Genetic factors associated with fibromyalgia: a narrative review

Authors

DOI:

https://doi.org/10.33448/rsd-v11i3.26421

Keywords:

Fibromyalgia; Genetic association studies; Genetic predisposition to disease; Genetic polymorphism.

Abstract

Fibromyalgia is a rheumatic disorder whose main symptom is chronic generalized pain accompanied by a set of symptoms such as sleep disturbance, depression, and chronic fatigue, among others. Its pathophysiology is complex, of multifactorial origin, including the influence of genetic factors. It has been shown that individuals with fibromyalgia show a pattern of family aggregation and that the probability of an individual developing this condition is about 50% attributed to genetic factors. In this sense, the present article aims to carry out a narrative review of the literature on the genetic aspects of fibromyalgia in its development and severity of symptoms. To this end, searches were carried out in PubMed and Scopus databases, using specific descriptors: “fibromyalgia” and “polymorphism”. Polymorphisms that influence pain modulation, such as those that occur in genes of the monoaminergic pathway or of catecholamine metabolism, are frequently found in studies of association with fibromyalgia. However, other target genes have emerged, related to the pathways responsible for the mechanisms of the most diverse symptoms that can affect these patients, such as neuroplasticity, neurotransmission, inflammation, vascularization, oxidative stress, cell cycle, among others.

References

Ablin, J. N., & Buskila, D. (2014). Predicting fibromyalgia, a narrative review: are we better than fools and children?. European journal of pain (London, England), 18(8), 1060–1066. https://doi.org/10.1002/j.1532-2149.2014.00481.x

Ablin, J. N., & Buskila, D. (2015). Update on the genetics of the fibromyalgia syndrome. Best practice & research. Clinical rheumatology, 29(1), 20–28. https://doi.org/10.1016/j.berh.2015.04.018

Affleck, G., Urrows, S., Tennen, H., Higgins, P., & Abeles, M. (1996). Sequential daily relations of sleep, pain intensity, and attention to pain among women with fibromyalgia. Pain, 68(2-3), 363–368. https://doi.org/10.1016/s0304-3959(96)03226-5

Araya-Quintanilla, F., Gutiérrez-Espinoza, H., Muñoz-Yánez, M. J., Cavero-Redondo, I., Álvarez-Bueno, C., & Martinez-Vizcaíno, V. (2020). Effectiveness of a multicomponent treatment versus conventional treatment in patients with fibromyalgia: Study protocol. Medicine, 99(4), e18833. https://doi.org/10.1097/MD.0000000000018833

Arnold, L. M., Hudson, J. I., Hess, E. V., Ware, A. E., Fritz, D. A., Auchenbach, M. B., Starck, L. O., & Keck, P. E., Jr (2004). Family study of fibromyalgia. Arthritis and rheumatism, 50(3), 944–952. https://doi.org/10.1002/art.20042

Bair, M. J., Robinson, R. L., Katon, W., & Kroenke, K. (2003). Depression and pain comorbidity: a literature review. Archives of internal medicine, 163(20), 2433–2445. https://doi.org/10.1001/archinte.163.20.2433

Balkarli, A., Sengül, C., Tepeli, E., Balkarli, H., & Cobankara, V. (2014). Synaptosomal-associated protein 25 (Snap-25) gene polymorphism frequency in fibromyalgia syndrome and relationship with clinical symptoms. BMC musculoskeletal disorders, 15, 191. https://doi.org/10.1186/1471-2474-15-191

Baraniuk, J. N., Whalen, G., Cunningham, J., & Clauw, D. J. (2004). Cerebrospinal fluid levels of opioid peptides in fibromyalgia and chronic low back pain. BMC musculoskeletal disorders, 5, 48. https://doi.org/10.1186/1471-2474-5-48

Barbosa, F. R., Matsuda, J. B., Mazucato, M., de Castro França, S., Zingaretti, S. M., da Silva, L. M., Martinez-Rossi, N. M., Júnior, M. F., Marins, M., & Fachin, A. L. (2012). Influence of catechol-O-methyltransferase (COMT) gene polymorphisms in pain sensibility of Brazilian fibromialgia patients. Rheumatology international, 32(2), 427–430. https://doi.org/10.1007/s00296-010-1659-z

Becker, R. M., da Silva, V. K., Machado, F., dos Santos, A. F., Meireles, D. C., Mergener, M., dos Santos, G. A., & de Andrade, F. M. (2010). Association between environmental quality, stress and APOE gene variation in fibromyalgia susceptibility determination. Revista brasileira de reumatologia, 50(6), 617–624. https://doi.org/10.1590/S0482-50042010000600003

Bennett, R. M., Friend, R., Jones, K. D., Ward, R., Han, B. K., & Ross, R. L. (2009). The Revised Fibromyalgia Impact Questionnaire (FIQR): validation and psychometric properties. Arthritis research & therapy, 11(4), R120. https://doi.org/10.1186/ar2783

Berger, A., Dukes, E., Martin, S., Edelsberg, J., & Oster, G. (2007). Characteristics and healthcare costs of patients with fibromyalgia syndrome. International journal of clinical practice, 61(9), 1498–1508. https://doi.org/10.1111/j.1742-1241.2007.01480.x

Bilge, U., Sari, Y. E., Balcioglu, H., Yasar Bilge, N. S., Kasifoglu, T., Kayhan, M., & Unluoglu, I. (2018). Prevalence of comorbid diseases in patients with fibromyalgia: A retrospective cross-sectional study. JPMA. The Journal of the Pakistan Medical Association, 68(5), 729–732.

Bond, C., LaForge, K. S., Tian, M., Melia, D., Zhang, S., Borg, L., Gong, J., Schluger, J., Strong, J. A., Leal, S. M., Tischfield, J. A., Kreek, M. J., & Yu, L. (1998). Single-nucleotide polymorphism in the human mu opioid receptor gene alters beta-endorphin binding and activity: possible implications for opiate addiction. Proceedings of the National Academy of Sciences of the United States of America, 95(16), 9608–9613. https://doi.org/10.1073/pnas.95.16.9608

Bondy B. (2003). Common genetic risk factors for psychiatric and simatic disorders. Dialogues in clinical neuroscience, 5(2), 129–138. https://doi.org/10.31887/DCNS.2003.5.2/bbondy

Bondy, B., Spaeth, M., Offenbaecher, M., Glatzeder, K., Stratz, T., Schwarz, M., de Jonge, S., Krüger, M., Engel, R. R., Färber, L., Pongratz, D. E., & Ackenheil, M. (1999). The T102C polymorphism of the 5-HT2A-receptor gene in fibromyalgia. Neurobiology of disease, 6(5), 433–439. https://doi.org/10.1006/nbdi.1999.0262

Boomershine, C. S., & Crofford, L. J. (2009). A symptom-based approach to pharmacologic management of fibromyalgia. Nature reviews. Rheumatology, 5(4), 191–199. https://doi.org/10.1038/nrrheum.2009.25

Borchers, A. T., & Gershwin, M. E. (2015). Fibromyalgia: A Critical and Comprehensive Review. Clinical reviews in allergy & immunology, 49(2), 100–151. https://doi.org/10.1007/s12016-015-8509-4

Burckhardt, C. S., Clark, S. R., & Bennett, R. M. (1991). The fibromyalgia impact questionnaire: development and validation. The Journal of rheumatology, 18(5), 728–733.

Buskila, D., Cohen, H., Neumann, L., & Ebstein, R. P. (2004). An association between fibromyalgia and the dopamine D4 receptor exon III repeat polymorphism and relationship to novelty seeking personality traits. Molecular psychiatry, 9(8), 730–731. https://doi.org/10.1038/sj.mp.4001506

Cabo-Meseguer, A., Cerdá-Olmedo, G., & Trillo-Mata, J. L. (2017). Fibromyalgia: Prevalence, epidemiologic profiles and economic costs. Fibromialgia: prevalencia, perfiles epidemiológicos y costes económicos. Medicina clinica, 149(10), 441–448. https://doi.org/10.1016/j.medcli.2017.06.008

Carville, S. F., Arendt-Nielsen, L., Bliddal, H., Blotman, F., Branco, J. C., Buskila, D., Da Silva, J. A., Danneskiold-Samsøe, B., Dincer, F., Henriksson, C., Henriksson, K. G., Kosek, E., Longley, K., McCarthy, G. M., Perrot, S., Puszczewicz, M., Sarzi-Puttini, P., Silman, A., Späth, M., Choy, E. H., … EULAR (2008). EULAR evidence-based recommendations for the management of fibromyalgia syndrome. Annals of the rheumatic diseases, 67(4), 536–541. https://doi.org/10.1136/ard.2007.071522

Chae, J. J., Centola, M., Aksentijevich, I., Dutra, A., Tran, M., Wood, G., Nagaraju, K., Kingma, D. W., Liu, P. P., & Kastner, D. L. (2000). Isolation, genomic organization, and expression analysis of the mouse and rat homologs of MEFV, the gene for familial mediterranean fever. Mammalian genome : official journal of the International Mammalian Genome Society, 11(6), 428–435. https://doi.org/10.1007/s003350010082

Chang, F. M., Kidd, J. R., Livak, K. J., Pakstis, A. J., & Kidd, K. K. (1996). The world-wide distribution of allele frequencies at the human dopamine D4 receptor locus. Human genetics, 98(1), 91–101. https://doi.org/10.1007/s004390050166

Chen, J., Lipska, B. K., Halim, N., Ma, Q. D., Matsumoto, M., Melhem, S., Kolachana, B. S., Hyde, T. M., Herman, M. M., Apud, J., Egan, M. F., Kleinman, J. E., & Weinberger, D. R. (2004). Functional analysis of genetic variation in catechol-O-methyltransferase (COMT): effects on mRNA, protein, and enzyme activity in postmortem human brain. American journal of human genetics, 75(5), 807–821. https://doi.org/10.1086/425589

Chinn, S., Caldwell, W., & Gritsenko, K. (2016). Fibromyalgia Pathogenesis and Treatment Options Update. Current pain and headache reports, 20(4), 25. https://doi.org/10.1007/s11916-016-0556-x

Chong, L. K., Chowdry, J., Ghahramani, P., & Peachell, P. T. (2000). Influence of genetic polymorphisms in the beta2-adrenoceptor on desensitization in human lung mast cells. Pharmacogenetics, 10(2), 153–162. https://doi.org/10.1097/00008571-200003000-00007

Ciampi de Andrade, D., Maschietto, M., Galhardoni, R., Gouveia, G., Chile, T., Victorino Krepischi, A. C., Dale, C. S., Brunoni, A. R., Parravano, D. C., Cueva Moscoso, A. S., Raicher, I., Kaziyama, H., Teixeira, M. J., & Brentani, H. P. (2017). Epigenetics insights into chronic pain: DNA hypomethylation in fibromyalgia-a controlled pilot-study. Pain, 158(8), 1473–1480. https://doi.org/10.1097/j.pain.0000000000000932

Clauw D. J. (2014). Fibromyalgia: a clinical review. JAMA, 311(15), 1547–1555. https://doi.org/10.1001/jama.2014.3266

Cohen, H., Buskila, D., Neumann, L., & Ebstein, R. P. (2002). Confirmation of an association between fibromyalgia and serotonin transporter promoter region (5- HTTLPR) polymorphism, and relationship to anxiety-related personality traits. Arthritis and rheumatism, 46(3), 845–847. https://doi.org/10.1002/art.10103

Cohen, H., Neumann, L., Glazer, Y., Ebstein, R. P., & Buskila, D. (2009). The relationship between a common catechol-O-methyltransferase (COMT) polymorphism val(158) met and fibromyalgia. Clinical and experimental rheumatology, 27(5 Suppl 56), S51–S56.

Cortes-Altamirano, J. L., Olmos-Hernandez, A., Jaime, H. B., Carrillo-Mora, P., Bandala, C., Reyes-Long, S., & Alfaro-Rodríguez, A. (2018). Review: 5-HT1, 5-HT2, 5-HT3 and 5-HT7 Receptors and their Role in the Modulation of Pain Response in the Central Nervous System. Current neuropharmacology, 16(2), 210–221. https://doi.org/10.2174/1570159X15666170911121027

Costa, B., Da Pozzo, E., & Martini, C. (2012). Translocator protein as a promising target for novel anxiolytics. Current topics in medicinal chemistry, 12(4), 270–285. https://doi.org/10.2174/156802612799078720

da Silveira Alves, C. F., Caumo, W., Silvestri, J. M., Zortea, M., Dos Santos, V. S., Cardoso, D. F., Regner, A., de Souza, A. H., & Simon, D. (2020). Pain catastrophizing is associated with the Val66Met polymorphism of the brain-derived neurotrophic factor in fibromyalgia. Advances in rheumatology (London, England), 60(1), 39. https://doi.org/10.1186/s42358-020-00141-9

Davies, P. A., Pistis, M., Hanna, M. C., Peters, J. A., Lambert, J. J., Hales, T. G., & Kirkness, E. F. (1999). The 5-HT3B subunit is a major determinant of serotonin-receptor function. Nature, 397(6717), 359–363. https://doi.org/10.1038/16941

D'Agnelli, S., Arendt-Nielsen, L., Gerra, M. C., Zatorri, K., Boggiani, L., Baciarello, M., & Bignami, E. (2019). Fibromyalgia: Genetics and epigenetics insights may provide the basis for the development of diagnostic biomarkers. Molecular pain, 15, 1744806918819944. https://doi.org/10.1177/1744806918819944

de Lima, L. O., Zicarelli, C., Matsumura, A. S., Moroti-Perugini, L. R., de Castro Teixeira, D., Fernandes, K., de Oliveira Perrucini, P. D., & Poli-Frederico, R. C. (2019). Lower limb muscle strength and serotonin receptor gene polymorphism as factors associated in women with fibromyalgia. Advances in rheumatology (London, England), 59(1), 59. https://doi.org/10.1186/s42358-019-0101-9

DeRijk, R. H., Wüst, S., Meijer, O. C., Zennaro, M. C., Federenko, I. S., Hellhammer, D. H., Giacchetti, G., Vreugdenhil, E., Zitman, F. G., & de Kloet, E. R. (2006). A common polymorphism in the mineralocorticoid receptor modulates stress responsiveness. The Journal of clinical endocrinology and metabolism, 91(12), 5083–5089. https://doi.org/10.1210/jc.2006-0915

Desmeules, J., Chabert, J., Rebsamen, M., Rapiti, E., Piguet, V., Besson, M., Dayer, P., & Cedraschi, C. (2014). Central pain sensitization, COMT Val158Met polymorphism, and emotional factors in fibromyalgia. The journal of pain, 15(2), 129–135. https://doi.org/10.1016/j.jpain.2013.10.004

Desmeules, J., Piguet, V., Besson, M., Chabert, J., Rapiti, E., Rebsamen, M., Rossier, M. F., Curtin, F., Dayer, P., & Cedraschi, C. (2012). Psychological distress in fibromyalgia patients: a role for catechol-O-methyl-transferase Val158met polymorphism. Health psychology : official journal of the Division of Health Psychology, American Psychological Association, 31(2), 242–249. https://doi.org/10.1037/a0025223

Doan, L., Manders, T., & Wang, J. (2015). Neuroplasticity underlying the comorbidity of pain and depression. Neural plasticity, 2015, 504691. https://doi.org/10.1155/2015/504691

Docampo, E., Escaramís, G., Gratacòs, M., Villatoro, S., Puig, A., Kogevinas, M., Collado, A., Carbonell, J., Rivera, J., Vidal, J., Alegre, J., Estivill, X., & Rabionet, R. (2014). Genome-wide analysis of single nucleotide polymorphisms and copy number variants in fibromyalgia suggest a role for the central nervous system. Pain, 155(6), 1102–1109. https://doi.org/10.1016/j.pain.2014.02.016

Dolcino, M., Tinazzi, E., Puccetti, A., & Lunardi, C. (2020). Gene Expression Profiling in Fibromyalgia Indicates an Autoimmune Origin of the Disease and Opens New Avenues for Targeted Therapy. Journal of clinical medicine, 9(6), 1814. https://doi.org/10.3390/jcm9061814

Ebstein, R. P., Novick, O., Umansky, R., Priel, B., Osher, Y., Blaine, D., Bennett, E. R., Nemanov, L., Katz, M., & Belmaker, R. H. (1996). Dopamine D4 receptor (D4DR) exon III polymorphism associated with the human personality trait of Novelty Seeking. Nature genetics, 12(1), 78–80. https://doi.org/10.1038/ng0196-78

Egan, M. F., Kojima, M., Callicott, J. H., Goldberg, T. E., Kolachana, B. S., Bertolino, A., Zaitsev, E., Gold, B., Goldman, D., Dean, M., Lu, B., & Weinberger, D. R. (2003). The BDNF val66met polymorphism affects activity-dependent secretion of BDNF and human memory and hippocampal function. Cell, 112(2), 257–269. https://doi.org/10.1016/s0092-8674(03)00035-7

Estacion, M., Harty, T. P., Choi, J. S., Tyrrell, L., Dib-Hajj, S. D., & Waxman, S. G. (2009). A sodium channel gene SCN9A polymorphism that increases nociceptor excitability. Annals of neurology, 66(6), 862–866. https://doi.org/10.1002/ana.21895

Estévez-López, F., Aparicio, V. A., Ruiz, J. R., Martínez-González, L. J., Delgado-Fernández, M., & Álvarez-Cubero, M. J. (2018a). The TT genotype of the rs6860 polymorphism of the charged multivesicular body protein 1A gene is associated with susceptibility to fibromyalgia in southern Spanish women. Rheumatology international, 38(3), 531–533. https://doi.org/10.1007/s00296-017-3896-x

Estévez-López, F., Camiletti-Moirón, D., Aparicio, V. A., Segura-Jiménez, V., Álvarez-Gallardo, I. C., Soriano-Maldonado, A., Borges-Cosic, M., Acosta-Manzano, P., Geenen, R., Delgado-Fernández, M., Martínez-González, L. J., Ruiz, J. R., & Álvarez-Cubero, M. J. (2018b). Identification of candidate genes associated with fibromyalgia susceptibility in southern Spanish women: the al-Ándalus project. Journal of translational medicine, 16(1), 43. https://doi.org/10.1186/s12967-018-1416-8

Estévez-López, F., Guerrero-González, J. M., Salazar-Tortosa, D., Camiletti-Moirón, D., Gavilán-Carrera, B., Aparicio, V. A., Acosta-Manzano, P., Álvarez-Gallardo, I. C., Segura-Jiménez, V., Soriano-Maldonado, A., Geenen, R., Delgado-Fernández, M., Martínez-González, L. J., Ruiz, J. R., & Álvarez-Cubero, M. J. (2021b). Interplay between genetics and lifestyle on pain susceptibility in women with fibromyalgia: The al-Ándalus project. Rheumatology (Oxford, England), keab911. Advance online publication. https://doi.org/10.1093/rheumatology/keab911

Estévez-López, F., Salazar-Tortosa, D. F., Camiletti-Moirón, D., Gavilán-Carrera, B., Aparicio, V. A., Acosta-Manzano, P., Segura-Jiménez, V., Álvarez-Gallardo, I. C., Carbonell-Baeza, A., Munguía-Izquierdo, D., Geenen, R., Lacerda, E., Delgado-Fernández, M., Martínez-González, L. J., Ruiz, J. R., & Álvarez-Cubero, M. J. (2021a). Fatigue in Women with Fibromyalgia: A Gene-Physical Activity Interaction Study. Journal of clinical medicine, 10(9), 1902. https://doi.org/10.3390/jcm10091902

Fanton, S., Sandström, A., Tour, J., Kadetoff, D., Schalling, M., Jensen, K. B., Sitnikov, R., Ellerbrock, I., & Kosek, E. (2021). The translocator protein gene is associated with endogenous pain modulation and the balance between glutamate and GABA in fibromyalgia and healthy subjects: a multimodal neuroimaging study. Pain, 10.1097/j.pain.0000000000002309. Advance online publication. https://doi.org/10.1097/j.pain.0000000000002309

Fernández-de-Las-Peñas, C., Ambite-Quesada, S., Gil-Crujera, A., Cigarán-Méndez, M., & Peñacoba-Puente, C. (2012). Catechol-O-methyltransferase Val158Met polymorphism influences anxiety, depression, and disability, but not pressure pain sensitivity, in women with fibromyalgia syndrome. The journal of pain, 13(11), 1068–1074. https://doi.org/10.1016/j.jpain.2012.08.001

Ferrera, D., Gómez-Esquer, F., Peláez, I., Barjola, P., Fernandes-Magalhaes, R., Carpio, A., De Lahoz, M. E., Díaz-Gil, G., & Mercado, F. (2020). Effects of COMT Genotypes on Working Memory Performance in Fibromyalgia Patients. Journal of clinical medicine, 9(8), 2479. https://doi.org/10.3390/jcm9082479

Ferrera, D., Mercado, F., Peláez, I., Martínez-Iñigo, D., Fernandes-Magalhaes, R., Barjola, P., Écija, C., Díaz-Gil, G., & Gómez-Esquer, F. (2021). Fear of pain moderates the relationship between self-reported fatigue and methionine allele of catechol-O-methyltransferase gene in patients with fibromyalgia. PloS one, 16(4), e0250547. https://doi.org/10.1371/journal.pone.0250547

Finan, P. H., Zautra, A. J., Davis, M. C., Lemery-Chalfant, K., Covault, J., & Tennen, H. (2010). Genetic influences on the dynamics of pain and affect in fibromyalgia. Health psychology : official journal of the Division of Health Psychology, American Psychological Association, 29(2), 134–142. https://doi.org/10.1037/a0018647

Finan, P. H., Zautra, A. J., Davis, M. C., Lemery-Chalfant, K., Covault, J., & Tennen, H. (2011). COMT moderates the relation of daily maladaptive coping and pain in fibromyalgia. Pain, 152(2), 300–307. https://doi.org/10.1016/j.pain.2010.10.024

Fitzcharles, M. A., Ste-Marie, P. A., Goldenberg, D. L., Pereira, J. X., Abbey, S., Choinière, M., Ko, G., Moulin, D. E., Panopalis, P., Proulx, J., Shir, Y., & National Fibromyalgia Guideline Advisory Panel (2013). 2012 Canadian Guidelines for the diagnosis and management of fibromyalgia syndrome: executive summary. Pain research & management, 18(3), 119–126. https://doi.org/10.1155/2013/918216

Fitzcharles, M. A., Perrot, S., & Häuser, W. (2018). Comorbid fibromyalgia: A qualitative review of prevalence and importance. European journal of pain (London, England), 22(9), 1565–1576. https://doi.org/10.1002/ejp.1252

Flatters S. J. (2015). The contribution of mitochondria to sensory processing and pain. Progress in molecular biology and translational science, 131, 119–146. https://doi.org/10.1016/bs.pmbts.2014.12.004

Gaughan, D. J., Barbaux, S., Kluijtmans, L. A., & Whitehead, A. S. (2000). The human and mouse methylenetetrahydrofolate reductase (MTHFR) genes: genomic organization, mRNA structure and linkage to the CLCN6 gene. Gene, 257(2), 279–289. https://doi.org/10.1016/s0378-1119(00)00392-9

Gelernter, J., Pakstis, A. J., & Kidd, K. K. (1995). Linkage mapping of serotonin transporter protein gene SLC6A4 on chromosome 17. Human genetics, 95(6), 677–680. https://doi.org/10.1007/BF00209486

Gerra, M. C., Carnevali, D., Ossola, P., González-Villar, A., Pedersen, I. S., Triñanes, Y., Donnini, C., Manfredini, M., Arendt-Nielsen, L., & Carrillo-de-la-Peña, M. T. (2021). DNA Methylation Changes in Fibromyalgia Suggest the Role of the Immune-Inflammatory Response and Central Sensitization. Journal of clinical medicine, 10(21), 4992. https://doi.org/10.3390/jcm10214992

Gerra, M. C., Carnevali, D., Pedersen, I. S., Donnini, C., Manfredini, M., González-Villar, A., Triñanes, Y., Pidal-Miranda, M., Arendt-Nielsen, L., & Carrillo-de-la-Peña, M. T. (2020). DNA methylation changes in genes involved in inflammation and depression in fibromyalgia: a pilot study. Scandinavian journal of pain, 21(2), 372–383. https://doi.org/10.1515/sjpain-2020-0124

Gogos, J. A., Morgan, M., Luine, V., Santha, M., Ogawa, S., Pfaff, D., & Karayiorgou, M. (1998). Catechol-O-methyltransferase-deficient mice exhibit sexually dimorphic changes in catecholamine levels and behavior. Proceedings of the National Academy of Sciences of the United States of America, 95(17), 9991–9996. https://doi.org/10.1073/pnas.95.17.9991

Graeff F. G. (1997). Serotonergic systems. The Psychiatric clinics of North America, 20(4), 723–739. https://doi.org/10.1016/s0193-953x(05)70342-7

Gursoy S. (2002). Absence of association of the serotonin transporter gene polymorphism with the mentally healthy subset of fibromyalgia patients. Clinical rheumatology, 21(3), 194–197. https://doi.org/10.1007/s10067-002-8284-5

Gürsoy, S., Erdal, E., Herken, H., Madenci, E., & Alaşehirli, B. (2001). Association of T102C polymorphism of the 5-HT2A receptor gene with psychiatric status in fibromyalgia syndrome. Rheumatology international, 21(2), 58–61. https://doi.org/10.1007/s002960100130

Gürsoy, S., Erdal, E., Herken, H., Madenci, E., Alaşehirli, B., & Erdal, N. (2003). Significance of catechol-O-methyltransferase gene polymorphism in fibromyalgia syndrome. Rheumatology international, 23(3), 104–107. https://doi.org/10.1007/s00296-002-0260-5

Gürsoy, S., Erdal, E., Sezgin, M., Barlas, I. O., Aydeniz, A., Alaşehirli, B., & Sahin, G. (2008). Which genotype of MAO gene that the patients have are likely to be most susceptible to the symptoms of fibromyalgia?. Rheumatology international, 28(4), 307–311. https://doi.org/10.1007/s00296-007-0454-y

Hannon, J., & Hoyer, D. (2008). Molecular biology of 5-HT receptors. Behavioural brain research, 195(1), 198–213. https://doi.org/10.1016/j.bbr.2008.03.020

Hanson, I. M., Seawright, A., & van Heyningen, V. (1992). The human BDNF gene maps between FSHB and HVBS1 at the boundary of 11p13-p14. Genomics, 13(4), 1331–1333. https://doi.org/10.1016/0888-7543(92)90060-6

Hariri, A. R., Mattay, V. S., Tessitore, A., Kolachana, B., Fera, F., Goldman, D., Egan, M. F., & Weinberger, D. R. (2002). Serotonin transporter genetic variation and the response of the human amygdala. Science (New York, N.Y.), 297(5580), 400–403. https://doi.org/10.1126/science.1071829

Harris, R. E., & Clauw, D. J. (2006). How do we know that the pain in fibromyalgia is "real"?. Current pain and headache reports, 10(6), 403–407. https://doi.org/10.1007/s11916-006-0069-0

Harris, R. E., Clauw, D. J., Scott, D. J., McLean, S. A., Gracely, R. H., & Zubieta, J. K. (2007). Decreased central mu-opioid receptor availability in fibromyalgia. The Journal of neuroscience : the official journal of the Society for Neuroscience, 27(37), 10000–10006. https://doi.org/10.1523/JNEUROSCI.2849-07.2007

Hatami, M., Sobhan, M. R., Dastgheib, S. A., Jarahzadeh, M. H., Jafari, M., Yadegari, A., Sadeghizadeh-Yazdi, J., & Neamatzadeh, H. (2020). Association of catechol-O-methyltranferase 472G>A (Val158Met) polymorphism with susceptibility to fibromyalgia syndrome. Journal of orthopaedics, 20, 257–260. https://doi.org/10.1016/j.jor.2020.01.013

Heddini, U., Bohm-Starke, N., Grönbladh, A., Nyberg, F., Nilsson, K. W., & Johannesson, U. (2014). Serotonin receptor gene (5HT-2A) polymorphism is associated with provoked vestibulodynia and comorbid symptoms of pain. The journal of sexual medicine, 11(12), 3064–3071. https://doi.org/10.1111/jsm.12685

Heils, A., Teufel, A., Petri, S., Stöber, G., Riederer, P., Bengel, D., & Lesch, K. P. (1996). Allelic variation of human serotonin transporter gene expression. Journal of neurochemistry, 66(6), 2621–2624. https://doi.org/10.1046/j.1471-4159.1996.66062621.x

Hocking, L. J., Smith, B. H., Jones, G. T., Reid, D. M., Strachan, D. P., & Macfarlane, G. J. (2010). Genetic variation in the beta2-adrenergic receptor but not catecholamine-O-methyltransferase predisposes to chronic pain: results from the 1958 British Birth Cohort Study. Pain, 149(1), 143–151. https://doi.org/10.1016/j.pain.2010.01.023

Hoehe, M. R., Berrettini, W. H., Schwinn, D. A., & Hsieh, W. T. (1992). A two-allele PstI RFLP for the alpha-1C adrenergic receptor gene (ADRA1C). Human molecular genetics, 1(5), 349. https://doi.org/10.1093/hmg/1.5.349-a

Inanir, A., Karakus, N., Ates, O., Sezer, S., Bozkurt, N., Inanir, S., & Yigit, S. (2014). Clinical symptoms in fibromyalgia are associated to catechol-O-methyltransferase (COMT) gene Val158Met polymorphism. Xenobiotica; the fate of foreign compounds in biological systems, 44(10), 952–956. https://doi.org/10.3109/00498254.2014.913083

Inanir, A., Yigit, S., Tekcan, A., Pinarli, F. A., Inanir, S., & Karakus, N. (2015). Angiotensin converting enzyme and methylenetetrahydrofolate reductase gene variations in fibromyalgia syndrome. Gene, 564(2), 188–192. https://doi.org/10.1016/j.gene.2015.03.051

Inoue, A., Yanagisawa, M., Takuwa, Y., Mitsui, Y., Kobayashi, M., & Masaki, T. (1989). The human preproendothelin-1 gene. Complete nucleotide sequence and regulation of expression. The Journal of biological chemistry, 264(25), 14954–14959.

Josep García-Fructuoso, F., Ignacio Lao-Villadóniga, J., Beyer, K., & Santos, C. (2006). Relación entre genotipos del gen catecol O-metiltransferasa y la gravedad de la fibromialgia [Relationship between COMT gene genotypes and severity of fibromyalgia]. Reumatologia clinica, 2(4), 168–172. https://doi.org/10.1016/S1699-258X(06)73042-X

Karakus, N., Yigit, S., Inanir, A., Inanir, S., Toprak, H., & Okan, S. (2012). Association between sequence variations of the Mediterranean fever gene and fibromyalgia syndrome in a cohort of Turkish patients. Clinica chimica acta; international journal of clinical chemistry, 414, 36–40. https://doi.org/10.1016/j.cca.2012.07.019

Karoly, P., & Ruehlman, L. S. (2006). Psychological "resilience" and its correlates in chronic pain: findings from a national community sample. Pain, 123(1-2), 90–97. https://doi.org/10.1016/j.pain.2006.02.014

Kato, K., Sullivan, P. F., Evengård, B., & Pedersen, N. L. (2009). A population-based twin study of functional somatic syndromes. Psychological medicine, 39(3), 497–505. https://doi.org/10.1017/S0033291708003784

Khalil, R., Al-Awaida, W. J., Al-Ameer, H. J., Jarrar, Y., Imraish, A., Al Bawareed, O., Qawadri, R., Al Madhoon, F., & Obeidat, L. (2020). Investigation of ACE rs4646994, MTHFR rs1801133 and VDR rs2228570 Genotypes in Jordanian Patients with Fibromyalgia Syndrome. Endocrine, metabolic & immune disorders drug targets, 10.2174/1871530321666201223104622. Advance online publication. https://doi.org/10.2174/1871530321666201223104622

Kilpatrick, L. A., Labus, J. S., Coveleskie, K., Hammer, C., Rappold, G., Tillisch, K., Bueller, J. A., Suyenobu, B., Jarcho, J. M., McRoberts, J. A., Niesler, B., & Mayer, E. A. (2011). The HTR3A polymorphism c. -42C>T is associated with amygdala responsiveness in patients with irritable bowel syndrome. Gastroenterology, 140(7), 1943–1951. https://doi.org/10.1053/j.gastro.2011.03.011

Kim, S. K., Kim, S. H., Nah, S. S., Lee, J. H., Hong, S. J., Kim, H. S., Lee, H. S., Kim, H. A., Joung, C. I., Bae, J., Choe, J. Y., & Lee, S. S. (2013). Association of guanosine triphosphate cyclohydrolase 1 gene polymorphisms with fibromyalgia syndrome in a Korean population. The Journal of rheumatology, 40(3), 316–322. https://doi.org/10.3899/jrheum.120929

Klengel, T., Pape, J., Binder, E. B., & Mehta, D. (2014). The role of DNA methylation in stress-related psychiatric disorders. Neuropharmacology, 80, 115–132. https://doi.org/10.1016/j.neuropharm.2014.01.013

Kling, A., Seddighzadeh, M., Arlestig, L., Alfredsson, L., Rantapää-Dahlqvist, S., & Padyukov, L. (2008). Genetic variations in the serotonin 5-HT2A receptor gene (HTR2A) are associated with rheumatoid arthritis. Annals of the rheumatic diseases, 67(8), 1111–1115. https://doi.org/10.1136/ard.2007.074948

Kosek, E., Martinsen, S., Gerdle, B., Mannerkorpi, K., Löfgren, M., Bileviciute-Ljungar, I., Fransson, P., Schalling, M., Ingvar, M., Ernberg, M., & Jensen, K. B. (2016). The translocator protein gene is associated with symptom severity and cerebral pain processing in fibromyalgia. Brain, behavior, and immunity, 58, 218–227. https://doi.org/10.1016/j.bbi.2016.07.150

Krzywkowski, K., Davies, P. A., Feinberg-Zadek, P. L., Bräuner-Osborne, H., & Jensen, A. A. (2008). High-frequency HTR3B variant associated with major depression dramatically augments the signaling of the human 5-HT3AB receptor. Proceedings of the National Academy of Sciences of the United States of America, 105(2), 722–727. https://doi.org/10.1073/pnas.0708454105

Kuryliszyn-Moskal, A., Klimiuk, P. A., Sierakowski, S., & Ciolkiewicz, M. (2006). A study on vascular endothelial growth factor and endothelin-1 in patients with extra-articular involvement of rheumatoid arthritis. Clinical rheumatology, 25(3), 314–319. https://doi.org/10.1007/s10067-005-0007-2

Lachaine, J., Beauchemin, C., & Landry, P. A. (2010). Clinical and economic characteristics of patients with fibromyalgia syndrome. The Clinical journal of pain, 26(4), 284–290. https://doi.org/10.1097/AJP.0b013e3181cf599f

Lang, U. E., Hellweg, R., Kalus, P., Bajbouj, M., Lenzen, K. P., Sander, T., Kunz, D., & Gallinat, J. (2005). Association of a functional BDNF polymorphism and anxiety-related personality traits. Psychopharmacology, 180(1), 95–99. https://doi.org/10.1007/s00213-004-2137-7

Ledermann, K., Hasler, G., Jenewein, J., Sprott, H., Schnyder, U., & Martin-Soelch, C. (2020). 5'UTR polymorphism in the serotonergic receptor HTR3A gene is differently associated with striatal Dopamine D2/D3 receptor availability in the right putamen in Fibromyalgia patients and healthy controls-Preliminary evidence. Synapse (New York, N.Y.), 74(5), e22147. https://doi.org/10.1002/syn.22147

Lee, C., Liptan, G., Kantorovich, S., Sharma, M., & Brenton, A. (2018). Association of Catechol-O-methyltransferase single nucleotide polymorphisms, ethnicity, and sex in a large cohort of fibromyalgia patients. BMC rheumatology, 2, 38. https://doi.org/10.1186/s41927-018-0045-4

Lee, Y. H., Choi, S. J., Ji, J. D., & Song, G. G. (2012). Candidate gene studies of fibromyalgia: a systematic review and meta-analysis. Rheumatology international, 32(2), 417–426. https://doi.org/10.1007/s00296-010-1678-9

Lesch, K. P., Bengel, D., Heils, A., Sabol, S. Z., Greenberg, B. D., Petri, S., Benjamin, J., Müller, C. R., Hamer, D. H., & Murphy, D. L. (1996). Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science (New York, N.Y.), 274(5292), 1527–1531. https://doi.org/10.1126/science.274.5292.1527

Levine, J. D., & Alessandri-Haber, N. (2007). TRP channels: targets for the relief of pain. Biochimica et biophysica acta, 1772(8), 989–1003. https://doi.org/10.1016/j.bbadis.2007.01.008

Lloyd, D. M., Wittkopf, P. G., Arendsen, L. J., & Jones, A. (2020). Is Transcranial Direct Current Stimulation (tDCS) Effective for the Treatment of Pain in Fibromyalgia? A Systematic Review and Meta-Analysis. The journal of pain, 21(11-12), 1085–1100. https://doi.org/10.1016/j.jpain.2020.01.003

Low, L. A., & Schweinhardt, P. (2012). Early life adversity as a risk factor for fibromyalgia in later life. Pain research and treatment, 2012, 140832. https://doi.org/10.1155/2012/140832

Lusis, A. J., Heinzmann, C., Sparkes, R. S., Scott, J., Knott, T. J., Geller, R., Sparkes, M. C., & Mohandas, T. (1986). Regional mapping of human chromosome 19: organization of genes for plasma lipid transport (APOC1, -C2, and -E and LDLR) and the genes C3, PEPD, and GPI. Proceedings of the National Academy of Sciences of the United States of America, 83(11), 3929–3933. https://doi.org/10.1073/pnas.83.11.3929

Macedo, J. A., Hesse, J., Turner, J. D., Meyer, J., Hellhammer, D. H., & Muller, C. P. (2008). Glucocorticoid sensitivity in fibromyalgia patients: decreased expression of corticosteroid receptors and glucocorticoid-induced leucine zipper. Psychoneuroendocrinology, 33(6), 799–809. https://doi.org/10.1016/j.psyneuen.2008.03.012

Macfarlane, G. J., Kronisch, C., Dean, L. E., Atzeni, F., Häuser, W., Fluß, E., Choy, E., Kosek, E., Amris, K., Branco, J., Dincer, F., Leino-Arjas, P., Longley, K., McCarthy, G. M., Makri, S., Perrot, S., Sarzi-Puttini, P., Taylor, A., & Jones, G. T. (2017). EULAR revised recommendations for the management of fibromyalgia. Annals of the rheumatic diseases, 76(2), 318–328. https://doi.org/10.1136/annrheumdis-2016-209724

Maricq, A. V., Peterson, A. S., Brake, A. J., Myers, R. M., & Julius, D. (1991). Primary structure and functional expression of the 5HT3 receptor, a serotonin-gated ion channel. Science (New York, N.Y.), 254(5030), 432–437. https://doi.org/10.1126/science.1718042

Markkula, R., Järvinen, P., Leino-Arjas, P., Koskenvuo, M., Kalso, E., & Kaprio, J. (2009). Clustering of symptoms associated with fibromyalgia in a Finnish Twin Cohort. European journal of pain (London, England), 13(7), 744–750. https://doi.org/10.1016/j.ejpain.2008.09.007

Marques, A. P., Santos, A. M. B., Assumpção, A., Matsutani, L. A., Lage, L. V. & Pereira, C. A. B. (2006). Validation of the brazilian version of the fibromyalgia impact questionnaire (FIQ). Revista brasileira de reumatologia, 46(1), 24–31. https://doi.org/10.1590/S0482-50042006000100006

Marques, A. P., Santo, A., Berssaneti, A. A., Matsutani, L. A., & Yuan, S. (2017). Prevalence of fibromyalgia: literature review update. Revista brasileira de reumatologia, 57(4), 356–363. https://doi.org/10.1016/j.rbre.2017.01.005

Martínez-Jauand, M., Sitges, C., Rodríguez, V., Picornell, A., Ramon, M., Buskila, D., & Montoya, P. (2013). Pain sensitivity in fibromyalgia is associated with catechol-O-methyltransferase (COMT) gene. European journal of pain (London, England), 17(1), 16–27. https://doi.org/10.1002/j.1532-2149.2012.00153.x

Masters, S. L., Lagou, V., Jéru, I., Baker, P. J., Van Eyck, L., Parry, D. A., Lawless, D., De Nardo, D., Garcia-Perez, J. E., Dagley, L. F., Holley, C. L., Dooley, J., Moghaddas, F., Pasciuto, E., Jeandel, P. Y., Sciot, R., Lyras, D., Webb, A. I., Nicholson, S. E., De Somer, L., … Liston, A. (2016). Familial autoinflammation with neutrophilic dermatosis reveals a regulatory mechanism of pyrin activation. Science translational medicine, 8(332), 332ra45. https://doi.org/10.1126/scitranslmed.aaf1471

Matsuda, J. B., Barbosa, F. R., Morel, L. J., França, S., Zingaretti, S. M., da Silva, L. M., Pereira, A. M., Marins, M., & Fachin, A. L. (2010). Serotonin receptor (5-HT 2A) and catechol-O-methyltransferase (COMT) gene polymorphisms: triggers of fibromyalgia?. Revista brasileira de reumatologia, 50(2), 141–149.

McAllister, S. J., Vincent, A., Hassett, A. L., Whipple, M. O., Oh, T. H., Benzo, R. P., & Toussaint, L. L. (2015). Psychological Resilience, Affective Mechanisms and Symptom Burden in a Tertiary-care Sample of Patients with Fibromyalgia. Stress and health : journal of the International Society for the Investigation of Stress, 31(4), 299–305. https://doi.org/10.1002/smi.2555

Meeker, T. J., Jupudi, R., Lenz, F. A., & Greenspan, J. D. (2020). New Developments in Non-invasive Brain Stimulation in Chronic Pain. Current physical medicine and rehabilitation reports, 8(3), 280–292. https://doi.org/10.1007/s40141-020-00260-w

Menzies, V., Lyon, D. E., Archer, K. J., Zhou, Q., Brumelle, J., Jones, K. H., Gao, G., York, T. P., & Jackson-Cook, C. (2013). Epigenetic alterations and an increased frequency of micronuclei in women with fibromyalgia. Nursing research and practice, 2013, 795784. https://doi.org/10.1155/2013/795784

Michiels, J. J., te Morsche, R. H., Jansen, J. B., & Drenth, J. P. (2005). Autosomal dominant erythermalgia associated with a novel mutation in the voltage-gated sodium channel alpha subunit Nav1.7. Archives of neurology, 62(10), 1587–1590. https://doi.org/10.1001/archneur.62.10.1587

Miyake, A., Mochizuki, S., Takemoto, Y., & Akuzawa, S. (1995). Molecular cloning of human 5-hydroxytryptamine3 receptor: heterogeneity in distribution and function among species. Molecular pharmacology, 48(3), 407–416.

Mogil J. S. (2012). Pain genetics: past, present and future. Trends in genetics : TIG, 28(6), 258–266. https://doi.org/10.1016/j.tig.2012.02.004

Moriam, S., & Sobhani, M. E. (2013). Epigenetic effect of chronic stress on dopamine signaling and depression. Genetics & epigenetics, 5, 11–16. https://doi.org/10.4137/GEG.S11016

Mortensen, E. L., & Høgh, P. (2001). A gender difference in the association between APOE genotype and age-related cognitive decline. Neurology, 57(1), 89–95. https://doi.org/10.1212/wnl.57.1.89

Nackley, A. G., Tan, K. S., Fecho, K., Flood, P., Diatchenko, L., & Maixner, W. (2007). Catechol-O-methyltransferase inhibition increases pain sensitivity through activation of both beta2- and beta3-adrenergic receptors. Pain, 128(3), 199–208. https://doi.org/10.1016/j.pain.2006.09.022

Nah, S. S., Lee, H., Hong, Y., Im, J., Won, H., Chang, S. H., Kim, H. K., Kwon, J. T., & Kim, H. J. (2017). Association between endothelin‑1 and fibromyalgia syndrome. Molecular medicine reports, 16(5), 6234–6239. https://doi.org/10.3892/mmr.2017.7395

Najera, K., Fagan, B. M., & Thompson, P. M. (2019). SNAP-25 in Major Psychiatric Disorders: A Review. Neuroscience, 420, 79–85. https://doi.org/10.1016/j.neuroscience.2019.02.008

Nicholl, B. I., Holliday, K. L., Macfarlane, G. J., Thomson, W., Davies, K. A., O'Neill, T. W., Bartfai, G., Boonen, S., Casanueva, F., Finn, J. D., Forti, G., Giwercman, A., Huhtaniemi, I. T., Kula, K., Punab, M., Silman, A. J., Vanderschueren, D., Wu, F. C., McBeth, J., & European Male Ageing Study Group (2010). No evidence for a role of the catechol-O-methyltransferase pain sensitivity haplotypes in chronic widespread pain. Annals of the rheumatic diseases, 69(11), 2009–2012. https://doi.org/10.1136/ard.2009.126086

Nugraha, B., Anwar, S. L., Gutenbrunner, C., & Korallus, C. (2020). Polymorphisms of brain-derived neurotrophic factor genes are associated with anxiety and body mass index in fibromyalgia syndrome patients. BMC research notes, 13(1), 402. https://doi.org/10.1186/s13104-020-05226-8

Offenbaecher, M., Bondy, B., de Jonge, S., Glatzeder, K., Krüger, M., Schoeps, P., & Ackenheil, M. (1999). Possible association of fibromyalgia with a polymorphism in the serotonin transporter gene regulatory region. Arthritis and rheumatism, 42(11), 2482–2488. https://doi.org/10.1002/1529-0131(199911)42:11<2482::AID-ANR27>3.0.CO;2-B

Pache, M., Schwarz, H. A., Kaiser, H. J., Wüest, P., Klöti, M., Dubler, B., & Flammer, J. (2002). Elevated plasma endothelin-1 levels and vascular dysregulation in patients with rheumatoid arthritis. Medical science monitor : international medical journal of experimental and clinical research, 8(9), CR616–CR619.

Pages, J. C., Drieu, C., Blanché, H., Beckmann, J., & Cann, H. M. (1993). A short tandem repeat polymorphism at the endothelin 1 (EDN1) locus. Human molecular genetics, 2(1), 90. https://doi.org/10.1093/hmg/2.1.90

Paiva, E. S., Heymann, R. E., Rezende, M. C., Helfenstein, M., Jr, Martinez, J. E., Provenza, J. R., Ranzolin, A., de Assis, M. R., Pasqualin, V. D., & Bennett, R. M. (2013). A Brazilian Portuguese version of the Revised Fibromyalgia Impact Questionnaire (FIQR): a validation study. Clinical rheumatology, 32(8), 1199–1206. https://doi.org/10.1007/s10067-013-2259-6

Park, D. J., Kang, J. H., Yim, Y. R., Kim, J. E., Lee, J. W., Lee, K. E., Wen, L., Kim, T. J., Park, Y. W., & Lee, S. S. (2015). Exploring Genetic Susceptibility to Fibromyalgia. Chonnam medical journal, 51(2), 58–65. https://doi.org/10.4068/cmj.2015.51.2.58

Park, D. J., Kim, S. H., Nah, S. S., Lee, J. H., Kim, S. K., Lee, Y. A., Hong, S. J., Kim, H. S., Lee, H. S., Kim, H. A., Joung, C. I., Kim, S. H., & Lee, S. S. (2016a). Association between catechol-O-methyl transferase gene polymorphisms and fibromyalgia in a Korean population: A case-control study. European journal of pain (London, England), 20(7), 1131–1139. https://doi.org/10.1002/ejp.837

Park, D. J., Kim, S. H., Nah, S. S., Lee, J. H., Kim, S. K., Lee, Y. A., Hong, S. J., Kim, H. S., Lee, H. S., Kim, H. A., Joung, C. I., Kim, S. H., & Lee, S. S. (2016b). Polymorphisms of the TRPV2 and TRPV3 genes associated with fibromyalgia in a Korean population. Rheumatology (Oxford, England), 55(8), 1518–1527. https://doi.org/10.1093/rheumatology/kew180

Park, D. J., Kim, S. H., Nah, S. S., Lee, J. H., Kim, S. K., Lee, Y. A., Hong, S. J., Kim, H. S., Lee, H. S., Kim, H. A., Joung, C. I., Kim, S. H., & Lee, S. S. (2018). Association between brain-derived neurotrophic factor gene polymorphisms and fibromyalgia in a Korean population: a multicenter study. Arthritis research & therapy, 20(1), 220. https://doi.org/10.1186/s13075-018-1726-5

Park, H., & Poo, M. M. (2013). Neurotrophin regulation of neural circuit development and function. Nature reviews. Neuroscience, 14(1), 7–23. https://doi.org/10.1038/nrn3379

Parsons, M. J., D'Souza, U. M., Arranz, M. J., Kerwin, R. W., & Makoff, A. J. (2004). The -1438A/G polymorphism in the 5-hydroxytryptamine type 2A receptor gene affects promoter activity. Biological psychiatry, 56(6), 406–410. https://doi.org/10.1016/j.biopsych.2004.06.020

Pearson-Fuhrhop, K. M., Dunn, E. C., Mortero, S., Devan, W. J., Falcone, G. J., Lee, P., Holmes, A. J., Hollinshead, M. O., Roffman, J. L., Smoller, J. W., Rosand, J., & Cramer, S. C. (2014). Dopamine genetic risk score predicts depressive symptoms in healthy adults and adults with depression. PloS one, 9(5), e93772. https://doi.org/10.1371/journal.pone.0093772

Pereira, M.G. (2012). Artigos científicos: como redigir, publicar e avaliar. Rio de Janeiro: Guanabara Koogan. p.47-48.

Pertovaara A. (2006). Noradrenergic pain modulation. Progress in neurobiology, 80(2), 53–83. https://doi.org/10.1016/j.pneurobio.2006.08.001

Polli, A., Ghosh, M., Bakusic, J., Ickmans, K., Monteyne, D., Velkeniers, B., Bekaert, B., Godderis, L., & Nijs, J. (2020). DNA Methylation and Brain-Derived Neurotrophic Factor Expression Account for Symptoms and Widespread Hyperalgesia in Patients With Chronic Fatigue Syndrome and Comorbid Fibromyalgia. Arthritis & rheumatology (Hoboken, N.J.), 72(11), 1936–1944. https://doi.org/10.1002/art.41405

Potvin, S., Larouche, A., Normand, E., de Souza, J. B., Gaumond, I., Grignon, S., & Marchand, S. (2009). DRD3 Ser9Gly polymorphism is related to thermal pain perception and modulation in chronic widespread pain patients and healthy controls. The journal of pain, 10(9), 969–975. https://doi.org/10.1016/j.jpain.2009.03.013

Pozzi, D., Corradini, I., & Matteoli, M. (2019). The Control of Neuronal Calcium Homeostasis by SNAP-25 and its Impact on Neurotransmitter Release. Neuroscience, 420, 72–78. https://doi.org/10.1016/j.neuroscience.2018.11.009

Queiroz L. P. (2013). Worldwide epidemiology of fibromyalgia. Current pain and headache reports, 17(8), 356. https://doi.org/10.1007/s11916-013-0356-5

Ramamoorthy, S., Bauman, A. L., Moore, K. R., Han, H., Yang-Feng, T., Chang, A. S., Ganapathy, V., & Blakely, R. D. (1993). Antidepressant- and cocaine-sensitive human serotonin transporter: molecular cloning, expression, and chromosomal localization. Proceedings of the National Academy of Sciences of the United States of America, 90(6), 2542–2546. https://doi.org/10.1073/pnas.90.6.2542

Reeser, J. C., Payne, E., Kitchner, T., & McCarty, C. A. (2011). Apolipoprotein e4 genotype increases the risk of being diagnosed with posttraumatic fibromyalgia. PM & R : the journal of injury, function, and rehabilitation, 3(3), 193–197. https://doi.org/10.1016/j.pmrj.2010.12.009

Reichborn-Kjennerud, T., Stoltenberg, C., Tambs, K., Roysamb, E., Kringlen, E., Torgersen, S., & Harris, J. R. (2002). Back-neck pain and symptoms of anxiety and depression: a population-based twin study. Psychological medicine, 32(6), 1009–1020. https://doi.org/10.1017/s0033291702005950

Reimann, F., Cox, J. J., Belfer, I., Diatchenko, L., Zaykin, D. V., McHale, D. P., Drenth, J. P., Dai, F., Wheeler, J., Sanders, F., Wood, L., Wu, T. X., Karppinen, J., Nikolajsen, L., Männikkö, M., Max, M. B., Kiselycznyk, C., Poddar, M., Te Morsche, R. H., Smith, S., … Woods, C. G. (2010). Pain perception is altered by a nucleotide polymorphism in SCN9A. Proceedings of the National Academy of Sciences of the United States of America, 107(11), 5148–5153. https://doi.org/10.1073/pnas.0913181107

Russell, I. J., Michalek, J. E., Vipraio, G. A., Fletcher, E. M., Javors, M. A., & Bowden, C. A. (1992a). Platelet 3H-imipramine uptake receptor density and serum serotonin levels in patients with fibromyalgia/fibrositis syndrome. The Journal of rheumatology, 19(1), 104–109.

Russell, I. J., Vaeroy, H., Javors, M., & Nyberg, F. (1992b). Cerebrospinal fluid biogenic amine metabolites in fibromyalgia/fibrositis syndrome and rheumatoid arthritis. Arthritis and rheumatism, 35(5), 550–556. https://doi.org/10.1002/art.1780350509

Sarzi-Puttini, P., Giorgi, V., Marotto, D., & Atzeni, F. (2020). Fibromyalgia: an update on clinical characteristics, aetiopathogenesis and treatment. Nature reviews. Rheumatology, 16(11), 645–660. https://doi.org/10.1038/s41584-020-00506-w

Schmechel, D. E., & Edwards, C. L. (2012). Fibromyalgia, mood disorders, and intense creative energy: A1AT polymorphisms are not always silent. Neurotoxicology, 33(6), 1454–1472. https://doi.org/10.1016/j.neuro.2012.03.001

Schwinn, D. A., Lomasney, J. W., Lorenz, W., Szklut, P. J., Fremeau, R. T., Jr, Yang-Feng, T. L., Caron, M. G., Lefkowitz, R. J., & Cotecchia, S. (1990). Molecular cloning and expression of the cDNA for a novel alpha 1-adrenergic receptor subtype. The Journal of biological chemistry, 265(14), 8183–8189.

Scott, I. C., Halila, R., Jenkins, J. M., Mehan, S., Apostolou, S., Winqvist, R., Callen, D. F., Prockop, D. J., Peltonen, L., & Kadler, K. E. (1996). Molecular cloning, expression and chromosomal localization of a human gene encoding a 33 kDa putative metallopeptidase (PRSM1). Gene, 174(1), 135–143. https://doi.org/10.1016/0378-1119(96)00510-0

Scudds, R. A., Li EK-M., Scudds R. J. (2006). The prevalence of fibromyalgia syndrome in Chinese people in Hong Kong. J Musculoskelet Pain, 14:3-11. https://doi.org/10.1300/J094v14n02_02

Senna, E. R., De Barros, A. L., Silva, E. O., Costa, I. F., Pereira, L. V., Ciconelli, R. M., & Ferraz, M. B. (2004). Prevalence of rheumatic diseases in Brazil: a study using the COPCORD approach. The Journal of rheumatology, 31(3), 594–597.

Shen, S., Battersby, S., Weaver, M., Clark, E., Stephens, K., & Harmar, A. J. (2000). Refined mapping of the human serotonin transporter (SLC6A4) gene within 17q11 adjacent to the CPD and NF1 genes. European journal of human genetics : EJHG, 8(1), 75–78. https://doi.org/10.1038/sj.ejhg.5200400

Shimizu, E., Hashimoto, K., Okamura, N., Koike, K., Komatsu, N., Kumakiri, C., Nakazato, M., Watanabe, H., Shinoda, N., Okada, S., & Iyo, M. (2003). Alterations of serum levels of brain-derived neurotrophic factor (BDNF) in depressed patients with or without antidepressants. Biological psychiatry, 54(1), 70–75. https://doi.org/10.1016/s0006-3223(03)00181-1

Sluka, K. A., & Clauw, D. J. (2016). Neurobiology of fibromyalgia and chronic widespread pain. Neuroscience, 338, 114–129. https://doi.org/10.1016/j.neuroscience.2016.06.006

Small, K. M., McGraw, D. W., & Liggett, S. B. (2003). Pharmacology and physiology of human adrenergic receptor polymorphisms. Annual review of pharmacology and toxicology, 43, 381–411. https://doi.org/10.1146/annurev.pharmtox.43.100901.135823

Smith, S. B., Maixner, D. W., Fillingim, R. B., Slade, G., Gracely, R. H., Ambrose, K., Zaykin, D. V., Hyde, C., John, S., Tan, K., Maixner, W., & Diatchenko, L. (2012). Large candidate gene association study reveals genetic risk factors and therapeutic targets for fibromyalgia. Arthritis and rheumatism, 64(2), 584–593. https://doi.org/10.1002/art.33338

Spurlock, G., Heils, A., Holmans, P., Williams, J., D'Souza, U. M., Cardno, A., Murphy, K. C., Jones, L., Buckland, P. R., McGuffin, P., Lesch, K. P., & Owen, M. J. (1998). A family based association study of T102C polymorphism in 5HT2A and schizophrenia plus identification of new polymorphisms in the promoter. Molecular psychiatry, 3(1), 42–49. https://doi.org/10.1038/sj.mp.4000342

Stein, M. B., Campbell-Sills, L., & Gelernter, J. (2009). Genetic variation in 5HTTLPR is associated with emotional resilience. American journal of medical genetics. Part B, Neuropsychiatric genetics : the official publication of the International Society of Psychiatric Genetics, 150B(7), 900–906. https://doi.org/10.1002/ajmg.b.30916

Sui, B. D., Xu, T. Q., Liu, J. W., Wei, W., Zheng, C. X., Guo, B. L., Wang, Y. Y., & Yang, Y. L. (2013). Understanding the role of mitochondria in the pathogenesis of chronic pain. Postgraduate medical journal, 89(1058), 709–714. https://doi.org/10.1136/postgradmedj-2012-131068

Sullivan, M. J., Adams, H., Rhodenizer, T., & Stanish, W. D. (2006). A psychosocial risk factor--targeted intervention for the prevention of chronic pain and disability following whiplash injury. Physical therapy, 86(1), 8–18. https://doi.org/10.1093/ptj/86.1.8

Sullivan, M. J., Thorn, B., Haythornthwaite, J. A., Keefe, F., Martin, M., Bradley, L. A., & Lefebvre, J. C. (2001). Theoretical perspectives on the relation between catastrophizing and pain. The Clinical journal of pain, 17(1), 52–64. https://doi.org/10.1097/00002508-200103000-00008

Szyf, M., & Bick, J. (2013). DNA methylation: a mechanism for embedding early life experiences in the genome. Child development, 84(1), 49–57. https://doi.org/10.1111/j.1467-8624.2012.01793.x

Tammimäki, A., & Männistö, P. T. (2012). Catechol-O-methyltransferase gene polymorphism and chronic human pain: a systematic review and meta-analysis. Pharmacogenetics and genomics, 22(9), 673–691. https://doi.org/10.1097/FPC.0b013e3283560c46

Tander, B., Gunes, S., Boke, O., Alayli, G., Kara, N., Bagci, H., & Canturk, F. (2008). Polymorphisms of the serotonin-2A receptor and catechol-O-methyltransferase genes: a study on fibromyalgia susceptibility. Rheumatology international, 28(7), 685–691. https://doi.org/10.1007/s00296-008-0525-8

Tegeder, I., Costigan, M., Griffin, R. S., Abele, A., Belfer, I., Schmidt, H., Ehnert, C., Nejim, J., Marian, C., Scholz, J., Wu, T., Allchorne, A., Diatchenko, L., Binshtok, A. M., Goldman, D., Adolph, J., Sama, S., Atlas, S. J., Carlezon, W. A., Parsegian, A., … Woolf, C. J. (2006). GTP cyclohydrolase and tetrahydrobiopterin regulate pain sensitivity and persistence. Nature medicine, 12(11), 1269–1277. https://doi.org/10.1038/nm1490

Thöny, B., Heizmann, C. W., & Mattei, M. G. (1995). Human GTP-cyclohydrolase I gene and sepiapterin reductase gene map to region 14q21-q22 and 2p14-p12, respectively, by in situ hybridization. Genomics, 26(1), 168–170. https://doi.org/10.1016/0888-7543(95)80101-q

Tour, J., Löfgren, M., Mannerkorpi, K., Gerdle, B., Larsson, A., Palstam, A., Bileviciute-Ljungar, I., Bjersing, J., Martin, I., Ernberg, M., Schalling, M., & Kosek, E. (2017). Gene-to-gene interactions regulate endogenous pain modulation in fibromyalgia patients and healthy controls-antagonistic effects between opioid and serotonin-related genes. Pain, 158(7), 1194–1203. https://doi.org/10.1097/j.pain.0000000000000896

Tsang, H. T., Connell, J. W., Brown, S. E., Thompson, A., Reid, E., & Sanderson, C. M. (2006). A systematic analysis of human CHMP protein interactions: additional MIT domain-containing proteins bind to multiple components of the human ESCRT III complex. Genomics, 88(3), 333–346. https://doi.org/10.1016/j.ygeno.2006.04.003

Tunbridge E. M. (2010). The catechol-O-methyltransferase gene: its regulation and polymorphisms. International review of neurobiology, 95, 7–27. https://doi.org/10.1016/B978-0-12-381326-8.00002-8

Turhanoglu A. D., Yilmaz S., Kaya S., Dursun M., Kararmaz A., Saka G. (2008). The epidemiological aspects of fibromyalgia syndrome in adults living in turkey: a population based study. J Musculoskelet Pain.16:141–7. https://doi.org/10.1080/10582450802161820

Uçeyler, N., Valenza, R., Stock, M., Schedel, R., Sprotte, G., & Sommer, C. (2006). Reduced levels of antiinflammatory cytokines in patients with chronic widespread pain. Arthritis and rheumatism, 54(8), 2656–2664. https://doi.org/10.1002/art.22026

van Tilburg, M., Parisien, M., Boles, R. G., Drury, G. L., Smith-Voudouris, J., Verma, V., Khoury, S., Chabot-Doré, A. J., Nackley, A. G., Smith, S. B., Whitehead, W. E., Zolnoun, D. A., Slade, G. D., Tchivileva, I., Maixner, W., & Diatchenko, L. (2020). A genetic polymorphism that is associated with mitochondrial energy metabolism increases risk of fibromyalgia. Pain, 161(12), 2860–2871. https://doi.org/10.1097/j.pain.0000000000001996

Vargas-Alarcon, G., Alvarez-Leon, E., Fragoso, J. M., Vargas, A., Martinez, A., Vallejo, M., & Martinez-Lavin, M. (2012). A SCN9A gene-encoded dorsal root ganglia sodium channel polymorphism associated with severe fibromyalgia. BMC musculoskeletal disorders, 13, 23. https://doi.org/10.1186/1471-2474-13-23

Vargas-Alarcón, G., Fragoso, J. M., Cruz-Robles, D., Vargas, A., Martinez, A., Lao-Villadóniga, J. I., García-Fructuoso, F., Vallejo, M., & Martínez-Lavín, M. (2009). Association of adrenergic receptor gene polymorphisms with different fibromyalgia syndrome domains. Arthritis and rheumatism, 60(7), 2169–2173. https://doi.org/10.1002/art.24655

Vargas-Alarcón, G., Fragoso, J. M., Cruz-Robles, D., Vargas, A., Vargas, A., Lao-Villadóniga, J. I., García-Fructuoso, F., Ramos-Kuri, M., Hernández, F., Springall, R., Bojalil, R., Vallejo, M., & Martínez-Lavín, M. (2007). Catechol-O-methyltransferase gene haplotypes in Mexican and Spanish patients with fibromyalgia. Arthritis research & therapy, 9(5), R110. https://doi.org/10.1186/ar2316

Walitt, B., Nahin, R. L., Katz, R. S., Bergman, M. J., & Wolfe, F. (2015). The Prevalence and Characteristics of Fibromyalgia in the 2012 National Health Interview Survey. PloS one, 10(9), e0138024. https://doi.org/10.1371/journal.pone.0138024

Wan, L., Li, Y., Zhang, Z., Sun, Z., He, Y., & Li, R. (2018). Methylenetetrahydrofolate reductase and psychiatric diseases. Translational psychiatry, 8(1), 242. https://doi.org/10.1038/s41398-018-0276-6

Wang, H., Moser, M., Schiltenwolf, M., & Buchner, M. (2008). Circulating cytokine levels compared to pain in patients with fibromyalgia -- a prospective longitudinal study over 6 months. The Journal of rheumatology, 35(7), 1366–1370.

Weiss, B., Mertz, A., Schröck, E., Koenen, M., & Rappold, G. (1995). Assignment of a human homolog of the mouse Htr3 receptor gene to chromosome 11q23.1-q23.2. Genomics, 29(1), 304–305. https://doi.org/10.1006/geno.1995.1254

Williams, J., Spurlock, G., McGuffin, P., Mallet, J., Nöthen, M. M., Gill, M., Aschauer, H., Nylander, P. O., Macciardi, F., & Owen, M. J. (1996). Association between schizophrenia and T102C polymorphism of the 5-hydroxytryptamine type 2a-receptor gene. European Multicentre Association Study of Schizophrenia (EMASS) Group. Lancet (London, England), 347(9011), 1294–1296. https://doi.org/10.1016/s0140-6736(96)90939-3

Wolfe, F., Clauw, D. J., Fitzcharles, M. A., Goldenberg, D. L., Häuser, W., Katz, R. L., Mease, P. J., Russell, A. S., Russell, I. J., & Walitt, B. (2016). 2016 Revisions to the 2010/2011 fibromyalgia diagnostic criteria. Seminars in arthritis and rheumatism, 46(3), 319–329. https://doi.org/10.1016/j.semarthrit.2016.08.012

Wolfe, F., Clauw, D. J., Fitzcharles, M. A., Goldenberg, D. L., Häuser, W., Katz, R. S., Mease, P., Russell, A. S., Russell, I. J., & Winfield, J. B. (2011). Fibromyalgia criteria and severity scales for clinical and epidemiological studies: a modification of the ACR Preliminary Diagnostic Criteria for Fibromyalgia. The Journal of rheumatology, 38(6), 1113–1122. https://doi.org/10.3899/jrheum.100594

Wolfe, F., Clauw, D. J., Fitzcharles, M. A., Goldenberg, D. L., Katz, R. S., Mease, P., Russell, A. S., Russell, I. J., Winfield, J. B., & Yunus, M. B. (2010). The American College of Rheumatology preliminary diagnostic criteria for fibromyalgia and measurement of symptom severity. Arthritis care & research, 62(5), 600–610. https://doi.org/10.1002/acr.20140

Wolfe, F., Ross, K., Anderson, J., Russell, I. J., & Hebert, L. (1995). The prevalence and characteristics of fibromyalgia in the general population. Arthritis and rheumatism, 38(1), 19–28. https://doi.org/10.1002/art.1780380104

Wolfe, F., Russell, I. J., Vipraio, G., Ross, K., & Anderson, J. (1997). Serotonin levels, pain threshold, and fibromyalgia symptoms in the general population. The Journal of rheumatology, 24(3), 555–559.

Wolfe, F., Smythe, H. A., Yunus, M. B., Bennett, R. M., Bombardier, C., Goldenberg, D. L., Tugwell, P., Campbell, S. M., Abeles, M., & Clark, P. (1990). The American College of Rheumatology 1990 Criteria for the Classification of Fibromyalgia. Report of the Multicenter Criteria Committee. Arthritis and rheumatism, 33(2), 160–172. https://doi.org/10.1002/art.1780330203

Xiao, Y., He, W., & Russell, I. J. (2011). Genetic polymorphisms of the beta2-adrenergic receptor relate to guanosine protein-coupled stimulator receptor dysfunction in fibromyalgia syndrome. The Journal of rheumatology, 38(6), 1095–1103. https://doi.org/10.3899/jrheum.101104

Xiao, Y., Russell, I. J., & Liu, Y. G. (2012). A brain-derived neurotrophic factor polymorphism Val66Met identifies fibromyalgia syndrome subgroup with higher body mass index and C-reactive protein. Rheumatology international, 32(8), 2479–2485. https://doi.org/10.1007/s00296-011-1990-z

Yang-Feng, T. L., Xue, F. Y., Zhong, W. W., Cotecchia, S., Frielle, T., Caron, M. G., Lefkowitz, R. J., & Francke, U. (1990). Chromosomal organization of adrenergic receptor genes. Proceedings of the National Academy of Sciences of the United States of America, 87(4), 1516–1520. https://doi.org/10.1073/pnas.87.4.1516

Yigit, S., Inanir, A., Tekcan, A., Inanir, S., Tural, S., & Ates, O. (2013). Association between fibromyalgia syndrome and polymorphism of the IL-4 gene in a Turkish population. Gene, 527(1), 62–64. https://doi.org/10.1016/j.gene.2013.04.020

Yunus M. B. (2015). Editorial review: an update on central sensitivity syndromes and the issues of nosology and psychobiology. Current rheumatology reviews, 11(2), 70–85. https://doi.org/10.2174/157339711102150702112236

Zennaro, M. C., Souque, A., Viengchareun, S., Poisson, E., & Lombès, M. (2001). A new human MR splice variant is a ligand-independent transactivator modulating corticosteroid action. Molecular endocrinology (Baltimore, Md.), 15(9), 1586–1598. https://doi.org/10.1210/mend.15.9.0689

Zhao, M., Chen, L., Yang, J., Han, D., Fang, D., Qiu, X., Yang, X., Qiao, Z., Ma, J., Wang, L., Jiang, S., Song, X., Zhou, J., Zhang, J., Chen, M., Qi, D., Yang, Y., & Pan, H. (2018). BDNF Val66Met polymorphism, life stress and depression: A meta-analysis of gene-environment interaction. Journal of affective disorders, 227, 226–235. https://doi.org/10.1016/j.jad.2017.10.024

Żmudzka, E., Sałaciak, K., Sapa, J., & Pytka, K. (2018). Serotonin receptors in depression and anxiety: Insights from animal studies. Life sciences, 210, 106–124. https://doi.org/10.1016/j.lfs.2018.08.050

Zorina-Lichtenwalter, K., Meloto, C. B., Khoury, S., & Diatchenko, L. (2016). Genetic predictors of human chronic pain conditions. Neuroscience, 338, 36–62. https://doi.org/10.1016/j.neuroscience.2016.04.041

Published

14/02/2022

How to Cite

MERINO, A. S.; SIMON, D. Genetic factors associated with fibromyalgia: a narrative review. Research, Society and Development, [S. l.], v. 11, n. 3, p. e11211326421, 2022. DOI: 10.33448/rsd-v11i3.26421. Disponível em: https://www.rsdjournal.org/index.php/rsd/article/view/26421. Acesso em: 26 apr. 2024.

Issue

Section

Health Sciences