Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Qualitative sex differences in pain processing: emerging evidence of a biased literature

Abstract

Although most patients with chronic pain are women, the preclinical literature regarding pain processing and the pathophysiology of chronic pain has historically been derived overwhelmingly from the study of male rodents. This Review describes how the recent adoption by a number of funding agencies of policies mandating the incorporation of sex as a biological variable into preclinical research has correlated with an increase in the number of studies investigating sex differences in pain and analgesia. Trends in the field are analysed, with a focus on newly published findings of qualitative sex differences: that is, those findings that are suggestive of differential processing mechanisms in each sex. It is becoming increasingly clear that robust differences exist in the genetic, molecular, cellular and systems-level mechanisms of acute and chronic pain processing in male and female rodents and humans.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Analysis of quantitative and qualitative sex differences in the pain literature.

Similar content being viewed by others

References

  1. Institute of Medicine. Relieving Pain in America: A Blueprint for Transforming Prevention, Care, Education, and Research (National Academies Press, 2011).

  2. GBD 2017 Disease and Injury Incidence and Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 392, 1789–1858 (2018).

    Article  Google Scholar 

  3. Ruau, D., Liu, L. Y., Clark, J. D., Angst, M. S. & Butte, A. J. Sex differences in reported pain across 11,000 patients captured in electronic medical records. J. Pain 13, 228–234 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Unruh, A. M. Gender variations in clinical pain experience. Pain 65, 123–167 (1996).

    Article  CAS  PubMed  Google Scholar 

  5. Berkley, K. J. Sex differences in pain. Behav. Brain Sci. 20, 371–380 (1997).

    Article  CAS  PubMed  Google Scholar 

  6. Mogil, J. S. Sex differences in pain and pain inhibition: multiple explanations of a controversial phenomenon. Nat. Rev. Neurosci. 13, 859–866 (2012).

    Article  CAS  PubMed  Google Scholar 

  7. Fillingim, R. B., King, C. D., Riberio-Dasilva, M. C., Rahim-Williams, B. & Riley, J. L. III Sex, gender, and pain: a review of recent clinical and experimental findings. J. Pain 10, 447–485 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Kannan, V. D. & Veazie, P. J. Who avoids going to the doctor and why? Audience segmentation analysis for application of message development. Health Commun. 30, 635–645 (2015).

    Article  PubMed  Google Scholar 

  9. Abraham, A. et al. Sex differences in neuropathic pain intensity in diabetes. J. Neurol. Sci. 388, 103–106 (2018).

    Article  PubMed  Google Scholar 

  10. Cardinez, N. et al. Sex differences in neuropathic pain in longstanding diabetes: results from the Canadian Study of Longevity in Type 1 Diabetes. J. Diabetes Complicat. 32, 660–664 (2018).

    Article  Google Scholar 

  11. Zheng, H. et al. Age and preoperative pain are major confounders for sex differences in postoperative pain outcome: a prospective database analysis. PLoS One 12, e0178659 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Ostrom, C. et al. Demographic predictors of pain sensitivity: results from the OPPERA study. J. Pain. 18, 295–307 (2017).

    Article  PubMed  Google Scholar 

  13. Gobina, I. et al. Prevalence of self-reported chronic pain among adolescents: evidence from 42 countries and regions. Eur. J. Pain 23, 316–326 (2019).

    Article  PubMed  Google Scholar 

  14. Pitcher, M. H., Von Korff, M., Bushnell, M. C. & Porter, L. Prevalence and profile of high-impact chronic pain in the United States. J. Pain 20, 146–160 (2019).

    Article  PubMed  Google Scholar 

  15. Steingrimsdottir, O. A., Landmark, T., Macfarlane, G. J. & Nielsen, C. S. Defining chronic pain in epidemiological studies: a systematic review and meta-analysis. Pain 158, 2092–2107 (2017).

    Article  PubMed  Google Scholar 

  16. Mogil, J. S. & Chanda, M. L. The case for the inclusion of female subjects in basic science studies of pain. Pain 117, 1–5 (2005).

    Article  PubMed  Google Scholar 

  17. Beery, A. K. & Zucker, I. Sex bias in neuroscience and biomedical research. Neurosci. Biobehav. Rev. 35, 565–572 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Will, T. R. et al. Problems and progress regarding sex bias and omission in neuroscience research. eNeuro 4, e0278.0217.2017 (2017).

    Article  Google Scholar 

  19. Fields, R. D. Sex in science: the NIH Gets it wrong? in BrainFacts.org https://www.brainfacts.org/in-the-lab/animals-in-research/2014/sex-in-science--the-nih-gets-it-wrong (2014).

  20. Prendergast, B. J., Onishi, K. G. & Zucker, I. Female mice liberated for inclusion in neuroscience and biomedical research. Neurosci. Biobehav. Rev. 40, 1–5 (2014).

    Article  PubMed  Google Scholar 

  21. Itoh, Y. & Arnold, A. P. Are females more variable than males in gene expression? Meta-analysis of microarray datasets. Biol. Sex. Diff. 6, 18 (2015).

    Article  CAS  Google Scholar 

  22. Becker, J. B., Prendergast, B. J. & Liang, J. W. Female rats are not more variable than male rats: a meta-analysis of neuroscience studies. Biol. Sex. Diff. 7, 34 (2016).

    Article  Google Scholar 

  23. Beery, A. K. Inclusion of females does not increase variability in rodent research studies. Curr. Opin. Behav. Sci. 23, 143–149 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Tannenbaum, C., Ellis, R., Eyssel, F., Zou, J. & Schiebinger, L. Sex and gender analysis improves science and engineering. Nature 575, 137–146 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Fields, R. D. NIH policy: mandate goes too far. Nature 510, 340 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Richardson, S. S., Reiches, M., Shattuck-Heidorn, H., LaBonte, M. L. & Consoli, T. Focus on preclinical sex differences will not address women’s and men’s health disparities. Proc. Natl Acad. Sci. USA 112, 13419–13420 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mogil, J. S. Equality need not be painful. Nature 535, S7 (2016).

    Article  PubMed  Google Scholar 

  28. Shansky, R. M. Are hormones a “female problem” for animal research? Science 364, 825–826 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Riley III, J. L., Robinson, M. E., Wise, E. A., Myers, C. D. & Fillingim, R. B. Sex differences in the perception of noxious experimental stimuli: a meta-analysis. Pain 74, 181–187 (1998).

    Article  PubMed  Google Scholar 

  30. Averitt, D. L., Eidson, L. N., Doyle, H. H. & Murphy, A. Z. Neuronal and glial factors contributing to sex differences in opioid modulation of pain. Neuropsychopharmacology 44, 155–165 (2019).

    Article  CAS  PubMed  Google Scholar 

  31. Sorge, R. E. & Strath, L. J. Sex differences in pain responses. Curr. Opin. Physiol. 6, 75–81 (2018).

    Article  Google Scholar 

  32. Mogil, J. S. Sex-based divergence of mechanisms underlying pain and pain inhibition. Curr. Opin. Behav. Sci. 23, 113–117 (2018).

    Article  Google Scholar 

  33. Coraggio, V. et al. Neuroimmune-driven neuropathic pain establishment: a focus on gender differences. Int. J. Mol. Sci. 19, 281 (2018).

    Article  PubMed Central  CAS  Google Scholar 

  34. Melchior, M., Poisbeau, P., Gaumond, I. & Marchand, S. Insights into the mechanisms and the emergence of sex-differences in pain. Neuroscience 338, 63–80 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Boerner, K. E. et al. The conceptual complexity of gender and its relevance to pain. Pain 159, 2137–2141 (2018).

    Article  PubMed  Google Scholar 

  36. Racine, M. et al. A systematic literature review of 10 years of research on sex/gender and experimental pain perception – Part 1: are there really differences between women and men? Pain 153, 602–618 (2012).

    Article  PubMed  Google Scholar 

  37. Hashmi, J. A. & Davis, K. D. Deconstructing sex differences in pain sensitivity. Pain 155, 10–13 (2014).

    Article  PubMed  Google Scholar 

  38. Chen, G., Luo, X., Qadri, M. Y., Berta, T. & Ji, R.-R. Sex-dependent glial signaling in pathological pain: distinct roles of spinal microglia and astrocytes. Neurosci. Bull. 34, 98–108 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Gutierrez, S., Hayashida, K. & Eisenach, J. C. The puerperium alters spinal cord plasticity following peripheral nerve injury. Neuroscience 228, 301–308 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Sorge, R. E. et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat. Neurosci. 18, 1081–1083 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mapplebeck, J. C. S. et al. Chloride dysregulation through downregulation of KCC2 mediates neuropathic pain in both sexes. Cell Rep. 28, 590–596 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. McCarthy, M. M., Arnold, A. P., Ball, G. F., Blaustein, J. D. & De Vries, G. J. Sex differences in the brain: the not so inconvenient truth. J. Neurosci. 32, 2241–2247 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Mogil, J. S. Interaction between sex and genotype in the mediation of pain and pain inhibition. Semin. Pain. Med. 1, 197–205 (2003).

    Article  Google Scholar 

  44. Mogil, J. S. & Belknap, J. K. Sex and genotype determine the selective activation of neurochemically-distinct mechanisms of swim stress-induced analgesia. Pharmacol. Biochem. Behav. 56, 61–66 (1997).

    Article  CAS  PubMed  Google Scholar 

  45. Kest, B., Wilson, S. G. & Mogil, J. S. Sex differences in supraspinal morphine analgesia are dependent on genotype. J. Pharmacol. Exp. Ther. 289, 1370–1375 (1999).

    CAS  PubMed  Google Scholar 

  46. Cook, C. D., Barrett, A. C., Roach, E. L., Bowman, J. R. & Picker, M. J. Sex-related differences in the antinociceptive effects of opioids: importance of rat genotype, nociceptive stimulus intensity, and efficacy at the µ opioid receptor. Psychopharmacology 150, 430–442 (2000).

    Article  CAS  PubMed  Google Scholar 

  47. Mogil, J. S., Chesler, E. J., Wilson, S. G., Juraska, J. M. & Sternberg, W. F. Sex differences in thermal nociception and morphine antinociception in rodents depend on genotype. Neurosci. Biobehav. Rev. 24, 375–389 (2000).

    Article  CAS  PubMed  Google Scholar 

  48. Barrett, A. C. et al. Sex and rat strain determine sensitivity to κ opioid-induced antinociception. Psychopharmacology 160, 170–181 (2002).

    Article  CAS  PubMed  Google Scholar 

  49. Terner, J. M., Barrett, A. C., Grossell, E. & Picker, M. J. Influence of gonadectomy on the antinociceptive effects of opioids in male and female rats. Psychopharmacology 163, 183–193 (2002).

    Article  CAS  PubMed  Google Scholar 

  50. Terner, J. M., Barrett, A. C., Cook, C. D. & Picker, M. J. Sex differences in (-)-pentazocine antinociception: comparison to morphine and spiradoline in four rat strains using a thermal nociceptive assay. Behav. Pharmacol. 14, 77–85 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Vendruscolo, L. F., Pamplona, F. A. & Takahashi, R. N. Strain and sex differences in the expression of nociceptive behavior and stress-induced analgesia in rats. Brain Res. 1030, 277–283 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. LaCroix-Fralish, M. L., Mogil, J. S., Weinstein, J. N., Rutkowski, M. D. & DeLeo, J. A. The magnitude of mechanical allodynia in a rodent model of lumbar radiculopathy is dependent on strain and sex. Spine 30, 1821–1827 (2005).

    Article  PubMed  Google Scholar 

  53. Abdus-Saboor, I. et al. Development of a mouse pain scale using sub-second behavioral mapping and statistical modeling. Cell Rep. 28, 1623–1634 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Devor, M. et al. Sex-specific variability and a ‘cage effect’ independently mask a neuropathic pain quantitative trait locus detected in a whole genome scan. Eur. J. Neurosci. 26, 681–688 (2007).

    Article  PubMed  Google Scholar 

  55. Mogil, J. S. et al. Identification of a sex-specific quantitative trait locus mediating nonopioid stress-induced analgesia in female mice. J. Neurosci. 17, 7995–8002 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Mogil, J. S. et al. Pain sensitivity and vasopressin analgesia are mediated by a gene-sex-environment interaction. Nat. Neurosci. 14, 1569–1573 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Bryant, C. D. et al. C57BL/6 substrain differences in inflammatory and neuropathic nociception and genetic mapping of a major quantitative trait locus underlying acute thermal nociception. Mol. Pain 15, 1–15 (2019).

    Article  CAS  Google Scholar 

  58. Mogil, J. S. et al. Variable sensitivity to noxious heat is mediated by differential expression of the CGRP gene. Proc. Natl. Acad. Sci. USA 102, 12938–12943 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Furuya, T. et al. Genetic dissection of a rat model for rheumatoid arthritis: significant gender influences on autosomal modifier loci. Hum. Mol. Genet. 9, 2241–2250 (2000).

    Article  CAS  PubMed  Google Scholar 

  60. Dominguez, C. A. et al. Genetic and sex influence on neuropathic pain-like behaviour after spinal cord injury in the rat. Eur. J. Pain 16, 1368–1377 (2012).

    Article  CAS  PubMed  Google Scholar 

  61. Mogil, J. S. et al. The melanocortin-1 receptor gene mediates female-specific mechanisms of analgesia in mice and humans. Proc. Natl Acad. Sci. USA 100, 4867–4872 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Valverde, P., Healy, E., Jackson, I., Rees, J. L. & Thody, A. J. Variants of the melanocyte-stimulating hormone receptor gene are associated with red hair and fair skin in humans. Nat. Genet. 11, 328–330 (1995).

    Article  CAS  PubMed  Google Scholar 

  63. Ballou, L. R., Botting, R. M., Goorha, S., Zhang, J. & Vane, J. R. Nociception in cyclooxygenase isozyme-deficient mice. Proc. Natl Acad. Sci. USA 97, 10272–10276 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Blednov, Y. A., Stoffel, M., Alva, H. & Harris, R. A. A pervasive mechanism for analgesia: activation of GIRK2 channels. Proc. Natl Acad. Sci. USA 100, 277–282 (2003).

    Article  CAS  PubMed  Google Scholar 

  65. Martin, M., Matifas, A., Maldonado, R. & Kieffer, B. L. Acute antinociceptive responses in single and combinatorial opioid receptor knockout mice: distinct mu, delta and kappa tones. Eur. J. Neurosci. 17, 701–708 (2003).

    Article  PubMed  Google Scholar 

  66. Mitrovic, I. et al. Contribution of GIRK2-mediated postsynaptic signaling to opiate and α2-adrenergic analgesia and analgesic sex differences. Proc. Natl Acad. Sci. USA 100, 271–276 (2003).

    Article  CAS  PubMed  Google Scholar 

  67. Patil, M. J., Green, D. P., Henry, M. A. & Akopian, A. N. Sex-dependent roles of prolactin and prolactin receptor in postoperative pain and hyperalgesia in mice. Neuroscience 253, 132–141 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Nasir, H. et al. Consistent sex-dependent effects of PKMζ gene ablation and pharmacological inhibition on the maintenance of referred pain. Mol. Pain 12, 1–14 (2016).

    Article  CAS  Google Scholar 

  69. AlSharari, S. D. et al. Sex differences and drug dose influence the role of the α7 nicotinic acetylcholine receptor in the mouse dextran sodium sulfate-induced colitis model. Nicotine Tob. Res. 19, 460–468 (2017).

    Article  CAS  PubMed  Google Scholar 

  70. Khariv, V. et al. Impaired sensitivity to pain stimuli in plasma membrane calcium ATPase 2 (PMCA2) heterozygous mice: a possible modality- and sex-specific role for PMCA2 in nociception. FASEB J. 31, 224–237 (2017).

    Article  CAS  PubMed  Google Scholar 

  71. Liu, P. et al. Dopamine D3 receptor knockout mice exhibit abnormal nociception in a sex-different manner. J. Neurosci. Res. 95, 1438–1445 (2017).

    Article  CAS  PubMed  Google Scholar 

  72. Megat, S. et al. A critical role for dopamine D5 receptors in pain chronicity in male mice. J. Neurosci. 38, 379–397 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. del Rivero, T., Fischer, R., Yang, F., Swanson, K. A. & Bethea, J. R. Tumor necrosis factor receptor 1 inhibition is therapeutic for neuropathic pain in males but not in females. Pain 160, 922–931 (2019).

    Article  PubMed  CAS  Google Scholar 

  74. Azkona, G. et al. Striatal-enriched protein tyrosine phosphatase modulates nociception: evidence from genetic deletion and pharmacological inhibition. Pain 157, 377–386 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Cowie, A. M., Menzel, A. D., O’hara, C., Lawlor, M. W. & Stucky, C. L. NOD-like receptor protein 3 inflammasome drives postoperative mechanical pain in a sex-dependent manner. Pain 160, 1794–1816 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Fillingim, R. B. et al. The A118G single nucleotide polymorphism of the µ-opioid receptor gene (OPRM1) is associated with pressure pain sensitivity in humans. J. Pain 6, 159–167 (2005).

    Article  CAS  PubMed  Google Scholar 

  77. Olsen, M. B. et al. Pain intensity the first year after lumbar disc herniation is associated with the A118G polymorphism in the opioid receptor mu 1 gene: evidence of a sex and genotype interaction. J. Neurosci. 32, 9831–9834 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Linnstaedt, S. D. et al. µ-Opioid receptor gene A118G variants and persistent pain symptoms among men and women experiencing motor vehicle collision. J. Pain 16, 637–644 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Aoki, J. et al. Association between 5-hydroxytryptamine 2A receptor gene polymorphism and postoperative analgesic requirements after major abdominal surgery. Neurosci. Lett. 479, 40–43 (2010).

    Article  CAS  PubMed  Google Scholar 

  80. Lebe, M. et al. Association of serotonin-1A and -2A receptor promoter polymorphisms with depressive symptoms, functional recovery, and pain in patients 6 months after lumbar disc surgery. Pain 154, 377–384 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Lindstedt, F., Lonsdorf, T. B., Schalling, M., Kosek, E. & Ingvar, M. Perception of thermal pain and the thermal grill illusion is associated with polymorphisms in the serotonin transporter gene. PLoS One 6, e17752 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Belfer, I. et al. Pain modality- and sex-specific effects of COMT functional variants. Pain 154, 1368–1376 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Meloto, C. B. et al. Modification of COMT-dependent pain sensitivity by psychological stress and sex. Pain 157, 858–867 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Sato, H. et al. Gender, variation in opioid receptor genes and sensitivity to experimental pain. Mol. Pain 9, 20 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Belfer, I. et al. A GCH1 haplotype confers sex-specific susceptibility to pain crises and altered endothelial function in adults with sickle cell anemia. Am. J. Hematol. 89, 187–193 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Ursu, D. et al. Gain and loss of function of P2X7 receptors: mechanisms, pharmacology and relevance to diabetic neuropathic pain. Mol. Pain 10, 37 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Meng, W. 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 2, 1386–1393 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Sanders, A. E. et al. GWAS identifies new loci for painful temporomandibular disorder: Hispanic community health study/study of Latinos. J. Dent. Res. 96, 277–284 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Smith, S. B. et al. Genome-wide association reveals contribution of MRAS to painful temporomandibular disorder in males. Pain 160, 579–591 (2019).

    Article  CAS  PubMed  Google Scholar 

  90. Korczeniewska, O. A. et al. Differential gene expression in trigeminal ganglia of male and female rats following chronic constriction of the infraorbital nerve. Eur. J. Pain 22, 875–888 (2018).

    Article  CAS  PubMed  Google Scholar 

  91. Stephens, K. E. et al. Sex differences in gene regulation in the dorsal root ganglion after nerve injury. BMC Genomics 20, 147 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  92. Ray, P. R. et al. Transcriptome analysis of the human tibial nerve identifies sexually dimorphic expression of genes involved in pain, inflammation, and neuro-immunity. Front. Mol. Neurosci. 12, 37 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. North, R. Y. et al. Electrophysiological and transcriptomic correlates of neuropathic pain in human dorsal root ganglion neurons. Brain 142, 1215–1226 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  94. Avona, A. et al. Dural calcitonin gene-related peptide produces female-specific responses in rodent migraine models. J. Neurosci. 39, 4323–4331 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Chow, L.-H. et al. Sex difference of angiotensin IV-, LVV-hemorphin 7-, and oxytocin-induced antiallodynia at the spinal level in mice with neuropathic pain. Anesth. Analg. 126, 2093–2101 (2019).

    Article  CAS  Google Scholar 

  96. Ferrari, L. F., Khomula, E. V., Araldi, D. & Levine, J. D. Marked sexual dimorphism in the role of the ryanodine receptor in a model of pain chronification in the rat. Sci. Rep. 6, 31221 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Joseph, E. K., Parada, C. A. & Levine, J. D. Hyperalgesic priming in the rat demonstrates marked sexual dimorphism. Pain 105, 143–150 (2003).

    Article  CAS  PubMed  Google Scholar 

  98. Araldi, D., Ferrari, L. F., Green, P. & Levine, J. D. Marked sexual dimorphism in 5-HT1 receptors mediating pronociceptive effects of sumatriptan. Neuroscience 344, 394–405 (2017).

    Article  CAS  PubMed  Google Scholar 

  99. Tonsfeldt, K. J. et al. Sex differences in GABAA signaling in the periaqueductal gray induced by persistent inflammation. J. Neurosci. 36, 1669–1681 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Mogil, J. S., Sternberg, W. F., Kest, B., Marek, P. & Liebeskind, J. C. Sex differences in the antagonism of swim stress-induced analgesia: effects of gonadectomy and estrogen replacement. Pain 53, 17–25 (1993).

    Article  CAS  PubMed  Google Scholar 

  101. Arout, C. A., Caldwell, M., Rossi, G. & Kest, B. Spinal and supraspinal N-methyl-D-aspartate and melanocortin-1 receptors contribute to a qualitative sex difference in morphine-induced hyperalgesia. Physiol. Behav. 147, 364–372 (2015).

    Article  CAS  PubMed  Google Scholar 

  102. George, N. C., Laferriere, A. & Coderre, T. J. Sex differences in the contributions of spinal atypical PKCs and downstream targets to the maintenance of nociceptive sensitization. Mol. Pain 15, 1–12 (2019).

    Article  CAS  Google Scholar 

  103. Hagiwara, H., Funabashi, T., Akema, T. & Kimura, F. Sex-specific differences in pain response by dopamine in the bed nucleus of the stria terminalis in rats. Neuroreport 24, 181–185 (2013).

    Article  CAS  PubMed  Google Scholar 

  104. Joseph, E. K. & Levine, J. D. Sexual dimorphism in the contribution of protein kinase C isoforms to nociception in the streptozotocin diabetic rat. Neuroscience 120, 907–913 (2003).

    Article  CAS  PubMed  Google Scholar 

  105. Ceriani, C. E. J., Wilhour, D. A. & Silberstein, S. D. Novel medications for the treatment of migraine. Headache 59, 1597–1608 (2019).

    Article  PubMed  Google Scholar 

  106. Morales-Medina, J. C., Flores, G., Vallelunga, A., Griffiths, N. H. & Iannitti, T. Cerebrolysin improves peripheral inflammatory pain: sex differences in two models of acute and chronic mechanical hypersensitivity. Drug. Dev. Res. 80, 513–518 (2019).

    Article  CAS  PubMed  Google Scholar 

  107. Zammataro, M. et al. Chronic treatment with fluoxetine induces sex-dependent analgesic effects and modulates HDAC2 and mGlu2 expression in female mice. Front. Pharmacol. 8, 743 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Mifflin, K. A. et al. Voluntary wheel running reveals sex-specific nociceptive factors in murine experimental autoimmune encephalomyelitis. Pain 160, 870–881 (2019).

    Article  CAS  PubMed  Google Scholar 

  109. Cooper, Z. D. & Haney, M. Sex-dependent effects of cannabis-induced analgesia. Drug Alcohol Depend. 167, 112–120 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  110. Gear, R. W. et al. Kappa-opioids produce significantly greater analgesia in women than in men. Nat. Med. 2, 1248–1250 (1996).

    Article  CAS  PubMed  Google Scholar 

  111. Fillingim, R. B. et al. Experimental pain models reveal no sex differences in pentazocine analgesia in humans. Anesthesiology 100, 1263–1270 (2004).

    Article  CAS  PubMed  Google Scholar 

  112. Scheff, N. N. et al. Neutrophil-mediated endogenous analgesia contributes to sex differences in oral cancer pain. Front. Integr. Neurosci. 12, 52 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Long, C. C., Sadler, K. E. & Kolber, B. J. Hormonal and molecular effects of restraint stress on formalin-induced pain-like behavior in male and female mice. Physiol. Behav. 165, 278–285 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Kuner, R. & Flor, H. Structural plasticity and reorganisation in chronic pain. Nat. Rev. Neurosci. 18, 20–30 (2016).

    Article  PubMed  CAS  Google Scholar 

  115. Juarez, I. et al. Tooth pulp injury induces sex-dependent neuronal reshaping in the ventral posterolateral nucleus of the rat thalamus. J. Chem. Neuroanat. 96, 16–21 (2019).

    Article  PubMed  Google Scholar 

  116. Shiers, S. et al. Neuropathic pain creates an enduring prefrontal cortex dysfunction corrected by the type II diabetic drug metformin but not by gapabentin. J. Neurosci. 38, 7337–7350 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Nag, S. & Mokha, S. S. Activation of the trigeminal α2-adrenoceptor produces sex-specific, estrogen dependent thermal antinociception and antihyperalgesia using an operant pain assay in the rat. Behav. Brain Res. 314, 152–158 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Nag, S. & Mokha, S. S. Activation of α2-adrenoceptors in the trigeminal region produces sex-specific modulation of nociception in the rat. Neuroscience 142, 1255–1262 (2006).

    Article  CAS  PubMed  Google Scholar 

  119. Thompson, A. D., Angelotti, T., Nag, S. & Mokha, S. S. Sex-specific modulation of spinal nociception by α2-adrenoceptors: differential regulation by estrogen and testosterone. Neuroscience 153, 1268–1277 (2008).

    Article  CAS  PubMed  Google Scholar 

  120. Chakrabarti, S., Liu, N.-J. & Gintzler, A. R. Formation of µ-/κ-opioid receptor heterodimer is sex-dependent and mediates female-specific opioid analgesia. Proc. Natl Acad. Sci. USA 107, 20115–20119 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Gupta, D. S., Von Gizycki, H. & Gintzler, A. R. Sex-/ovarian steroid-dependent release of endomorphin 2 from spinal cord. J. Pharmacol. Exp. Ther. 321, 635–641 (2007).

    Article  CAS  PubMed  Google Scholar 

  122. Kumar, A., Liu, N.-J., Madia, P. A. & Gintzler, A. R. Contribution of endogenous spinal endomorphin 2 to intrathecal opioid antinociception in rats is agonist dependent and sexually dimorphic. J. Pain 16, 1200–1210 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Liu, N.-J., Schnell, S., Wessendorf, M. W. & Gintzler, A. R. Sex, pain, and opioids: interdependent influences of sex and pain modality on dynorphin-mediated antinociception in rats. J. Pharmacol. Exp. Ther. 344, 522–530 (2013).

    Article  CAS  PubMed  Google Scholar 

  124. Liu, N.-J., von Gizycki, H. & Gintzler, A. R. Sexually dimorphic recruitment of spinal opioid analgesic pathways by the spinal application of morphine. J. Pharmacol. Exp. Ther. 322, 654–660 (2007).

    Article  CAS  PubMed  Google Scholar 

  125. Lawson, K. P., Nag, S., Thompson, A. D. & Mokha, S. S. Sex-specificity and estrogen-dependence of kappa opioid receptor-mediated antinociception and antihyperalgesia. Pain 151, 806–815 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Loyd, D. R., Wang, X. & Murphy, A. Z. Sex differences in μ-opioid receptor expression in the rat midbrain periaqueductal gray are essential for eliciting sex differences in morphine analgesia. J. Neurosci. 28, 14007–14017 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Tershner, S. A., Mitchell, J. M. & Fields, H. L. Brainstem pain modulating circuitry is sexually dimorphic with respect to mu and kappa opioid receptor function. Pain 85, 153–159 (2000).

    Article  CAS  PubMed  Google Scholar 

  128. Ji, R.-R., Chamessian, A. & Zhang, Y.-Q. Pain regulation by non-neuronal cells and inflammation. Science 354, 572–577 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Grace, P. M., Hutchinson, M. R., Maier, S. F. & Watkins, L. R. Pathological pain and the neuroimmune interface. Nat. Rev. Immunol. 14, 217–231 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Chen, G., Zhang, Y.-Q., Qadri, Y. J., Serhan, C. N. & Ji, R.-R. Microglia in pain: detrimental and protective roles in pathogenesis and resolution of pain. Neuron 100, 1292–1311 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Tanga, F. Y., Nutile-McMenemy, N. & DeLeo, J. A. The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proc. Natl Acad. Sci. USA 102, 5856–5861 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Sorge, R. E. et al. Genetically determined P2X7 receptor pore formation regulates variability in chronic pain sensitivity. Nat. Med. 18, 595–599 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Bastos, L. F. S. et al. Sex-independent suppression of experimental inflammatory pain by minocycline in two mouse strains. Neurosci. Lett. 553, 110–114 (2013).

    Article  CAS  PubMed  Google Scholar 

  134. Sorge, R. E. et al. Spinal cord Toll-like receptor 4 mediates inflammatory and neuropathic hypersensitivity in male but not female mice. J. Neurosci. 31, 15450–15454 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Beggs, S. & Salter, M. W. Microglia-neuronal signalling in neuropathic pain hypersensitivity 2.0. Curr. Opin. Neurobiol. 20, 474–480 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. The Editorial Board. Why science needs female mice. New York Times 10 (19 July 2015).

  137. Berta, T., Qadri, Y. J., Chen, G. & Ji, R.-R. Microglial signaling in chronic pain with a special focus on caspase 6, p38 MAP kinase, and sex dependence. J. Dent. Res. 95, 1124–1131 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Taves, S. et al. Spinal inhibition of p38 MAP kinase reduces inflammatory and neuropathic pain in male but not female mice: sex-dependent microglial signaling in the spinal cord. Brain Behav. Immun. 55, 70–81 (2016).

    Article  CAS  PubMed  Google Scholar 

  139. Woller, S. A. et al. Systemic TAK-242 prevents intrathecal LPS evoked hyperalgesia in male, but not female mice and prevents delayed allodynia following intraplantar formalin in both male and female mice: The role of TLR4 in the evolution of a persistent pain state. Brain Behav. Immun. 56, 271–280 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Luo, X. et al. Intrathecal administration of antisense oligonucleotide against p38a but not p38b MAP kinase isoform reduces neuropathic and postoperative pain and TLR4-induced pain in male mice. Brain Behav. Immun. 72, 34–44 (2018).

    Article  CAS  PubMed  Google Scholar 

  141. Mapplebeck, J. C. S. et al. Microglial P2X4R-evoked pain hypersensitivity is sexually dimorphic in rats. Pain 159, 1752–1763 (2018).

    CAS  PubMed  Google Scholar 

  142. Paige, C., Mejia, G., Batchalli, G. M., Dussor, G. & Price, T. Spinal inhibition of P2XR or p38 signaling disrupts hyperalgesic priming in male, but not female, mice. Neuroscience 385, 133–142 (2018).

    Article  CAS  PubMed  Google Scholar 

  143. Fonken, L. K. et al. Neuroinflammatory priming to stress is differentially regulated in male and female rats. Brain Behav. Immun. 70, 257–267 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  144. Moriarty, O. et al. Priming of adult incision response by early-life injury: neonatal microglial inhibition has persistent but sexually dimorphic effects in adult rats. J. Neurosci. 39, 3081–3093 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Inyang, K. E. et al. The antidiabetic drug metformin prevents and reverses neuropathic pain and spinal cord microglial activation in male but not female mice. Pharmacol. Res. 139, 1–16 (2019).

    Article  CAS  PubMed  Google Scholar 

  146. Rosen, S. F. et al. T cell mediation of pregnancy analgesia affecting chronic pain in mice. J. Neurosci. 37, 9819–9827 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Chen, M. J. et al. Astrocytic Cx43 hemichannels and gap junctions play a crucial role in development of chronic neuropathic pain following spinal cord injury. Glia 60, 1660–1670 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  148. Yang, Y. et al. Delayed activation of spinal microglia contributes to the maintenance of bone cancer pain in female Wistar rats via P2X7 receptor and IL-18. J. Neurosci. 35, 7950–7963 (2015).

    Article  CAS  PubMed  Google Scholar 

  149. Nieto, F. R. et al. Neuron-immune mechanisms contribute to pain in early stages of arthritis. J. Neuroinflamm. 13, 96 (2016).

    Article  CAS  Google Scholar 

  150. Peng, J. et al. Microglia and monocytes synergistically promote the transition from acute to chronic pain after nerve injury. Nat. Commun. 7, 12029 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Posillico, C. K., Terasaki, L. S., Bilbo, S. D. & Schwarz, J. M. Examination of sex and minocycline treatment on acute morphine-induced analgesia and inflammatory gene expression along the pain pathway in Sprague–Dawley rats. Biol. Sex. Diff. 6, 33 (2015).

    Article  CAS  Google Scholar 

  152. Doyle, H. H., Eidson, L. N., Sinkiewicz, D. M. & Murphy, A. Z. Sex differences in microglia activity within the peraqueductal gray of the rat: a potential mechanism driving the dimorphic effects of morphine. J. Neurosci. 37, 3202–3214 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Catuneanu, A., Paylor, J. W., Winship, I., Colbourne, F. & Kerr, B. J. Sex differences in central nervous system plasticity and pain in experimental autoimmune encephalomyelitis. Pain 160, 1037–1049 (2019).

    Article  PubMed  Google Scholar 

  154. Moalem-Taylor, G., Allbutt, H. N., Iordanova, M. D. & Tracey, D. J. Pain hypersensitivity in rats with experimental autoimmune neuritis, an animal model of human inflammatory demyelinating neuropathy. Brain Behav. Immun. 21, 699–710 (2007).

    Article  CAS  PubMed  Google Scholar 

  155. Sweitzer, S. M., Hickey, W. F., Rutkowski, M. D., Pahl, J. L. & Deleo, J. A. Focal peripheral nerve injury induces leukocyte trafficking into the central nervous system: potential relationship to neuropathic pain. Pain 100, 163–170 (2002).

    Article  PubMed  Google Scholar 

  156. Costigan, M. et al. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J. Neurosci. 29, 14415–14422 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Moalem, G., Xu, K. & Yu, L. T lymphocytes play a role in neuropathic pain following peripheral nerve injury in rats. Neuroscience 129, 767–777 (2004).

    Article  CAS  PubMed  Google Scholar 

  158. Kleinschnitz, C. et al. T cell infiltration after chronic constriction injury of mouse sciatic nerve is associated with interleukin-17 expression. Exp. Neurol. 200, 480–485 (2006).

    Article  CAS  PubMed  Google Scholar 

  159. Hu, P., Bembrick, A. L., Keay, K. A. & McLachlan, E. M. Immune cell involvement in dorsal root ganglia and spinal cord after chronic constriction or transection of the rat sciatic nerve. Brain Behav. Immun. 21, 599–616 (2007).

    Article  CAS  PubMed  Google Scholar 

  160. Cao, L. & DeLeo, J. A. CNS-infiltrating CD4+ T lymphocytes contribute to murine spinal nerve transection-induced neuropathic pain. Eur. J. Immunol. 38, 448–458 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Lopes, D. M. et al. Sex differences in peripheral not central immune responses to pain-inducing injury. Sci. Rep. 7, 16460 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  162. Gattlen, C. et al. Spinal cord T-cell infiltration in the rat spared nerve injury model: a time course study. Int. J. Mol. Sci. 17, 352 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  163. Du, B. et al. CD4+ αβ T cell infiltration into the leptomeninges of lumbar dorsal roots contributes to the transition from acute to chronic mechanical allodynia after adult rat tibial nerve injuries. J. Neuroinflamm. 15, 81 (2018).

    Article  CAS  Google Scholar 

  164. Krukowski, K. et al. CD8+ T cells and endogenous IL-10 are required for resolution of chemotherapy-induced neuropathic pain. J. Neurosci. 36, 11074–11083 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Duffy, S. S. et al. Regulatory T cells and their derived cytokine, interleukin-35, reduce pain in experiemental autoimmune encephalomyelitis. J. Neurosci. 39, 2326–2346 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  166. Liu, X.-J. et al. Nociceptive neurons regulate innate and adaptive immunity and neuropathic pain through MyD88 adapter. Cell Res. 24, 1374–1377 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Baddack-Werncke, U. et al. Cytotoxic T cells modulate inflammation and endogenous opioid analgesia in chronic arthritis. J. Neuroinflamm. 14, 30 (2017).

    Article  CAS  Google Scholar 

  168. Rosen, S. et al. Increased pain sensitivity and decreased opioid analgesia in T cell-deficient mice and implications for sex differences. Pain 160, 358–366 (2019).

    Article  CAS  PubMed  Google Scholar 

  169. Hartlehnert, M. et al. Schwann cells promote posttraumatic nerve inflammation and neuropathic pain through MHC class II. Sci. Rep. 7, 12518 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  170. Song, Z. et al. High-fat diet exacerbates postoperative pain and inflammation in a sex-dependent manner. Pain 159, 1731–1741 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Luo, X. et al. Macrophage Toll-like receptor 9 contributes to chemotherapy-induced neuropathic pain in male mice. J. Neurosci. 39, 6848–6864 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Luo, X., Gu, Y., Tao, X., Serhan, C. N. & Ji, R.-R. Resolvin D5 inhibits neuropathic and inflammatory pain in male but not female mice: distinct actions of D-series resolvins in chemotherapy-induced peripheral neuropathy. Front. Pharmacol. 10, 745 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Martin, L. J. et al. Male-specific conditioned pain hypersensitivity in mice and humans. Curr. Biol. 29, 291–201.e294 (2019).

    Article  CAS  Google Scholar 

  174. Chesler, E. J., Wilson, S. G., Lariviere, W. R., Rodriguez-Zas, S. L. & Mogil, J. S. Identification and ranking of genetic and laboratory environment factors influencing a behavioral trait, thermal nociception, via computational analysis of a large data archive. Neurosci. Biobehav. Rev. 26, 907–923 (2002).

    Article  PubMed  Google Scholar 

  175. Mogil, J. S. Laboratory environmental factors and pain behavior: the relevance of unknown unknowns to reproducibility and translation. Lab. Anim. 46, 136–141 (2017).

    Article  Google Scholar 

  176. Sorge, R. E. et al. Olfactory exposure to males, including men, causes stress and related analgesia in rodents. Nat. Meth. 11, 629–632 (2014).

    Article  CAS  Google Scholar 

  177. Greenwood-Van Meerveld, B. & Johnson, A. C. Stress-induced chronic visceral pain of gastrointestinal origin. Front. Syst. Neurosci. 11, 86 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Prusator, D. K. & Greenwood-Van Meerveld, B. Amygdala-mediated mechanisms regulate visceral hypersensitivity in adult females following early life stress: importance of the glucocorticoid receptor and corticotropin-releasing factor. Pain 158, 296–305 (2017).

    Article  CAS  PubMed  Google Scholar 

  179. Brown, K. J. & Grunberg, N. E. Effects of housing on male and female rats: crowding stresses males but calms females. Physiol. Behav. 58, 1085–1089 (1995).

    Article  CAS  PubMed  Google Scholar 

  180. Langford, D. L. et al. Social approach to pain in laboratory mice. Soc. Neurosci. 5, 163–170 (2010).

    Article  PubMed  Google Scholar 

  181. Langford, D. L. et al. Varying perceived social threat modulates pain behavior in male mice. J. Pain 12, 125–132 (2011).

    Article  PubMed  Google Scholar 

  182. Levine, F. M. & De Simone, L. L. The effects of experimenter gender on pain report in male and female subjects. Pain 44, 69–72 (1991).

    Article  CAS  PubMed  Google Scholar 

  183. Carter, L. E. et al. Effects of emotion on pain reports, tolerance and physiology. Pain. Res. Manag. 7, 21–30 (2002).

    Article  PubMed  Google Scholar 

  184. Essick, G., Guest, S., Martinez, E., Chen, C. & McGlone, F. Site-dependent and subject-related variations in perioral thermal sensitivity. Somatosens. Mot. Res. 21, 159–175 (2004).

    Article  PubMed  Google Scholar 

  185. Otto, M. W. & Dougher, M. J. Sex differences and personality factors in responsivity to pain. Percept. Mot. Skills 61, 383–390 (1985).

    Article  CAS  PubMed  Google Scholar 

  186. Gijsbers, K. & Nicholson, F. Experimental pain thresholds influenced by sex of experimenter. Percept. Mot. Skills 101, 803–807 (2005).

    Article  CAS  PubMed  Google Scholar 

  187. Aslaksen, P. M., Myrbakk, I. N., Hoifodt, R. S. & Flaten, M. A. The effect of experimenter gender on autonomic and subjective responses to pain stimuli. Pain 129, 260–268 (2007).

    Article  PubMed  Google Scholar 

  188. Kallai, I., Barke, A. & Voss, U. The effects of experimenter characteristics on pain reports in women and men. Pain 112, 142–147 (2004).

    Article  PubMed  Google Scholar 

  189. Stanke, K. M. & Ivanec, D. Pain threshold - measure of pain sensitivity or social behavior? Psihologija 49, 37–50 (2016).

    Article  Google Scholar 

  190. Vigil, J. M., Rowell, L. N., Alcock, J. & Maestes, R. Laboratory personnel gender and cold pressor apparatus affect subjective pain reports. Pain. Res. Manag. 19, e13–e18 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  191. Edwards, R., Eccleston, C. & Keogh, E. Observer influences on pain: an experimental series examining same-sex and opposite-sex friends, strangers, and romantic partners. Pain 158, 846–855 (2017).

    Article  PubMed  Google Scholar 

  192. Engebretsen, S. et al. The peer effect on pain tolerance. Scand. J. Pain 18, 467–477 (2018).

    Article  PubMed  Google Scholar 

  193. Vigil, J. M. et al. Sex differences in how social networks and relationship quality influence experimental pain sensitivity. PLoS One 8, e78663 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Tansley, S. N. et al. Modulation of social behavior and dominance status by chronic pain in mice. Genes. Brain Behav. 18, e12514 (2019).

    Article  PubMed  CAS  Google Scholar 

  195. Farmer, M. A. et al. Pain reduces sexual motivation in female but not male mice. J. Neurosci. 34, 5747–5753 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  196. Block, P., Heathcote, L. C. & Heyes, S. B. Social interaction and pain: an arctic expedition. Soc. Sci. Med. 196, 47–55 (2018).

    Article  PubMed  Google Scholar 

  197. Arnold, A. P. A general theory of sexual differentiation. J. Neurosci. Res. 95, 291–300 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Gioiosa, L., Chen, X., Watkins, R., Umeda, E. A. & Arnold, A. P. Sex chromosome complement affects nociception and analgesia in newborn mice. J. Pain 9, 962–969 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  199. Gioiosa, L. et al. Sex chromosome complement affects nociception in tests of acute and chronic exposure to morphine in mice. Horm. Behav. 53, 124–130 (2008).

    Article  CAS  PubMed  Google Scholar 

  200. Verriotis, M. et al. The distribution of pain activity across the human neonatal brain is sex dependent. NeuroImage 178, 69–77 (2018).

    Article  PubMed  Google Scholar 

  201. Burke, N. N. & Trang, T. Neonatal injury results in sex-dependent nociceptive hypersensitivity and social behavioral deficits during adolescence, without altering morphine response. J. Pain 18, 1384–1396 (2017).

    Article  PubMed  Google Scholar 

  202. Craft, R. M., Mogil, J. S. & Aloisi, A. M. Sex differences in pain and analgesia: the role of gonadal hormones. Eur. J. Pain 8, 397–411 (2004).

    Article  CAS  PubMed  Google Scholar 

  203. Craft, R. M. Modulation of pain by estrogens. Pain 132, S3–S12 (2007).

    Article  CAS  PubMed  Google Scholar 

  204. Da Silva, J. T., Zhang, Y., Asgar, J., Ro, J. Y. & Seminowicz, D. A. Diffuse noxious inhibitory controls and brain networks are modulated in a testosterone-dependent manner in Sprague Dawley rats. Behav. Brain Res. 2018, 91–97 (2018).

    Article  CAS  Google Scholar 

  205. Kasielska-Trojan, A., Stabryla, P. & Antoszewski, B. Digit ratio (2D:4D) and postoperative pain perception. Early Hum. Dev. 110, 25–30 (2017).

    Article  PubMed  Google Scholar 

  206. Bodnar, R. J., Commons, K. & Pfaff, D. W. Central Neural States Relating Sex and Pain (Johns Hopkins University Press, 2002).

  207. Stringer, S., Polderman, T. & Posthuma, D. Majority of human traits do not show evidence for sex-specific genetic and environmental effects. Sci. Rep. 7, 8688 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  208. Mayne, B. T. et al. Large scale gene expression meta-analysis reveals tissue-specific, sex-biased gene expression in humans. Front. Genet. 7, 183 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  209. Karp, N. A. et al. Prevalence of sexual dimorphism in mammalian phenotypic traits. Nat. Commun. 8, 15475 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Khramtsova, E. A., Davis, L. K. & Stranger, B. E. The role of sex in the genomics of human complex traits. Nat. Rev. Genet. 20, 173–190 (2019).

    Article  CAS  PubMed  Google Scholar 

  211. Clayton, J. A. & Collins, F. S. Policy: NIH to balance sex in cell and animal studies. Nature 509, 282–283 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  212. Hughes, R. N. Sex still matters: has the prevalence of male-only studies of drug effects on rodent behaviour changed during the past decade? Behav. Pharmacol. 30, 95–99 (2019).

    Article  PubMed  Google Scholar 

  213. Moseley, G. L. & Vlaeyen, J. W. S. Beyond nociception: the imprecision hypothesis of chronic pain. Pain 156, 35–38 (2015).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The author is supported by funding from the Canadian Institutes of Health Research, the Natural Sciences and Engineering Research Council of Canada and the Louise and Alan Edwards Foundation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jeffrey S. Mogil.

Ethics declarations

Competing interests

The author declares no competing interests.

Additional information

Data availability

The data that support the findings in the analyses are available from the author on request.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

related links

Canadian Institute of Health Research - How CIHR is supporting the integration of SGBA: https://cihr-irsc.gc.ca/e/50837.html

NIH Policy on Sex as a Biological Variable: https://orwh.od.nih.gov/sex-gender/nih-policy-sex-biological-variable

Supplementary information

Glossary

Quantitative trait locus

(QTL). The approximate genomic location of a gene variant responsible for a difference between strains, used to identify the gene responsible.

Candidate gene association study

A study of the potential role of a preidentified gene in the mediation of variability in a trait.

Pleiotropy

A situation in which a single gene influences two or more seemingly unrelated traits.

Genome-wide association studies

Studies looking for genetic association between thousands of known DNA variants and a trait of interest.

RNA sequencing

A technique that examines the quantity and sequences of RNA in a sample using next-generation sequencing.

Priming

A short-lasting hypersensitivity state that produces a long-lasting one on the introduction of a second noxious stimulus.

Microgliosis

A reactive change in the structure and function of microglia in response to damage to the CNS.

Schwann cell

A glial cell type in the peripheral nervous system that produce the myelin sheath around axons.

Organizational effects

Permanent effects of hormones on structure and function that occur during development.

Activational effects

Temporary effects of hormones on structure and function that depend on the presence or absence of the hormone.

X inactivation

The process by which one of the two copies of the X chromosome is inactivated in female mammals.

Four-core genotypes model

Strains of mice genetically engineered such that sex chromosome complement (XX vs XY) is unrelated to the animal’s gonadal sex.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mogil, J.S. Qualitative sex differences in pain processing: emerging evidence of a biased literature. Nat Rev Neurosci 21, 353–365 (2020). https://doi.org/10.1038/s41583-020-0310-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41583-020-0310-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing