Skip to main content

The incidence of persistent, episodic, or chronic visceral pain disorders such as irritable bowel syndrome and other “functional” syndromes (fibromyalgia, interstitial cystitis, chronic pelvic pain and others) are more prevalent in females. Defining the site(s) and mechanisms through which sex steroid hormones modulate visceral nociception is an important step in understanding the gender differences in pain perception. One such mechanism may be the convergence of nociceptive stimuli and estrogen input on the primary afferent neurons, which innervate viscera. In our study we determined how estrogen interferes with pain transmission at the level of primary sensory neurons. Based on our results, it is likely that estrogen receptors expressed in primary afferent neurons located in the dorsal root ganglion modulate purinergic and opiate receptors-mediating chemical signaling associated with nociception. Nociception is a balance of pro- and anti-nociceptive inputs that is subject to regulation depending on the state of the organism. The modulation of purinergic receptor- mediated increase in intracellular calcium concentration and attenuation of opioid receptors functions by estrogen observed in primary afferent neurons strongly suggest that estrogen modulates visceral pain processing peripherally. Moreover, estrogen appears to have different actions on nociceptive signaling depending on the input acting as anti-nociceptive by inhibiting purinergic receptors-mediating response but also interferes with anti-nociceptive actions of opioids.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Berkley KJ. Sex differences in pain. Behav Brain Sci 1997; 20(3):371–380; discussion 435–513.

    PubMed  CAS  Google Scholar 

  2. Lee OY, Mayer EA, Schmulson M, Chang L, et al. Gender-related differences in IBS symptoms. Am J Gastroenterol 2001; 96(7):2184–2193.

    Article  PubMed  CAS  Google Scholar 

  3. Toner BB, Akman D. Gender role and irritable bowel syndrome: literature review and hypothesis. Am J Gastroenterol 2000; 95(1):11–16.

    Article  PubMed  CAS  Google Scholar 

  4. Mayer EA, Naliboff BD, Chang L. Basic pathophysiologic mechanisms in irritable bowel syndrome. Dig Dis 2001; 19(3):212–218.

    Article  PubMed  CAS  Google Scholar 

  5. Mayer EA, Naliboff BD, Chang L, et al. V. Stress and irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol 2001; 280(4):G519–G524.

    PubMed  CAS  Google Scholar 

  6. Fillingim RB, Ness TJ. Sex-related hormonal influences on pain and analgesic responses. Neurosci Biobehav Rev 2000; 24(4):485–501.

    Article  PubMed  CAS  Google Scholar 

  7. Mogil JS, Chesler EJ, Wilson SG, et al. Sex differences in thermal nociception and morphine antinociception in rodents depend on genotype. Neurosci Biobehav Rev 2000; 24(3):375–389.

    Article  PubMed  CAS  Google Scholar 

  8. Azpiroz F, Dapoigny M, Pace F, et al. Nongastrointestinal disorders in the irritable bowel syndrome. Digestion 2000; 62(1):66–72.

    Article  PubMed  CAS  Google Scholar 

  9. Riley JL, 3rd, Robinson ME, Wise EA, et al. Sex differences in the perception of noxious experimental stimuli: a meta-analysis. Pain 1998; 74(2–3):181–187.

    Article  PubMed  Google Scholar 

  10. Burnstock G. Purine-mediated signalling in pain and visceral perception. Trends Pharmacol Sci 2001; 22(4):182–188.

    Article  PubMed  CAS  Google Scholar 

  11. Burnstock G. P2X receptors in sensory neurones. Br J Anaesth 2000; 84(4):476–488.

    PubMed  CAS  Google Scholar 

  12. Matsuka Y, Neubert JK, Maidment NT, et al. Concurrent release of ATP and substance P within guinea pig trigeminal ganglia in vivo. Brain Res 2001; 915(2):248–255.

    Article  PubMed  CAS  Google Scholar 

  13. McRoberts JA, Coutinho SV, Marvizon JC, et al., Role of peripheral N-methyl-D-aspartate (NMDA) receptors in visceral nociception in rats. Gastroenterology 2001; 120(7):1737–1748.

    Article  PubMed  CAS  Google Scholar 

  14. Papka RE, Storey-Workley M. Estrogen receptor-alpha and -beta coexist in a subpopulation of sensory neurons of female rat dorsal root ganglia. Neurosci Lett 2002; 319(2):71–74.

    Article  PubMed  CAS  Google Scholar 

  15. Chaban V, Mayer E, Ennes H, Micevych P. Rapid effect of estrogen on ATP-induced [Ca2+]i signaling in cultured DRG neurons from wild type and ERKO mice. Gastroenterology 2003; 124(4):A1.

    Article  Google Scholar 

  16. Sapsed-Byrne S, Ma D, Ridout D, Holdcroft A. Estrous cycle phase variations in visceromotor and cardiovascular responses to colonic distension in the anesthetized rat. Brain Res 1996; 742(1–2):10–16.

    Article  PubMed  CAS  Google Scholar 

  17. Traub RJ, Murphy A. Colonic inflammation induces fos expression in the thoracolumbar spinal cord increasing activity in the spinoparabrachial pathway. Pain 2002; 95(1–2):93–102.

    Article  PubMed  CAS  Google Scholar 

  18. Sand P. Chronic pain syndromes of gynecologic origin. J. Reprod. Med. 2004; 49:230–234.

    PubMed  Google Scholar 

  19. Chaban VV, Mayer EA, Ennes HS, Micevych PE. Estradiol inhibits ATP-induced intracellular calcium concentration increase in dorsal root ganglia neurons. Neuroscience 2003; 118(4):941–948.

    Article  PubMed  CAS  Google Scholar 

  20. Aloisi AM, Ceccarelli I, Herdegen T. Gonadectomy and persistent pain differently affect hippocampal c-Fos expression in male and female rats. Neurosci Lett 2000; 281(1):29–32.

    Article  PubMed  CAS  Google Scholar 

  21. Nadal A, Diaz M, Valverde MA. The estrogen trinity: membrane, cytosolic, and nuclear effects. News Physiol Sci 2001; 16:251–255.

    PubMed  CAS  Google Scholar 

  22. Mermelstein PG, Becker JB, Surmeier DJ. Estradiol reduces calcium currents in rat neostriatal neurons via a membrane receptor. J Neurosci 1996; 16(2):595–604.

    PubMed  CAS  Google Scholar 

  23. Kelly MJ, Wagner EJ. Estrogen modulation of G-protein-coupled receptors. Trends Endocrinol Metab 1999; 10(9):369–374.

    Article  PubMed  CAS  Google Scholar 

  24. Eckersell CB, Popper P, Micevych PE. Estrogen-induced alteration of mu-opioid receptor immunoreactivity in the medial preoptic nucleus and medial amygdala. J Neurosci 1998; 18(10):3967–3976.

    PubMed  CAS  Google Scholar 

  25. Razandi M, Pedram A, Greene GL, Levin ER. Cell membrane and nuclear estrogen receptors (ERs) originate from a single transcript: studies of ERalpha and ERbeta expressed in Chinese hamster ovary cells. Mol Endocrinol 1999; 13(2):307–319.

    Article  PubMed  CAS  Google Scholar 

  26. Amandusson A, et al. Estrogen-induced alterations of spinal cord enkephalin gene expression. Pain 1999; 83(2):243–248.

    Article  PubMed  CAS  Google Scholar 

  27. Williams SJ, Papka RE. Estrogen receptor-immunoreactive neurons are present in the female rat lumbosacral spinal cord. J Neurosci Res 1996; 46(4):492–501.

    Article  PubMed  CAS  Google Scholar 

  28. Taleghany N, Sarajari S, DonCarlos LL, et al. Differential expression of estrogen receptor alpha and beta in rat dorsal root ganglion neurons. J Neurosci Res 1999; 57(5):603–615.

    Article  PubMed  CAS  Google Scholar 

  29. Papka RE, Storey-Workley M, Shughrue PJ, et al. Estrogen receptor-alpha and beta- immunoreactivity and mRNA in neurons of sensory and autonomic ganglia and spinal cord. Cell Tissue Res 2001; 304(2):193–214.

    Article  PubMed  CAS  Google Scholar 

  30. Lee DY, Chai YG, Lee EB, Kim KW, et al. 17Beta-estradiol inhibits high-voltage-activated calcium channel currents in rat sensory neurons via a non-genomic mechanism. Life Sci 2002; 70(17):2047–2059.

    Article  PubMed  CAS  Google Scholar 

  31. Dunn PM, Zhong Y, Burnstock G. P2X receptors in peripheral neurons. Prog Neurobiol 2001; 65(2):107–134.

    Article  PubMed  CAS  Google Scholar 

  32. Koshimizu TA, Van Goor F, Tomic M, et al. Characterization of calcium signaling by purinergic receptor-channels expressed in excitable cells. Mol Pharmacol 2000; 58(5):936–945.

    PubMed  CAS  Google Scholar 

  33. Bodin P, Burnstock G. Purinergic signalling: ATP release. Neurochem Res 2001; 26(8–9):959–969.

    Article  PubMed  CAS  Google Scholar 

  34. Ueno S, Tsuda M, Iwanaga T, Inoue K. Cell type-specific ATP-activated responses in rat dorsal root ganglion neurons. Br J Pharmacol 1999; 126(2):429–436.

    Article  PubMed  CAS  Google Scholar 

  35. Chen CC, Akopian AN, Sivilotti L, et al. A P2X purinoceptor expressed by a subset of sensory neurons. Nature 1995; 377(6548):428–431.

    Article  PubMed  CAS  Google Scholar 

  36. Cockayne DA, Hamilton SG, Zhu QM, Dunn PM, et al. Urinary bladder hyporeflexia and reduced pain-related behaviour in P2X3-deficient mice. Nature 2000; 407(6807):1011–1015.

    Article  PubMed  CAS  Google Scholar 

  37. Zhong Y, Dunn PM, Bardini M, et al. Changes in P2X receptor responses of sensory neurons from P2X3-deficient mice. Eur J Neurosci 2001; 14(11):1784–1792.

    Article  PubMed  CAS  Google Scholar 

  38. Xu GY, Huang LY. Peripheral inflammation sensitizes P2X receptor-mediated responses in rat dorsal root ganglion neurons. J Neurosci 2002; 22(1):93–102.

    PubMed  CAS  Google Scholar 

  39. Chaban VV, McRoberts JA, Ennes HS, Mayer EA. Nitric oxide synthase inhibitors enhance mechanosensitive Ca(2+) influx in cultured dorsal root ganglion neurons. Brain Res 2001; 903(1–2):74–85.

    Article  PubMed  CAS  Google Scholar 

  40. Levin ER. Cell localization, physiology, and nongenomic actions of estrogen receptors. J Appl Physiol 2001; 91(4):1860–1867.

    PubMed  CAS  Google Scholar 

  41. Norman AW, Mizwicki MT, Norman DP. Steroid-hormone rapid actions, membrane receptors and a conformational ensemble model. Nat Rev Drug Discov 2004; 3(1):27–41.

    Article  PubMed  CAS  Google Scholar 

  42. Rusin KI, Moises HC. Mu-Opioid receptor activation reduces multiple components of high-threshold calcium current in rat sensory neurons. J Neurosci 1995; 15(6):4315–4327.

    PubMed  CAS  Google Scholar 

  43. Kim YJ, Hur EM, Park TJ, Kim KT Nongenomic inhibition of catecholamine secretion by 17beta-estradiol in PC12 cells. J Neurochem 2000; 74(6):2490–2496.

    Article  PubMed  CAS  Google Scholar 

  44. Kurata K, Takebayashi M, Kagaya A, et al. Effect of beta-estradiol on voltage-gated Ca(2+) channels in rat hippocampal neurons: a comparison with dehydroepiandrosterone. Eur J Pharmacol 2001; 416(3):203–212.

    Article  PubMed  CAS  Google Scholar 

  45. Chaban VV, Micevych PE. Estrogen receptor-alpha mediates estradiol attenuation of ATP-induced Ca2 + signaling in mouse dorsal root ganglion neurons. J Neurosci Res 2005; 81(1):31–37.

    Article  PubMed  CAS  Google Scholar 

  46. Micevych PE, Eckersell CB, Brecha N, Holland KL. Estrogen modulation of opioid and cholecystokinin systems in the limbic-hypothalamic circuit. Brain Res Bull 1997; 44(4):335–343.

    Article  PubMed  CAS  Google Scholar 

  47. Micevych P, Sinchak K. Estrogen and endogenous opioids regulate CCK in reproductive circuits. Peptides 2001; 22(8):1235–1244.

    Article  PubMed  CAS  Google Scholar 

  48. Sinchak K, Micevych P. Visualizing activation of opioid circuits by internalization of G protein-coupled receptors. Mol. Neurobiol. 2003; 27(2):197–222.

    Article  PubMed  CAS  Google Scholar 

  49. Priest CA, Vink KL, Micevych PE. Temporal regulation by estrogen of beta-preprotachykinin mRNA expression in the rat ventromedial nucleus of the hypothalamus. Brain Res Mol Brain Res 1995; 28(1):61–71.

    Article  PubMed  CAS  Google Scholar 

  50. Holland KL, Popper P, Micevych PE. Infusion of CCK-A receptor mRNA antisense oligodeoxynucleotides inhibits lordosis behavior. Physiol Behav 1997; 62(3):537–543.

    Article  PubMed  CAS  Google Scholar 

  51. Flores CA, et al. Orphanin FQ produces gender-specific modulation of trigeminal nociception: behavioral and electrophysiological observations. Neuroscience 2001; 105(2):489–498.

    Article  PubMed  CAS  Google Scholar 

  52. Sandner-Kiesling A and Eisenach JC. Estrogen reduces efficacy of mu- but Not kappa-opioid agonist inhibition in response to uterine cervical distension. Anesthesiology 2002; 96(2):375–379.

    Article  PubMed  CAS  Google Scholar 

  53. Miaskowski C. Women and pain. Crit Care Nurs Clin North Am 1997; 9(4): 453–458.

    PubMed  CAS  Google Scholar 

  54. Zubieta JK, Smith YR, Bueller JA, et al. Mu-opioid receptor-mediated antinociceptive responses differ in men and women. J Neurosci 2002; 22(12):5100–5107.

    PubMed  CAS  Google Scholar 

  55. Maggi R, Ma ZQ, Pimpinelli F, et al. Decrease of the number of opioid receptors and of the responsiveness to morphineduring neuronal differentiation induced by 17beta-estradiol in estrogen receptor-transfected neuroblastoma cells (SK-ER3). Neuroendocrinology 1999; 69(1):54–62.

    Article  PubMed  CAS  Google Scholar 

  56. Lagrange AH, Ronnekleiv OK, Kelly MJ. Modulation of G protein-coupled receptors by an estrogen receptor that activates protein kinase A. Mol Pharmacol 1997; 51(4):605–612.

    PubMed  CAS  Google Scholar 

  57. Dai Y, Fukuoka T, Wang H, et al. Contribution of sensitized P2X receptors in inflamed tissue to the mechanical hypersensitivity revealed by phosphorylated ERK in DRG neurons. Pain 2004; 108(3):258–266.

    Article  PubMed  CAS  Google Scholar 

  58. Gold MS, Levine JD, Correa AM. Modulation of TTX-R INa by PKC and PKA and their role in PGE2-induced sensitization of rat sensory neurons in vitro. J Neurosci 1998; 18(24):10345–10355.

    PubMed  CAS  Google Scholar 

  59. Wildman SS, King BF, Burnstock G. Potentiation of ATP-responses at a recombinant P2×2 receptor by neurotransmitters and related substances. Br J Pharmacol 1997; 120(2):221–224.

    Article  PubMed  CAS  Google Scholar 

  60. Stoop R, Surprenant A, North RA. Different sensitivities to pH of ATP-induced currents at four cloned P2X receptors. J Neurophysiol 1997; 78(4):1837–1840.

    PubMed  CAS  Google Scholar 

  61. Woolf CJ, Doubell TP. The pathophysiology of chronic pain–increased sensitivity to low threshold A beta-fibre inputs. Curr Opin Neurobiol 1994; 4(4):525–534.

    Article  PubMed  CAS  Google Scholar 

  62. Reichling DB, Levine JD. The primary afferent nociceptor as pattern generator. Pain 1999; Suppl 6:S103–S109.

    Article  PubMed  Google Scholar 

  63. Chaban V, Christensen A, Wakamatsu M, et al. The same dorsal root ganglion neurons innervate uterus and colon in the rat. Neuroreport 2007; 18(3):209–212.

    Article  PubMed  Google Scholar 

  64. Al-Chaer ED, Traub RJ. Biological basis of visceral pain: recent developments. Pain 2002; 96(3):221–225.

    Article  PubMed  Google Scholar 

  65. Martinez-Gomez M, Cruz Y, Salas M, et al. Assessing pain threshold in the rat: changes with estrus and time of day. Physiol Behav 1994; 55(4):651–657.

    Article  PubMed  CAS  Google Scholar 

  66. Giamberardino MA, Affaitati G, Valente R, et al. Changes in visceral pain reactivity as a function of estrous cycle in female rats with artificial ureteral calculosis. Brain Res 1997; 774(1–2):234–238.

    Article  PubMed  CAS  Google Scholar 

  67. Bradshaw HB, Berkley KJ. Estrogen replacement reverses ovariectomy-induced vaginal hyperalgesia in the rat. Maturitas 2002; 41(2):157–165.

    Article  PubMed  CAS  Google Scholar 

  68. Holdcroft A. Hormones and the gut. Br J Anaesth 2000; 85(1):58–68.

    Article  PubMed  CAS  Google Scholar 

  69. Bereiter DA. Sex differences in brainstem neural activation after injury to the TMJ region. Cells Tissues Organs 2001; 169(3):226–237.

    Article  PubMed  CAS  Google Scholar 

  70. Fillingim RB, Edwards RR. The association of hormone replacement therapy with experimental pain responses in postmenopausal women. Pain 2001; 92(1–2):229–234.

    Article  PubMed  CAS  Google Scholar 

  71. Gschossmann JM, Chaban VV, McRoberts JA, et al. Mechanical activation of dorsal root ganglion cells in vitro: comparison with capsaicin and modulation by kappa-opioids. Brain Res 2000; 856(1–2):101–110.

    Article  PubMed  CAS  Google Scholar 

  72. Papka RE, Mowa CN. Estrogen receptors in the spinal cord, sensory ganglia, and pelvic autonomic ganglia. Int Rev Cytol 2003; 231:91–127.

    Article  PubMed  CAS  Google Scholar 

  73. Bahr R, Blumberg H, Janig W. Do dichotomizing afferent fibers exist which supply visceral organs as well as somatic structures? A contribution to the problem or referred pain. Neurosci Lett 1981; 24(1):25–28.

    Article  PubMed  CAS  Google Scholar 

  74. Devor M, Obermayer M. Membrane differentiation in rat dorsal root ganglia and possible consequences for back pain. Neurosci Lett 1984; 51(3):341–346.

    Article  PubMed  CAS  Google Scholar 

  75. Pierau FK, Fellmer G, Taylor DC. Somato-visceral convergence in cat dorsal root ganglion neurones demonstrated by double-labelling with fluorescent tracers. Brain Res 1984; 321(1):63–70.

    Article  PubMed  CAS  Google Scholar 

  76. Habler HJ, Janig W, Koltzenburg M. Dichotomizing unmyelinated afferents supplying pelvic viscera and perineum are rare in the sacral segments of the cat. Neurosci Lett 1988; 94(1–2):119–124.

    Article  PubMed  CAS  Google Scholar 

  77. Gold MS, Dastmalchi S, Levine JD. Co-expression of nociceptor properties in dorsal root ganglion neurons from the adult rat in vitro. Neuroscience 1996; 71(1):265–275.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2008 Springer Science + Business Media, B.V

About this chapter

Cite this chapter

Chaban, V.V. (2008). Estrogen Modulation of Visceral Nociception. In: Ritsner, M.S., Weizman, A. (eds) Neuroactive Steroids in Brain Function, Behavior and Neuropsychiatric Disorders. Springer, Dordrecht. https://doi.org/10.1007/978-1-4020-6854-6_4

Download citation

  • DOI: https://doi.org/10.1007/978-1-4020-6854-6_4

  • Publisher Name: Springer, Dordrecht

  • Print ISBN: 978-1-4020-6853-9

  • Online ISBN: 978-1-4020-6854-6

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics