;4:295–306.

Source: PubMed

3
Prenumerera