Publication Cover
Canadian Journal of Pain
Revue canadienne de la douleur
Volume 4, 2020 - Issue 1
3,391
Views
9
CrossRef citations to date
0
Altmetric
Review

Biomarkers in temporomandibular disorder and trigeminal neuralgia: A conceptual framework for understanding chronic pain

ORCID Icon, , &
Pages 1-18 | Received 07 Aug 2019, Accepted 21 Dec 2019, Published online: 23 Jan 2020

References

  • Macfarlane TV, Blinkhorn AS, Davies RM, Kincey J, Worthington HV. Oro-facial pain in the community: prevalence and associated impact. Community Dent Oral Epidemiol. 2002;30(1):52–60. doi:10.1034/j.1600-0528.2002.300108.x.
  • Watt-Watson J, McGillion M, Hunter J, Choiniere M, Clark AJ, Dewar A, Johnston C, Lynch M, Morley-Forster P, Moulin D, et al. A survey of prelicensure pain curricula in health science faculties in Canadian universities. Pain Res Manag. 2009;14(6):439–44. doi:10.1155/2009/307932.
  • Briggs EV, Carr ECJ, Whittaker MS. Survey of undergraduate pain curricula for healthcare professionals in the United Kingdom. Eur J Pain. 2011;15(8):789–95. doi:10.1016/j.ejpain.2011.01.006.
  • Institute of Medicine. Relieving pain in America. Washington (D.C.): National Academies Press; 2011. doi:10.17226/13172.
  • Mezei L, Murinson BB. Pain education in North American Medical Schools. J Pain. 2011;12(12):1199–208. doi:10.1016/j.jpain.2011.06.006.
  • Shipton EE, Bate F, Garrick R, Steketee C, Shipton EA, Visser EJ. Systematic review of pain medicine content, teaching, and zssessment in medical school curricula internationally. Pain Ther. 2018;7(2):139–61. doi:10.1007/s40122-018-0103-z.
  • List T, Jensen RH. Temporomandibular disorders: old ideas and new concepts. Cephalalgia. 2017;37(7):692–704. doi:10.1177/0333102416686302.
  • Schiffman E, Ohrbach R, Truelove E, Look J, Anderson G, Goulet J-P, List T, Svensson P, Gonzalez Y, Lobbezoo F, et al. Diagnostic criteria for temporomandibular disorders (DC/TMD) for clinical and research applications: recommendations of the International RDC/TMD Consortium Network and Orofacial Pain Special Interest Group. J Oral Facial Pain Headache. 2014;28(1):6–27. doi:10.11607/jop.1151.
  • White JC, Sweet WH Pain and the neurosurgeon: a forty-year experience. Springfield, IL: C.C. Thomas; 1969. https://www.worldcat.org/title/pain-and-the-neurosurgeon-a-forty-year-experience/oclc/21431. Accessed July 23, 2019.
  • Cruccu G, Finnerup NB, Jensen TS, Scholz J, Sindou M, Svensson P, Treede R-D, Zakrzewska JM, Nurmikko T. Trigeminal neuralgia: new classification and diagnostic grading for practice and research. Neurology. 2016;87(2):220–28. doi:10.1212/WNL.0000000000002840.
  • Vincent M, Wang S. Headache classification committee of the International Headache Society (IHS) The international classification of headache disorders, 3rd edition. Cephalalgia. 2018;38(1):1–211. doi:10.1177/0333102417738202.
  • Scrivani SJ, Keith DA, Kaban LB. Temporomandibular disorders. N Engl J Med. 2008;359(25):2693–705. doi:10.1056/NEJMra0802472.
  • Zakrzewska JM, Linskey ME. Trigeminal neuralgia. BMJ. 2014;348(feb17 9):g474–g474. doi:10.1136/bmj.g474.
  • Zakrzewska JM. Facial pain: neurological and non-neurological. J Neurol Neurosurg Psychiatry. 2002;72(Suppl 2):ii27–ii32. doi:10.1136/jnnp.72.suppl_2.ii27.
  • Maixner W, Fillingim RB, Williams DA, Smith SB, Slade GD. Overlapping chronic pain conditions: implications for diagnosis and classification. J Pain. 2016;17(9):T93–T107. doi:10.1016/j.jpain.2016.06.002.
  • Goulet JP, Velly AM. Orofacial Pain Biomarkers. In: Goulet J-P, Velly AM editors. Part III: biomarkers in orofacial pain. Heidelberg, Germany: Springer Berlin Heidelberg; 2017. p. 65–131. doi: 10.1007/978-3-662-53994-1.
  • National Institute of Neurological Disorders and Stroke. Discovery and validation of biomarkers to develop non-addictive therapeutics for pain—home. https://meetings.ninds.nih.gov/Home/General/20748. Published 2018. Accessed July 19, 2019.
  • United Nations Environment Programme, International Labour Organisation, World Health Organization, International Program on Chemical Safety. Biomarkers in risk assessment: validity and validation. World Health Organization; 2001.
  • FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and Other Tools) resource. Bethesda (MD): Food and Drug Administration (US). 2016 [Accessed July 18, 2019.]. http://www.ncbi.nlm.nih.gov/pubmed/27010052.
  • Sedgwick P. Prospective cohort studies: advantages and disadvantages. BMJ. 2013;347(nov08 1):f6726–f6726. doi:10.1136/bmj.f6726.
  • Maixner W, Diatchenko L, Dubner R, Fillingim RB, Greenspan JD, Knott C, Ohrbach R, Weir B, Slade GD. Orofacial pain prospective evaluation and risk assessment study - the OPPERA study. J Pain. 2011;12(11SUPPL):T4-11.e1-2. doi:10.1016/j.jpain.2011.08.002.
  • Völzke H, Alte D, Schmidt CO, Radke D, Lorbeer R, Friedrich N, Aumann N, Lau K, Piontek M, Born G, et al. Cohort profile: the study of health in Pomerania. Int J Epidemiol. 2011;40(2):294–307. doi:10.1093/ije/dyp394.
  • Sanders AE, Jain D, Sofer T, Kerr KF, Laurie CC, Shaffer JR, Marazita ML, Kaste LM, Slade GD, Fillingim RB, et al. GWAS identifies new loci for painful temporomandibular disorder: hispanic Community Health Study/Study of Latinos. J Dent Res. 2017;96(3):277–84. doi:10.1177/0022034516686562.
  • Sudlow C, Gallacher J, Allen N, Beral V, Burton P, Danesh J, Downey P, Elliott P, Green J, Landray M, et al. UK Biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 2015;12(3). doi:10.1371/journal.pmed.1001779
  • Smith SB, Parisien M, Bair E, Belfer I, Chabot-Doré A-J, Gris P, Khoury S, Tansley S, Torosyan Y, Zaykin DV, et al. Genome-wide association reveals contribution of MRAS to painful temporomandibular disorder in males. Pain. 2019;160(3):579–91. doi:10.1097/j.pain.0000000000001438.
  • Kim HJ, Greenspan JD, Ohrbach R, Fillingim RB, Maixner W, Renn CL, Johantgen M, Zhu S, Dorsey SG. Racial/ethnic differences in experimental pain sensitivity and associated factors – cardiovascular responsiveness and psychological status. Koenig J, ed. PLoS One. 2019;14(4):e0215534. doi:10.1371/journal.pone.0215534.
  • Katusic S, Beard CM, Bergstralth E, Kurland LT. Incidence and clinical features of trigeminal neuralgia, Rochester, Minnesota, 1945–1984. Ann Neurol. 1990;27(1):89–95. doi:10.1002/ana.410270114.
  • Diatchenko L, Slade GD, Nackley AG, Bhalang K, Sigurdsson A, Belfer I, Goldman D, Xu K, Shabalina SA, Shagin D, et al. Genetic basis for individual variations in pain perception and the development of a chronic pain condition. Hum Mol Genet. 2005;14(1):135–43. doi:10.1093/hmg/ddi013.
  • Slade GD, Diatchenko L, Ohrbach R, Maixner W. Orthodontic treatment, genetic factors, and risk of temporomandibular disorder. Semin Orthod. 2008;14(2):146–56. doi:10.1053/j.sodo.2008.02.005.
  • Kim MR, Graber TM, Viana MA. Orthodontics and temporomandibular disorder: A meta-analysis. Am J Orthod Dentofac Orthop. 2002;121(5):438–46. doi:10.1067/mod.2002.121665.
  • Macfarlane TV, Kenealy P, Kingdon HA, Mohlin BO, Pilley JR, Richmond S, Shaw WC. Twenty-year cohort study of health gain from orthodontic treatment: temporomandibular disorders. Am J Orthod Dentofac Orthop. 2009;135(6):692.e1–692.e8. doi:10.1016/j.ajodo.2008.10.017.
  • Mladenovic I, Krunic J, Supic G, Kozomara R, Bokonjic D, Stojanovic N, Magic Z. Pulp sensitivity: influence of sex, psychosocial variables, COMT gene, and chronic facial pain. J Endod. 2018;44(5):717–721.e1. doi:10.1016/j.joen.2018.02.002.
  • Mladenovic I, Supic G, Kozomara R, Dodic S, Ivkovic N, Milicevic B, Simic I, Magic Z. Genetic polymorphisms of catechol-o-methyltransferase: association with temporomandibular disorders and postoperative pain. J Oral Facial Pain Headache. 2016;30(4):302–10. doi:10.11607/ofph.1688.
  • Michelotti A, Liguori R, Toriello M, D’Antò V, Vitale D, Castaldo G, Sacchetti L. Catechol-o-methyltransferase (COMT) gene polymorphisms as risk factor in temporomandibular disorders patients from southern Italy. Clin J Pain. 2014;30(2):129–33. doi:10.1097/AJP.0b013e318287a358.
  • Smith SB, Reenilä I, Männistö PT, Slade GD, Maixner W, Diatchenko L, Nackley AG. Epistasis between polymorphisms in COMT, ESR1, and GCH1 influences COMT enzyme activity and pain. Pain. 2014;155(11):2390–99. doi:10.1016/j.pain.2014.09.009.
  • Meloto CB, Segall SK, Smith S, Parisien M, Shabalina SA, Rizzatti-Barbosa CM, Gauthier J, Tsao D, Convertino M, Piltonen MH, et al. COMT gene locus: new functional variants. Pain. 2015;156(10):2072–83. doi:10.1097/j.pain.0000000000000273.
  • Visscher CM, Lobbezoo F. TMD pain is partly heritable. A systematic review of family studies and genetic association studies. J Oral Rehabil. 2015;42(5):386–99. doi:10.1111/joor.12263.
  • Smith SB, Maixner DW, Greenspan JD, Dubner R, Fillingim RB, Ohrbach R, Knott C, Slade GD, Bair E, Gibson DG, et al. Potential genetic risk factors for chronic TMD: genetic associations from the OPPERA case control study. J Pain. 2011;12(11):T92–T101. doi:10.1016/j.jpain.2011.08.005.
  • D’Agnelli S, Arendt-Nielsen L, Gerra MC, Zatorri K, Boggiani L, Baciarello M, Bignami E. Fibromyalgia: genetics and epigenetics insights may provide the basis for the development of diagnostic biomarkers. Mol Pain. 2019;15:174480691881994. doi:10.1177/1744806918819944.
  • Ciampi De Andrade D, Maschietto M, Galhardoni R, Gouveia G, Chile T, Victorino Krepischi AC, Dale CS, Brunoni AR, Parravano DC, Cueva Moscoso AS, et al. Epigenetics insights into chronic pain: DNA hypomethylation in fibromyalgia - A controlled pilot-study. Pain. 2017;158(8):1473–80. doi:10.1097/j.pain.0000000000000932.
  • Velly AM, Fricton J. Comorbidities in individuals with orofacial pain and their impact on biomarkers: orofacial pain comorbidities and their impact on biomarkers. In: J-P Goulet, AM Velly, editors. Orofacial pain biomarkers. Berlin (Heidelberg): Springer Berlin Heidelberg; 2017. p. 21–32. doi:10.1007/978-3-662-53994-1_2.
  • Fleetwood IG, Innes AM, Hansen SR, Steinberg GK. Familial trigeminal neuralgia. J Neurosurg. 2001;95(3):513–17. doi:10.3171/jns.2001.95.3.0513.
  • Cervera-Martinez C, Martinez-Manrique JJ, Revuelta-Gutierrez R. Surgical management of familial trigeminal neuralgia with different inheritance patterns: a case report. Front Neurol. 2018:9. doi:10.3389/fneur.2018.00316.
  • Panchagnula S, Sularz AK, Kahle KT. Familial trigeminal neuralgia cases implicate genetic factors in disease pathogenesis. JAMA Neurol. 2019;76(1):9–10. doi:10.1001/jamaneurol.2018.3322.
  • Cui W, Yu X, Zhang H. The serotonin transporter gene polymorphism is associated with the susceptibility and the pain severity in Idiopathic Trigeminal Neuralgia patients. J Headache Pain. 2014;15(1):1–6. doi:10.1186/1129-2377-15-42.
  • Tanaka BS, Zhao P, Dib-Hajj FB, Morisset V, Tate S, Waxman SG, Dib-Hajj SD. A gain-of-function mutation in Nav1.6 in a case of trigeminal neuralgia. Mol Med. 2016;22(1):338–48. doi:10.2119/molmed.2016.00131.
  • Siqueira SRDT, Alves B, Malpartida HMG, Teixeira MJ, Siqueira JTT. Abnormal expression of voltage-gated sodium channels Nav1.7, Nav1.3 and Nav1.8 in trigeminal neuralgia. Neuroscience. 2009;164(2):573–77. doi:10.1016/j.neuroscience.2009.08.037.
  • Costa GMF, Rocha LPC, de Siqueira SRDT, Moreira PR, Almeida-Leite CM. No association of polymorphisms in Nav1.7 or nerve growth factor receptor genes with trigeminal neuralgia. Pain Med. 2019;20(7):1362–69. doi:10.1093/pm/pny191.
  • Zakrzewska JM, Palmer J, Morisset V, Giblin GM, Obermann M, Ettlin DA, Cruccu G, Bendtsen L, Estacion M, Derjean D, et al. Safety and efficacy of a Nav1.7 selective sodium channel blocker in patients with trigeminal neuralgia: a double-blind, placebo-controlled, randomised withdrawal phase 2a trial. Lancet Neurol. 2017;16(4):291–300. doi:10.1016/S1474-4422(17)30005-4.
  • Chung WH, Hung SI, Hong HS, Hsih M-S, Yang L-C, Ho H-C, Wu J-Y, Chen Y-T. A marker for Stevens-Johnson syndrome. Nature. 2004;428(6982):486. doi:10.1038/428486a.
  • McCormack M, Alfirevic A, Bourgeois S, Farrell JJ, Kasperavičiūtė D, Carrington M, Sills GJ, Marson T, Jia X, de Bakker PIW, et al. HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans. N Engl J Med. 2011;364(12):1134–43. doi:10.1056/NEJMoa1013297.
  • Dean L. Carbamazepine therapy and HLA genotype. Bethesda (MD): National Center for Biotechnology Information (US); 2012. [Accessed July 25, 2019.]. http://www.ncbi.nlm.nih.gov/pubmed/28520367.
  • Obermann M, Yoon M-S, Ese D, Maschke M, Kaube H, Diener H-C, Katsarava Z. Impaired trigeminal nociceptive processing in patients with trigeminal neuralgia. Neurology. 2007;69(9):835–41. doi:10.1212/01.wnl.0000269670.30045.6b.
  • Cairns BE. Pathophysiology of TMD pain - basic mechanisms and their implications for pharmacotherapy. J Oral Rehabil. 2010;37(6):391–410. doi:10.1111/j.1365-2842.2010.02074.x.
  • Castrillon EE, Ernberg M, Cairns BE, Wang K, Sessle BJ, Arendt-Nielsen L, Svensson P. Interstitial glutamate concentration is elevated in the masseter muscle of myofascial temporomandibular disorder patients. J Orofac Pain. 2010; 24(4): 350–60. http://www.ncbi.nlm.nih.gov/pubmed/21197506
  • Louca Jounger S, Christidis N, Svensson P, List T, Ernberg M. Increased levels of intramuscular cytokines in patients with jaw muscle pain. J Headache Pain. 2017;18(1). doi:10.1186/s10194-017-0737-y.
  • Dubner R, Harper DE, Schrepf A, Clauw DJ. Pain mechanisms and centralized pain in temporomandibular disorders. J Dent Res. 2016;95(10):1102–08. doi:10.1177/0022034516657070.
  • Ericson H, Abu Hamdeh S, Freyhult E, Stiger F, Bäckryd E, Svenningsson A, Gordh T, Kultima K. Cerebrospinal fluid biomarkers of inflammation in trigeminal neuralgia patients operated with microvascular decompression. Pain. 2019;160(11):2603–11. doi:10.1097/j.pain.0000000000001649.
  • Liu MX, Zhong J, Xia L, Dou NN, Li ST. A correlative analysis between inflammatory cytokines and trigeminal neuralgia or hemifacial spasm. Neurol Res. 2019;41(4):335–40. doi:10.1080/01616412.2018.1564188.
  • Moayedi M, Weissman-Fogel I, Salomons TV, Crawley AP, Goldberg MB, Freeman BV, Tenenbaum HC, Davis KD. White matter brain and trigeminal nerve abnormalities in temporomandibular disorder. Pain. 2012;153(7):1467–77. doi:10.1016/j.pain.2012.04.003.
  • Monaco A, Cattaneo R, Mesin L, Ortu E, Giannoni M, Pietropaoli D. Dysregulation of the descending pain system in temporomandibular disorders revealed by low-frequency sensory transcutaneous electrical nerve stimulation: a pupillometric study. PLoS One. 2015;10(4):e0122826. doi:10.1371/journal.pone.0122826.
  • Fu K, Xuchen MA, Zhang Z, Chen W. Tumor necrosis factor in synovial fluid of patients with temporomandibular disorders. J Oral Maxillofac Surg. 1995;53(4):424–26. doi:10.1016/0278-2391(95)90717-3.
  • Takahashi T, Kondoh T, Fukuda M, Yamazaki Y, Toyosaki T, Suzuki R. Proinflammatory cytokines detectable in synovial fluids from patients with temporomandibular disorders. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 1998;85(2):135–41. doi:10.1016/S1079-2104(98)90415-2.
  • Emshoff R, Puffer P, Rudisch A, Gaßner R. Temporomandibular joint pain: relationship to internal derangement type, osteoarthrosis, and synovial fluid mediator level of tumor necrosis factor-α. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2000;90(4):442–49. doi:10.1067/moe.2000.108801.
  • Kaneyama K, Segami N, Sun W, Sato J, Fujimura K. Analysis of tumor necrosis factor-α, interleukin-6, interleukin-1β, soluble tumor necrosis factor receptors I and II, interleukin-6 soluble receptor, interleukin-1 soluble receptor type II, interleukin-1 receptor antagonist, and protein in the synovial fluid of patients with temporomandibular joint disorders. Oral Surgery, Oral Med Oral Pathol Oral Radiol Endodontology. 2005;99(3):276–84. doi:10.1016/J.TRIPLEO.2004.06.074.
  • Nordahl S, Alstergren P, Kopp S. Tumor necrosis factor-alpha in synovial fluid and plasma from patients with chronic connective tissue disease and its relation to temporomandibular joint pain. J Oral Maxillofac Surg. 2000;58(5):525–30. doi:10.1016/S0278-2391(00)90015-6.
  • Fredriksson L, Alstergren P, Kopp S. Tumor necrosis factor-α in temporomandibular joint synovial fluid predicts treatment effects on pain by intra-articular glucocorticoid treatment. Mediators Inflamm. 2006;2006(6):1–7. doi:10.1155/MI/2006/59425.
  • Shafer DM, Assael L, White LB, Rossomando EF. Tumor necrosis factor-α as a biochemical marker of pain and outcome in temporomandibular joints with internal derangements. J Oral Maxillofac Surg. 1994;52(8):786–91. doi:10.1016/0278-2391(94)90217-8.
  • Kubota E, Kubota T, Matsumoto J, Shibata T, Murakami KI. Synovial fluid cytokines and proteinases as markers of temporomandibular joint disease. J Oral Maxillofac Surg. 1998;56(2):192–98. doi:10.1016/S0278-2391(98)90868-0.
  • Suzuki T, Segami N, Kaneyama K, Nishimura M, Nojima T. Specific expression of interleukin-1β in temporomandibular joints with internal derangement: correlation with clinical findings. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 1999;88(4):413–17. doi:10.1016/S1079-2104(99)70054-5.
  • Shinoda C, Takaku S. Oral and maxillofacial pathology: interleukin-1 β, interleukin-6, and tissue inhibitor of metalloproteinase-1 in the synovial fluid of the temporomandibular joint with respect to cartilage destruction. Oral Dis. 2008;6(6):383–90. doi:10.1111/j.1601-0825.2000.tb00131.x.
  • Nishimura M, Segami N, Kaneyama K, Suzuki T, Miyamaru M. Proinflammatory cytokines and arthroscopic findings of patients with internal derangement and osteoarthritis of the temporomandibular joint. Br J Oral Maxillofac Surg. 2002;40(1):68–71. doi:10.1054/bjom.2001.0742.
  • Sato J, Segami N, Nishimura M, Demura N, Yoshimura H, Yoshitake Y, Nishikawa K. Expression of interleukin 6 in synovial tissues in patients with internal derangement of the temporomandibular joint. Br J Oral Maxillofac Surg. 2003;41(2):95–101. doi:10.1016/S0266-4356(02)00294-2.
  • Rodríguez de Sotillo D, Velly AM, Hadley M, Fricton JR. Evidence of oxidative stress in temporomandibular disorders: a pilot study. J Oral Rehabil. 2011;38(10):722–28. doi:10.1111/j.1365-2842.2011.02216.x.
  • Clark AK, Old EA, Malcangio M. Neuropathic pain and cytokines: current perspectives. J Pain Res. 2013;2013:803–14. https://doaj.org/article/dbec995b4eb246549a1e0f1e3c0ff649.
  • Goebel A, Lee MK, Cacciola F, Cross A, Eldridge P. A technique to assess perineuronal mediators. Br J Neurosurg. December 2017;1–3. doi:10.1080/02688697.2017.1416058.
  • Alstergren P, Pigg M, Kopp S. Clinical diagnosis of temporomandibular joint arthritis. J Oral Rehabil. 2018;45(4):269–81. doi:10.1111/joor.12611.
  • Basi DL, Velly AM, Schiffman EL, Lenton PA, Besspiata DA, Rankin AM, Hughes PJ, Swift JQ, Kehl LJ. Human temporomandibular joint and myofascial pain biochemical profiles: a case-control study. J Oral Rehabil. 2012;39(5):326–37. doi:10.1111/j.1365-2842.2011.02271.x.
  • Zhou X, Fragala MS, McElhaney JE, Kuchel GA. Conceptual and methodological issues relevant to cytokine and inflammatory marker measurements in clinical research. Curr Opin Clin Nutr Metab Care. 2010;13(5):541–47. doi:10.1097/MCO.0b013e32833cf3bc.
  • Schuster NM, Rapoport AM. New strategies for the treatment and prevention of primary headache disorders. Nat Rev Neurol. 2016;12(11):635–50. doi:10.1038/nrneurol.2016.143.
  • Alstergren P, Kopp S. Pain and synovial fluid concentration of serotonin in arthritic temporomandibular joints. Pain. 1997;72(1–2):137–43. doi:10.1016/S0304-3959(97)00022-5.
  • Fredriksson L, Alstergren P, Kopp S. Serotonergic mechanisms influence the response to glucocorticoid treatment in TMJ arthritis. Mediators Inflamm. 2005;2005(4):194–201. doi:10.1155/MI.2005.194.
  • Christidis N, Kang I, Cairns BE, Kumar U, Dong X, Rosén A, Kopp S, Ernberg M. Expression of 5-HT3 receptors and TTX resistant sodium channels (NaV1.8) on muscle nerve fibers in pain-free humans and patients with chronic myofascial temporomandibular disorders. J Headache Pain. 2014;15(1):1–12. doi:10.1186/1129-2377-15-63.
  • Dawson A, Stensson N, Ghafouri B,  Gerdle B, List T, Svensson P, Ernberg M. Dopamine in plasma – a biomarker for myofascial TMD pain?. J Headache Pain. 2016;17(1). doi:10.1186/s10194-016-0656-3
  • Sato J, Segami N, Kaneyama K, Yoshimura H, Fujimura K, Yoshitake Y. Relationship of calcitonin gene-related peptide in synovial tissues and temporomandibular joint pain in humans. Oral Surgery, Oral Med Oral Pathol Oral Radiol Endodontology. 2004;98(5):533–40. doi:10.1016/j.tripleo.2004.02.057.
  • Yoshida H, Fukumura Y, Fujita S, Nishida M, Iizuka T. The expression of cyclooxygenase-2 in human temporomandibular joint samples: an immunohistochemical study. J Oral Rehabil. 2002;29(12):1146–52. doi:10.1046/j.1365-2842.2002.00969.x.
  • Sato J, Segami N, Yoshitake Y, Kaneyama K, Yoshimura H, Fujimura K, Kitagawa Y. Specific expression of substance P in synovial tissues of patients with symptomatic, non-reducing internal derangement of the temporomandibular joint: comparison with clinical findings. Br J Oral Maxillofac Surg. 2007;45(5):372–77. doi:10.1016/j.bjoms.2006.09.011.
  • Tanaka E, Detamore MS, Mercuri LG. Degenerative disorders of the temporomandibular joint: etiology, diagnosis, and treatment. J Dent Res. 2008;87(4):296–307. doi:10.1177/154405910808700406.
  • Deseure K, Koek W, Colpaert FC, Adriaensen H. The 5-HT1A receptor agonist F 13640 attenuates mechanical allodynia in a rat model of trigeminal neuropathic pain. Eur J Pharmacol. 2002;456(1–3):51–57. doi:10.1016/S0014-2999(02)02640-7.
  • Nakai K, Nakae A, Oba S, Mashimo T, Ueda K. 5-HT2C receptor agonists attenuate pain-related behaviour in a rat model of trigeminal neuropathic pain. Eur J Pain. 2010;14(10):999–1006. doi:10.1016/j.ejpain.2010.04.008.
  • Sommer C. Is serotonin hyperalgesic or analgesic?. Curr Pain Headache Rep. 2006;10(2):101–06. doi:10.1007/s11916-006-0020-4.
  • Strittmatter M, Grauer M, Isenberg E, Hamann G, Fischer C, Hoffmann KH, Blaes F, Schimrigk K. Cerebrospinal fluid neuropeptides and monoaminergic transmitters in patients with trigeminal neuralgia. Headache. 1997;37(4):211–16. doi:10.1046/j.1526-4610.1997.3704211.x.
  • Qin Z, Yang L, Li N, Yue JN, Wu BS, Tang YZ, Guo YN, Lai GH, Ni JX. Clinical study of cerebrospinal fluid neuropeptides in patients with primary trigeminal neuralgia. Clin Neurol Neurosurg. 2016;143:111–15. doi:10.1016/j.clineuro.2016.02.012.
  • McDermott JE, Wang J, Mitchell H, Webb-Robertson BJ, Hafen R, Ramey J, Rodland KD. Challenges in biomarker discovery: combining expert insights with statistical analysis of complex omics data. Expert Opin Med Diagn. 2013;7(1):37–51. doi:10.1517/17530059.2012.718329.
  • Xu J, Liu Y, Deng M, Li J, Cai H, Meng Q, Fang W, Long X, Ke J. MicroRNA221-3p modulates Ets-1 expression in synovial fibroblasts from patients with osteoarthritis of temporomandibular joint. Osteoarthr Cartil. 2016;24(11):2003–11. doi:10.1016/j.joca.2016.06.011.
  • Hackshaw KV, Aykas DP, Sigurdson GT, Plans M, Madiai F, Yu L, Buffington CAT, Giusti MM, Rodriguez-Saona L. Metabolic fingerprinting for diagnosis of fibromyalgia and other rheumatologic disorders. J Biol Chem. 2019;294(7):2555–68. doi:10.1074/jbc.RA118.005816.
  • Farajzadeh A, Bathaie SZ, Arabkheradmand J, Ghodsi SM, Faghihzadeh S. Different pain states of trigeminal neuralgia make significant changes in the plasma proteome and some biochemical parameters: a preliminary cohort study. J Mol Neurosci. 2018;66(4):524–34. doi:10.1007/s12031-018-1183-2.
  • Martucci KT, MacKey SC. Neuroimaging of pain: human evidence and clinical relevance of central nervous system processes and modulation. Anesthesiology. 2018;128(6):1241–54. doi:10.1097/ALN.0000000000002137.
  • Tracey I. Neuroimaging mechanisms in pain: from discovery to translation. Pain. 2017;158(4):S115–S122. doi:10.1097/j.pain.0000000000000863.
  • Moayedi M, Weissman-Fogel I, Crawley AP, Goldberg MB, Freeman BV, Tenenbaum HC, Davis KD. Contribution of chronic pain and neuroticism to abnormal forebrain gray matter in patients with temporomandibular disorder. Neuroimage. 2011;55(1):277–86. doi:10.1016/j.neuroimage.2010.12.013.
  • Walitt B, Ceko M, Gracely JL, Gracely RH, Gracely JL, Gracely RH. Neuroimaging of central sensitivity syndromes: key insights from the scientific literature. Curr Rheumatol Rev. 2016;12(1):55–87. doi:10.2174/1573397112666151231111104.
  • Nebel MB, Folger S, Tommerdahl M, Hollins M, McGlone F, Essick G. Temporomandibular disorder modifies cortical response to tactile stimulation. J Pain. 2010;11(11):1083–94. doi:10.1016/j.jpain.2010.02.021.
  • Alonso AA, Koutlas IG, Leuthold AC, Lewis SM, Georgopoulos AP. Cortical processing of facial tactile stimuli in temporomandibular disorder as revealed by magnetoencephalography. Exp Brain Res. 2010;204(1):33–45. doi:10.1007/s00221-010-2291-6.
  • Gerstner GE, Gracely RH, Deebajah A, Ichesco E, Quintero A, Clauw DJ, Sundgren PC. Posterior insular molecular changes in myofascial pain. J Dent Res. 2012;91(5):485–90. doi:10.1177/0022034512443366.
  • Harfeldt K, Alexander L, Lam J, Månsson S, Westergren H, Svensson P, Sundgren PC, Alstergren P. Spectroscopic differences in posterior insula in patients with chronic temporomandibular pain. Scand J Pain. 2018;18(3):351–61. doi:10.1515/sjpain-2017-0159.
  • DeSouza DD, Hodaie M, Davis KD. Structural magnetic resonance imaging can identify trigeminal system abnormalities in classical trigeminal neuralgia. Front Neuroanat. 2016;10:(October):1–14. doi:10.3389/fnana.2016.00095.
  • Anderson VC, Berryhill PC, Sandquist MA, Ciaverella DP, Nesbit GM, Burchiel KJ. High-resolution three-dimensional magnetic resonance angiography and three-dimensional spoiled gradient-recalled imaging in the evaluation of neurovascular compression in patients with trigeminal neuralgia: a double-blind pilot study. Neurosurgery. 2006;58(4):666–71. doi:10.1227/01.NEU.0000197117.34888.DE.
  • Lorenzoni J, David P, Levivier M. Patterns of neurovascular compression in patients with classic trigeminal neuralgia: a high-resolution MRI-based study. Eur J Radiol. 2012;81(8):1851–57. doi:10.1016/j.ejrad.2009.09.017.
  • Maarbjerg S, Wolfram F, Gozalov A, Olesen J, Bendtsen L. Significance of neurovascular contact in classical trigeminal neuralgia. Brain. 2015;138(2):311–19. doi:10.1093/brain/awu349.
  • Hung -PS-P, Chen DQ, Davis KD, Zhong J, Hodaie M. Predicting pain relief: use of pre-surgical trigeminal nerve diffusion metrics in trigeminal neuralgia. NeuroImage Clin. 2017;15:710–18. doi:10.1016/j.nicl.2017.06.017.
  • Chen ST, Yang JT, Yeh MY, Weng HH, Chen CF, Tsai YH. Using diffusion tensor imaging to evaluate microstructural changes and outcomes after radiofrequency rhizotomy of trigeminal nerves in patients with trigeminal neuralgia. PLoS One. 2016;11(12):e0167584. doi:10.1371/journal.pone.0167584.
  • Lee Y-L, Chen S-T, Yang J-T, Weng -H-H, Wang H-L, Tsai Y-H. Diffusivity parameters of diffusion tensor imaging and apparent diffusion coefficient as imaging markers for predicting the treatment response of patients with trigeminal neuralgia. J Neurosurg. 2019:1–7. doi:10.3171/2019.2.jns183008.
  • Moayedi M, Hodaie M. Trigeminal nerve and white matter brain abnormalities in chronic orofacial pain disorders. PAIN Rep. 2019;4(4):e755. doi:10.1097/pr9.0000000000000755.
  • Wilcox SL, Gustin SM, Eykman EN, Fowler G, Peck CC, Murray GM, Henderson LA. Trigeminal nerve anatomy in neuropathic and non-neuropathic orofacial pain patients. J Pain. 2013;14(8):865–72. doi:10.1016/j.jpain.2013.02.014.
  • Lin CS. Brain signature of chronic orofacial pain: a systematic review and meta-analysis on neuroimaging research of trigeminal neuropathic pain and temporomandibular joint disorders. PLoS One. 2014;9:4. doi:10.1371/journal.pone.0094300.
  • Youssef AM, Gustin SM, Nash PG, Reeves JM, Petersen ET, Peck CC, Murray GM, Henderson LA. Differential brain activity in subjects with painful trigeminal neuropathy and painful temporomandibular disorder. Pain. 2014;155(3):467–75. doi:10.1016/j.pain.2013.11.008.
  • Rolke R, Baron R, Maier C, Tölle TR, Treede DR, Beyer A, Binder A, Birbaumer N, Birklein F, Bötefür IC, et al. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): standardized protocol and reference values. Pain. 2006;123(3):231–43. doi:10.1016/j.pain.2006.01.041.
  • Magerl W, Krumova EK, Baron R, Tölle T, Treede RD, Maier C. Reference data for quantitative sensory testing (QST): refined stratification for age and a novel method for statistical comparison of group data. Pain. 2010;151(3):598–605. doi:10.1016/j.pain.2010.07.026.
  • Pfau DB, Krumova EK, Treede R-D, Baron R, Toelle T, Birklein F, Eich W, Geber C, Gerhardt A, Weiss T, et al. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): reference data for the trunk and application in patients with chronic postherpetic neuralgia. Pain. 2014;155(5):1002–15. doi:10.1016/j.pain.2014.02.004.
  • Backonja MM, Attal N, Baron R, Bouhassira D, Drangholt M, Dyck PJ, Edwards RR, Freeman R, Gracely R, Haanpaa MH, et al. Value of quantitative sensory testing in neurological and pain disorders: neuPSIG consensus. Pain. 2013;154(9):1807–19. doi:10.1016/j.pain.2013.05.047.
  • Greenspan JD, Slade GD, Bair E, Dubner R, Fillingim RB, Ohrbach R, Knott C, Diatchenko L, Liu Q, Maixner W, et al. Pain sensitivity and autonomic factors associated with development of TMD: the OPPERA prospective cohort study. J Pain. 2013;14(12 SUPPL):T63–T74.e6. doi:10.1016/j.jpain.2013.06.007.
  • Yang G, Baad-Hansen L, Wang K, Fu K, Xie QF, Svensson P. Somatosensory abnormalities in Chinese patients with painful temporomandibular disorders. J Headache Pain. 2016;17(1):31. doi:10.1186/s10194-016-0632-y.
  • Kothari SF, Baad-Hansen L, Oono Y, Svensson P. Somatosensory assessment and conditioned pain modulation in temporomandibular disorders pain patients. Pain. 2015;156(12):2545–55. doi:10.1097/j.pain.0000000000000325.
  • Yang G, Baad-Hansen L, Wang K, Xie QF, Svensson P. A study on variability of quantitative sensory testing in healthy participants and painful temporomandibular disorder patients. Somatosens Mot Res. 2014;31(2):62–71. doi:10.3109/08990220.2013.869493.
  • Carvalho GF, Chaves TC, Florencio LL, Dach F, Bigal ME, Bevilaqua-Grossi D. Reduced thermal threshold in patients with temporomandibular disorders. J Oral Rehabil. 2016;43(6):401–08. doi:10.1111/joor.12386.
  • La Touche R, Paris-Alemany A, Hidalgo-Pérez A, López-de-Uralde-Villanueva I, Angulo-Diaz-Parreño S, Muñoz-García D. Evidence for central sensitization in patients with temporomandibular disorders: a systematic review and meta-analysis of observational studies. Pain Pract. 2018;18(3):388–409. doi:10.1111/papr.12604.
  • Slade GD, Sanders AE, Ohrbach R, Fillingim RB, Dubner R, Gracely RH, Bair E, Maixner W, Greenspan JD. Pressure pain thresholds fluctuate with, but do not usefully predict, the clinical course of painful temporomandibular disorder. Pain. 2014;155(10):2134–43. doi:10.1016/j.pain.2014.08.007.
  • Fillingim RB, Slade GD, Greenspan JD, Dubner R, Maixner W, Bair E, Ohrbach R. Long-term changes in biopsychosocial characteristics related to temporomandibular disorder: findings from the OPPERA study. Pain. 2018;159(11):2403–13. doi:10.1097/j.pain.0000000000001348.
  • Ichida MC, Alvarenga da Silva L, Teixeira MJ, de Siqueira JTT, de Siqueira SRDT. Functional and sensory evaluation of patients with idiopathic trigeminal neuralgia: comparison with controls. Clin Neurol Neurosurg. 2015;130:114–21. doi:10.1016/j.clineuro.2014.12.024.
  • Sinay VJ, Bonamico LH, Dubrovsky A. Subclinical sensory abnormalities in trigeminal neuralgia. Cephalalgia. 2003;23(7):541–44. doi:10.1046/j.1468-2982.2003.00581.x.
  • Flor H, Rasche D, Islamian AP, Rolko C, Yilmaz P, Ruppolt M, Capelle HH, Tronnier V, Krauss JK. Subtle sensory abnormalities detected by quantitative sensory testing in patients with trigeminal neuralgia. Pain Physician. 2016;19:507–18.
  • Maier C, Baron R, Tölle TR, Binder A, Birbaumer N, Birklein F, Gierthmühlen J, Flor H, Geber C, Huge V, et al. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): somatosensory abnormalities in 1236 patients with different neuropathic pain syndromes. Pain. 2010;150(3):439–50. doi:10.1016/j.pain.2010.05.002.
  • Younis S, Maarbjerg S, Reimer M, Wolfram F, Olesen J, Baron R, Bendtsen L. Quantitative sensory testing in classical trigeminal neuralgia-a blinded study in patients with and without concomitant persistent pain. Pain. 2016;157(7):1407–14. doi:10.1097/j.pain.0000000000000528.
  • Ichida MC, de Almeida AN, da Nobrega JCM, Teixeira MJ, de Siqueira JTT, de Siqueira SRDT. Sensory abnormalities and masticatory function after microvascular decompression or balloon compression for trigeminal neuralgia compared with carbamazepine and healthy controls. J Neurosurg. 2015;122(6):1315–23. doi:10.3171/2014.9.JNS14346.
  • Smith SM, Dworkin RH, Turk DC, Baron R, Polydefkis M, Tracey I, Borsook D, Edwards RR, Harris RE, Wager TD, et al. The potential role of sensory testing, skin biopsy, and functional brain imaging as biomarkers in chronic pain clinical trials: IMMPACT considerations. J Pain. 2017;18(7):757–77. doi:10.1016/j.jpain.2017.02.429.