Genetics of pain: From rare Mendelian disorders to genetic predisposition to pain

Genetics of pain: From rare Mendelian disorders to genetic predisposition to pain

Authors

  • Zakira Naureen Department of Biological Sciences and Chemistry, College of Arts and Sciences, University of Nizwa, Nizwa, Oman
  • Lorenzo Lorusso ASST Lecco, UOC Neurology and Stroke Unit, Merate (LC), Italy
  • Paolo Manganotti Clinical Unit of Neurology, Department of Medicine, Surgery and Health Sciences, Cattinara University Hospital ASUGI, University of Trieste, Trieste, Italy
  • Paola Caruso Clinical Unit of Neurology, Department of Medicine, Surgery and Health Sciences, Cattinara University Hospital ASUGI, University of Trieste, Trieste, Italy
  • Giulia Mazzon Clinical Unit of Neurology, Department of Medicine, Surgery and Health Sciences, Cattinara University Hospital ASUGI, University of Trieste, Trieste, Italy
  • Stefano Cecchin MAGI’S LAB, Rovereto (TN), Italy
  • Giuseppe Marceddu MAGI EUREGIO, Bolzano, Italy
  • Matteo Bertelli MAGI’S LAB, Rovereto (TN), Italy; MAGI EUREGIO, Bolzano, Italy; EBTNA-LAB, Rovereto (TN), Italy

Keywords:

Chronic pain, painlessness, pain predisposition, genetic test

Abstract

Background and aim of the work: Pain is defined by the International Association for the Study of Pain as “an unpleasant sensory and emotional experience associated with actual or potential tissue damage, or described in terms of such damage”. In this mini-review, we focused on the Mendelian disorders with chronic pain as the main characteristic or where pain perception is disrupted, and on the polymorphisms that can impart susceptibility to chronic pain. Methods: We searched PubMed and Online Mendelian Inheritance in Man (OMIM) databases and selected only syndromes in which pain or insensitivity to pain were among the main characteristics. Polymorphisms were selected from the database GWAS catalog (https://www.ebi.ac.uk/gwas/home). Results: We retrieved a total of 28 genes associated with Mendelian inheritance in which pain or insensitivity to pain were the main characteristics and 70 polymorphisms associated with modulation of pain perception. Conclusions: This mini-review highlights the importance of genetics in phenotypes characterized by chronic pain or pain insensitivity. We think that an effective genetic test should analyze all genes associated with Mendelian pain disorders and all SNPs that can increase the risk of pain.

References

[1] https://www.iasp-pain.org/terminology?navItemNumber=576#Pain
[2] Bolles RC, Fanselow MS. Endorphins and behavior. Annu Rev Psychol 1982;33:87–101
[3] Garland EL. Pain processing in the human nervous system: a selective review of nociceptive and biobehavioral pathways. Prim Care 2012;39:561-71
[4] Dubin AE, Patapoutian A. Nociceptors: the sensors of the pain pathway. J Clin Invest 2010;120:3760-3772
[5] Young EE, Lariviere WR, Belfer I. Genetic basis of pain variability. J Med Genet 2012;49:1–19
[6] Diatchenko L, Slade GD, Nackley AG, et al. Genetic basis for individual variations in pain perception and the development of a chronic pain condition. Hum Mol Genet 2005;14:135–143
[7] Lacroix-Fralish ML, Mogil JS. Progress in genetic studies of pain and analgesia. Annu Rev Pharmacol Toxicol 2009;49:97–121
[8] Reimann F, Cox JJ, Belfer I, Diatchenko L, Zaykin DV, Mchale DP, Drenth JP, Dai F, Wheeler J, Sanders F, Wood L, Wu TX, Karppinen J, Nikolajsen L, Mannikko M, Max MB, Kiselycznyk C, Poddar M, Te Morsche RH, Smith S, Gibson D, Kelempisioti A, Maixner W, Gribble FM, Woods CG. Pain perception is altered by a nucleotide polymorphism in SCN9A. Proc Natl Acad Sci USA 2010;107:5148–5153
[9] Neely GG, Hess A, Costigan M, Keene AC, Goulas S, Langeslag M, Griffin RS, Belfer, I, Dai F, Smith SB, Diatchenko L, Gupta V, et al. A genome-wide Drosophila screen for heat nociception identifies alpha-2 delta-3 as an evolutionarily conserved pain gene. Cell 2010;143:628-638
[10] Costigan M, Belfer I, Griffin RS, Dai F, Barrett LB, Coppola G, Wu T, Kiselycznyk C, Poddar M, Lu Y, Diatchenko L, Smith S, Cobos EJ, Zaykin D, Allchorne A, Gershon E, Livneh J, Shen PH, Nikolajsen L, Karppinen J, Männikkö M, Kelempisioti A, Goldman D, Maixner W, Geschwind DH, Max MB, Seltzer Z, Woolf CJ. Multiple chronic pain states are associated with a common amino acid-changing allele in KCNS1. Brain 2010;133:2519-2527
[11] Nissenbaum J, Devor M, Seltzer Z, Gebauer M, Michaelis M, Tal M, Dorfman R, Abitbul-Yarkoni M, Lu Y, Elahipanah T, delCanho S, Minert A, Fried K, Persson AK, Shpigler H, Shabo E, Yakir B, Pisanté A, Darvasi A. Susceptibility to chronic pain following nerve injury is genetically affected by CACNG2. Genome Res 2010;20:1180-1190
[12] Kambur O, Kaunisto MA, Winsvold BS, Wilsgaard T, Stubhaug A, Zwart JA, Kalso E, Nielsen CS. Genetic variation in P2RX7 and pain tolerance. Pain 2018;159:1064-1073
[13] Duan G, Han C, Wang Q, Guo S, Zhang Y, Ying Y, Huang P, Zhang L, Macala L, Shah P, Zhang M, Li N, Dib-Hajj SD, Waxman SG, Zhang X. A SCN10A SNP biases human pain sensitivity. Mol Pain 2016;12:1744806916666083
[14] Gonzalez-Lopez E, Kawasawa Y, Walter V, Zhang L, Koltun W, Huang X, Vrana K, Coates M. Homozygosity for the SCN10A polymorphism rs6795970 is associated with hypoalgesic inflammatory bowel disease phenotype. Front Med 2018;5:324
[15] Lee E, Takita C, Wright JL, et al. Genome-wide enriched pathway analysis of acute post-radiotherapy pain in breast cancer patients: a prospective cohort study. Hum Genomics 2019; 13: 28
[16] Johnston KJA, Adams MJ, Nicholl BI, et al. Genome-wide association study of multisite chronic pain in UK Biobank. PLoS Genet 2019; 15: e1008164
[17] Suri P, Palmer MR, Tsepilov YA, et al. Genome-wide meta-analysis of 158,000 individuals of European ancestry identifies three loci associated with chronic back pain. PLoS Genet 2018; 14: e1007601
[18] Peters MJ, Broer L, Willemen HL, et al. Genome-wide association study meta-analysis of chronic widespread pain: evidence for involvement of the 5p15.2 region. Ann Rheum Dis 2013; 72: 427-36
[19] Meng W, Deshmukh HA, van Zuydam NR, et al. A genome-wide association study suggests an association of Chr8p21.3 (GFRA2) with diabetic neuropathic pain. Eur J Pain 2015; 19: 392-9
[20] Lee E, Takita C, Wright JL, et al. Genome-wide enriched pathway analysis of acute post-radiotherapy pain in breast cancer patients: a prospective cohort study. Hum Genomics 2019; 13: 28
[21] Warner SC, van Meurs JB, Schiphof D, et al. Genome-wide association scan of neuropathic pain symptoms post total joint replacement highlights a variant in the protein-kinase C gene. Eur J Hum Genet 2017; 25: 446-51
[22] Meng W, Adams MJ, Palmer CNA, et al. Genome-wide association study of knee pain identifies associations with GDF5 and COL27A1 in UK Biobank. Commun Biol 2019; 2: 321
[23] Meng W, Deshmukh HA, Donnelly LA, et al. A genome-wide association study provides evidence of sex-specific involvement of chr1p35.1 (ZSCAN20-TLR12P) and chr8p23.1 (HMGB1P46) with diabetic neuropathic pain. EBioMedicine 2015; 2: 1386-93
[24] Reyes-Gibby CC, Wang J, Silvas MR, Yu RK, Hanna EY, Shete S. Genome-wide association study suggests common variants within RP11-634B7.4 gene influencing severe pre-treatment pain in head and neck cancer patients. Sci Rep 2016; 6: 34206
[25] Reyes-Gibby CC, Wang J, Yeung SJ, et al. Genome-wide association study identifies genes associated with neuropathy in patients with head and neck cancer. Sci Rep 2018; 8: 8789

Downloads

Published

09-11-2020

How to Cite

1.
Naureen Z, Lorusso L, Manganotti P, Caruso P, Mazzon G, Cecchin S, et al. Genetics of pain: From rare Mendelian disorders to genetic predisposition to pain. Acta Biomed [Internet]. 2020 Nov. 9 [cited 2024 Jul. 18];91(13-S):e2020010. Available from: https://mattioli1885journals.com/index.php/actabiomedica/article/view/10682