Archives

  • 2018-07
  • 2018-10
  • 2018-11
  • 2019-04
  • 2019-05
  • 2019-06
  • 2019-07
  • 2019-08
  • 2019-09
  • 2019-10
  • 2019-11
  • 2019-12
  • 2020-01
  • 2020-02
  • 2020-03
  • 2020-04
  • 2020-05
  • 2020-06
  • 2020-07
  • 2020-08
  • 2020-09
  • 2020-10
  • 2020-11
  • 2020-12
  • 2021-01
  • 2021-02
  • 2021-03
  • 2021-04
  • 2021-05
  • 2021-06
  • 2021-07
  • 2021-08
  • 2021-09
  • 2021-10
  • 2021-11
  • 2021-12
  • 2022-01
  • 2022-02
  • 2022-03
  • 2022-04
  • 2022-05
  • 2022-06
  • 2022-07
  • 2022-08
  • 2022-09
  • 2022-10
  • 2022-11
  • 2022-12
  • 2023-01
  • 2023-02
  • 2023-03
  • 2023-04
  • 2023-05
  • 2023-06
  • 2023-08
  • 2023-09
  • 2023-10
  • 2023-11
  • 2023-12
  • 2024-01
  • 2024-02
  • 2024-03
  • 2024-04
  • Using an additive model integrated in

    2018-10-30

    Using an additive model integrated in the CaTS, we had 80% power for the overall dataset (961 cases and 3260 controls), assuming a minor disease allele frequency of 0.20, a genotypic relative risk for this variant of 1.31, a prevalence of neuropathic pain in the diabetic population of 0.25, and the significance level is 10 (Skol et al., 2006). In our previous analysis, our case definition also included evidence of neuropathy, based on recorded results of monofilament testing (Meng et al., 2015). As we did not consider the results of monofilament testing in this study, our case definition was more inclusive and therefore less specific. Although there are power benefits of including more cases, there is also a possibility that neuropathic pain with and without neuropathy evidence might have separate genetic risk markers, as well as shared genetic mechanisms. No studies have been reported examining whether there is any genetic difference between neuropathic pain with and without neuropathy evidence. The peaks we have identified in this paper could reflect some ‘general’ genetic mechanisms of neuropathic pain while the different peaks identified in our previous GWAS may be specifically associated with neuropathic pain with neuropathy evidence (Meng et al., 2015). In other disorders, a nitric oxide booster and its subtypes have been shown to have both shared and different genetic risks (Kessler et al., 2013). Similarly, we did not remove those who were prescribed strong opioid drugs from the control group since opioid drugs are neither indicated first- or second-line treatments, nor commonly used to treat diabetic neuropathic pain (Torrance et al., 2013). A good phenotype, endophenotype and subgroup definition should aim to reflect the underlying genetic mechanisms. There are some recent GWAS published in the field of pain research. A locus between CCT5 and FAM173B located at Chr5p15.2 has been proposed to be associated with chronic widespread pain (Peters et al., 2013). TAOK3 was suggested to be associated with morphine requirement and postoperative pain in a retrospective paediatric day surgery population (Cook-Sather et al., 2014). Rs11127292 in the MYT1L was found to be associated to fibromyalgia with low comorbidities (Docampo et al., 2014). Another GWAS study suggested rs2952768 in the Chr2q33.3 was involved with analgesic requirements in humans (Nishizawa et al., 2014). These GWAS have shed light on the elucidation of the genetic pathways for pain while further research is needed, including replication studies, functional studies, and agreement on feasible, valid and reproducible phenotype ascertainment. The limitations of our study include that the P values of tops SNPs are only close to GWAS significance but yet reached; no replication study to confirm the results; though the case definition is matched with those used epidemiological studies, we might misclassify some cases who have neuropathic pain but not prescribed medications into controls; we might also misclassify an individual into a control group who uses opioid to treat neuropathic pain. We have provided genetic evidence that SNPs in Chr1p35.1 (ZSCAN20-TLR12P) and Chr8p23.1 (HMGB1P46) may be involved with neuropathic pain in diabetes. Sex-specific associations are also suggested. Our findings should be treated with caution and, while we have also presented their consistency with known biological factors, they can only guide the nature of future research, which will be based on the findings reported in this paper. Any replication of our findings will help to confirm hypothesised pathways involved in the genetic mechanisms of neuropathic pain and provoke research on new potential drug targets for the treatment of pain. The following is the supplementary data related to this article.
    Competing Interests
    Author Contributions
    Role of the Funding Sources
    Acknowledgements
    Introduction