• Pain · Mar 2019

    Meta Analysis

    Genome-wide association reveals contribution of MRAS to painful temporomandibular disorder in males.

    • Shad B Smith, Marc Parisien, Eric Bair, Inna Belfer, Anne-Julie Chabot-Doré, Pavel Gris, Samar Khoury, Shannon Tansley, Yelizaveta Torosyan, Dmitri V Zaykin, Olaf Bernhardt, Priscila de Oliveira Serrano, Richard H Gracely, Deepti Jain, Marjo-Riitta Järvelin, Linda M Kaste, Kathleen F Kerr, Thomas Kocher, Raija Lähdesmäki, Nadia Laniado, Cathy C Laurie, Cecelia A Laurie, Minna Männikkö, Carolina B Meloto, Andrea G Nackley, Sarah C Nelson, Paula Pesonen, Margarete C Ribeiro-Dasilva, Celia M Rizzatti-Barbosa, Anne E Sanders, Christian Schwahn, Kirsi Sipilä, Tamar Sofer, Alexander Teumer, Jeffrey S Mogil, Roger B Fillingim, Joel D Greenspan, Richard Ohrbach, Gary D Slade, William Maixner, and Luda Diatchenko.
    • Department of Anesthesiology, Center for Translational Pain Medicine, Duke University, Durham, NC, United States.
    • Pain. 2019 Mar 1; 160 (3): 579-591.

    AbstractPainful temporomandibular disorders (TMDs) are the leading cause of chronic orofacial pain, but its underlying molecular mechanisms remain obscure. Although many environmental factors have been associated with higher risk of developing painful TMD, family and twin studies support a heritable genetic component as well. We performed a genome-wide association study assuming an additive genetic model of TMD in a discovery cohort of 999 cases and 2031 TMD-free controls from the Orofacial Pain: Prospective Evaluation and Risk Assessment (OPPERA) study. Using logistic models adjusted for sex, age, enrollment site, and race, we identified 3 distinct loci that were significant in combined or sex-segregated analyses. A single-nucleotide polymorphism on chromosome 3 (rs13078961) was significantly associated with TMD in males only (odds ratio = 2.9, 95% confidence interval: 2.02-4.27, P = 2.2 × 10). This association was nominally replicated in a meta-analysis of 7 independent orofacial pain cohorts including 160,194 participants (odds ratio = 1.16, 95% confidence interval: 1.0-1.35, P = 2.3 × 10). Functional analysis in human dorsal root ganglia and blood indicated this variant is an expression quantitative trait locus, with the minor allele associated with decreased expression of the nearby muscle RAS oncogene homolog (MRAS) gene (beta = -0.51, P = 2.43 × 10). Male mice, but not female mice, with a null mutation of Mras displayed persistent mechanical allodynia in a model of inflammatory pain. Genetic and behavioral evidence support a novel mechanism by which genetically determined MRAS expression moderates the resiliency to chronic pain. This effect is male-specific and may contribute to the lower rates of painful TMD in men.

      Pubmed     Full text   Copy Citation     Plaintext  

      Add institutional full text...

    Notes

     
    Knowledge, pearl, summary or comment to share?
    300 characters remaining
    help        
    You can also include formatting, links, images and footnotes in your notes
    • Simple formatting can be added to notes, such as *italics*, _underline_ or **bold**.
    • Superscript can be denoted by <sup>text</sup> and subscript <sub>text</sub>.
    • Numbered or bulleted lists can be created using either numbered lines 1. 2. 3., hyphens - or asterisks *.
    • Links can be included with: [my link to pubmed](http://pubmed.com)
    • Images can be included with: ![alt text](https://bestmedicaljournal.com/study_graph.jpg "Image Title Text")
    • For footnotes use [^1](This is a footnote.) inline.
    • Or use an inline reference [^1] to refer to a longer footnote elseweher in the document [^1]: This is a long footnote..

    hide…

Want more great medical articles?

Keep up to date with a free trial of metajournal, personalized for your practice.
1,624,503 articles already indexed!

We guarantee your privacy. Your email address will not be shared.