Article Data

  • Views 272
  • Dowloads 45

Original Research

Open Access

Identifying Genetic And Environmental Risk Factors For Chronic Orofacial Pain Syndromes: Human Models

  • Ze’ev Seltzer1,*,
  • Ruslan Dorfman2

1University of Toronto Centre for the Study of Pain, Faculty of Dentistry, Toronto, Ontario, Canada

2Canadian Institute of Health Research, Department of Genetics, Hospital for Sick Children, Toronto, Ontario, Canada

DOI: 10.11607/jofph.18311 Vol.18,Issue 4,December 2004 pp.311-317

Published: 30 December 2004

*Corresponding Author(s): Ze’ev Seltzer E-mail: zeev.seltzer@utoronto.ca

Abstract

Chronic orofacial pain syndromes are produced by nerve injury, diseases, and toxins. They constitute an unsolved medical problem because they affect a considerable number of adults and are difficult to treat. There is a remarkable variability among adults in terms of susceptibility to chronic orofacial pain and its characteristics, which suggests that these syndromes are complex heritable traits controlled by alleles of certain polymorphic genes that interact with the environment. Each syndrome is assumed to be deter-mined by a unique set of genes. In the present report, a practical study design is proposed to identify the genes responsible for interindividual variability in orofacial pain levels. This design is based on research strategies that have been used for studying other human diseases as well as pain syndromes outside the orofacial region. Specifically, this design has been used successfully by the authors and others over the past 8 years to study chronic pain syn-dromes such as migraines, radiculopathy, amputation pain, and postmastectomy pain. The strategies used to study these topics have been adapted to address the unique problems of orofacial pain. The authors believe that the study of genetics provides a novel research approach from which to identify targets for the development of individually tailored approaches in orofacial pain medicine, such as diagnostic and prognostic kits and novel drugs that would prevent pain chronicity in susceptible individuals or alleviate it once it had developed. This report focuses on human models. A follow-up report is intended to extend this design into animal models of oro-facial pain syndromes.

Keywords

genetic susceptibility; orofacial pain; polymorphic genes

Cite and Share

Ze’ev Seltzer,Ruslan Dorfman. Identifying Genetic And Environmental Risk Factors For Chronic Orofacial Pain Syndromes: Human Models. Journal of Oral & Facial Pain and Headache. 2004. 18(4);311-317.

References

1. Bennett GJ. Neuropathic pain in the orofacial region: Clinical and research challenges. J Orofac Pain 2004;18: 281–286.

2. Zakrzewska J. Classification issues related to trigeminal pain. J Orofac Pain 2004;18:325–331.

3. Truelove E. Management issues of neuropathic trigeminal pain from a dental perspective. J Orofac Pain 2004;18: 374–380.

4. Watson CPN. Management issues of neuropathic trigeminal pain from a medical perspective. J Orofac Pain 2004;18:366–373.

5. Kirkpatrick DB. Familial trigeminal neuralgia: Case report. Neurosurgery 1989;24:758–761.

6. Ophoff RA, Terwindt GM, Vergouwe MN, et al. Familial hemiplegic migraine and episodic ataxia type-2 are caused by mutations in the Ca2+ channel gene CACNL1A4. Cell 1996;87:543–552.

7. Kusumi M, Araki H, Ijiri T, et al. Serotonin 2C receptor gene Cys23Ser polymorphism: A candidate genetic risk factor of migraine with aura in Japanese population. Acta Neurol Scand 2004;109:407–409.

8. Estevez M, Gardner KL. Update on the genetics of migraine. Hum Genet 2004;114:225–235.

9. Violon A, Giugea D. Familial models for chronic pain. Pain 1984;18:199–203.

10. Edwards PW, Zeichner A, Kuczmierczyk AR, Boczkowski J. Familial pain models: The relationship between family history of pain and current pain experience. Pain 1985;21:379–384.

11. Heikkila JK, Koskenvuo M, Heliovaara M, et al. Genetic and environmental factors in sciatica. Evidence from a nationwide panel of 9365 adult twin pairs. Ann Med 1989;21:393–398.

12. Bengtsson B, Thorson J. Back pain: A study of twins. Acta Genet Med Gemellol (Roma) 1991;40:83–90.

13. Golik A, Modai D, Pervin R, Marcus EL, Fried K. Auto-somal dominant carpal tunnel syndrome in a Karaite family. Isr J Med Sci 1988;24:295–297.

14. Matsui H, Terahata N, Tsuji H, Hirano N, Naruse Y. Familial predisposition and clustering for juvenile lumbar disc herniation. Spine 1992;17:1323–1328.

15. Dyck PJ, Low PA, Stevens JC. “Burning feet” as the only manifestation of dominantly inherited sensory neuropathy. Mayo Clin Proc 1983;58:426–429.

16. Yunus MB, Khan MA, Rawlings KK, Green JR, Olson JM, Shah S. Genetic linkage analysis of multicase families with fibromyalgia syndrome. J Rheumatol 1999;26: 408–412.

17. Mailis A, Wade J. Profile of Caucasian women with possible genetic predisposition to reflex sympathetic dystrophy: A pilot study. Clin J Pain 1994;10:210–217.

18. Kemler MA, van de Vusse AC, van den Berg-Loonen EM, Barendse GA, van Kleef M, Weber WE. HLA-DQ1 associated with reflex sympathetic dystrophy. Neurology 1999; 53:1350–1351.

19. van de Beek WJ, Roep BO, van der Slik AR, Giphart MJ, van Hilten BJ. Susceptibility loci for complex regional pain syndrome. Pain 2003;103:93–97.

20. Sato M, Ohashi J, Tsuchiya N, et al. Association of HLA-A*3303-B*4403-DRB1*1302 haplotype, but not of TNFa promoter and NKp30 polymorphism, with postherpetic neuralgia (PHN) in the Japanese population. Genes Immun 2002;3:477–481.

21. Cohen H, Buskila D, Neumann L, Ebstein RP. Confirmation of an association between fibromyalgia and serotonin transporter promoter region (5-HTTLPR) polymor-phism, and relationship to anxiety-related personality traits. Arthritis Rheum 2002;46:845–847.

22. Foster DC, Sazenski TM, Stodgell CJ. Impact of genetic variation in interleukin-1 receptor antagonist and melano-cortin-1 receptor genes on vulvar vestibulitis syndrome. J Reprod Med 2004;49:503–509.

23. Devor M, Raber P. Heritability of symptoms in an experimental model of neuropathic pain. Pain 1990;42:51–67.

24. Mogil JS, Wilson SG, Bon K, et al. Heritability of nociception I: Responses of 11 inbred mouse strains on 12 measures of nociception. Pain 1999;80:67–82.

25. Shir Y, Zeltser R, Vatine JJ, et al. Correlation of intact sensibility and neuropathic pain-related behaviors in eight inbred and outbred rat strains and selection lines. Pain 2001;90:75–82.

26. Seltzer Z, Wu T, Max MB, Diehl SR. Mapping a gene for neuropathic pain-related behavior following peripheral neurectomy in the mouse. Pain 2001;93:101–106.

27. Max MB. Assessing pain candidate gene studies. Pain 2004;109:1–3.

28. Risch N, Merikangas K. The future of genetic studies of complex human diseases. Science 1996;273:1516–1517.

29. Belfer I, Wu T, Kingman A, Krishnaraju RK, Goldman D, Max MB. Candidate gene studies of human pain mechanisms: Methods for optimizing choice of polymorphisms and sample size. Anesthesiology 2004;100:1562–1572.

30. MacGregor AJ. The heritability of pain in humans. In: Mogil JS (ed). Progress in Pain Research and Manage-ment. Vol 28: The genetics of pain. Seattle: IASP Press, 2004:151–170.

31. Ulrich V, Gervil M, Olesen J. The relative influence of environment and genes in episodic tension-type headache. Neurology 2004;62:2065–2069.

32. Neumann L, Buskila D. Epidemiology of fibromyalgia. Curr Pain Headache Rep 2003;7:362–368.

33. Shir Y, Seltzer Z. Heat hyperalgesia following partial sciatic ligation in rats: Interacting nature and nurture. Neuroreport 2001;12:809–813.

34. Raber P, Devor M. Social variables affect phenotype in the neuroma model of neuropathic pain. Pain 2002;97: 139–150.

35. Mogil JS, Seltzer Z, Devor M. Social and environmental influences on pain: Implications for pain genetics. In: Mogil JS (ed). Progress in Pain Research and Management. Vol 28: The genetics of pain. Seattle: IASP Press, 2004:257–282.

36. Wall FE, Henkel RD, Stern MP, Jenson HB, Moyer MP. An efficient method for routine Epstein-Barr virus immortalization of human B lymphocytes. In Vitro Cell Dev Biol Anim 1995;31:156–159.

37. Makowski GS, Nadeau FL, Hopfer SM. Single tube multiplex PCR detection of 27 cystic fibrosis mutations and 4 polymorphisms using neonatal blood samples collected on Guthrie cards. Ann Clin Lab Sci 2003;33:243–250.

38. Moore A. Of mice and Mendel: The predicted rise in the use of knock-out and transgenic mice should cause us to reflect on our justification for the use of animals in research. EMBO Rep 2001;2:554–558 [erratum 2002; 2:197].

39. National Centre for Biotechnology Information. Comparative Maps. Available at: http://www.ncbi.nlm.nih.gov/Homology

40. Costigan M, Griffin RS, Woolf CJ. Microarray analysis of the pain pathway. In: Mogil JS (ed). Progress in Pain Research and Management. Vol 28: The genetics of pain. Seattle: IASP Press, 2004:65–84.

41. Seltzer Z. The relevance of animal neuropathy models for chronic pain in humans. Seminars Neurosci 1995;7: 211–219.

42. Harbo HF, Datta P, Oturai A, et al. Two genome-wide linkage disequilibrium screens in Scandinavian multiple sclerosis patients. J Neuroimmunol 2003;143:101–106.

43. Grossman I, Avidan N, Singer C, et al. Genomic profiling of interpopulation diversity guides prioritization of candidate-genes for autoimmunity. Genes Immun 2004;5: 493–504.

Abstracted / indexed in

Science Citation Index (SCI)

Science Citation Index Expanded (SCIE)

BIOSIS Previews

Scopus

Cumulative Index to Nursing and Allied Health Literature (CINAHL)

Submission Turnaround Time

Conferences

Top