Brazilian Journal of Anesthesiology
https://bjan-sba.org/article/doi/10.1590/S0034-70942011000600014
Brazilian Journal of Anesthesiology
Review Article

Sexo e percepção da dor e analgesia

Sex and Pain perception and analgesia

Cláudia Carneiro de Araújo Palmeira; Hazem Adel Ashmawi; Irimar de Paula Posso

Downloads: 2
Views: 869

Resumo

Sexo é um fator importante na modulação da experiência dolorosa. Um grande volume de evidências revela que a experiência dolorosa é diferente em machos e fêmeas, assim como a resposta a algumas classes de analgésicos. Inúmeros experimentos laboratoriais sugerem que as mulheres têm limiar doloroso menor que os homens em dor provocada por estímulos nociceptivos como calor, frio, pressão e estímulo elétrico. A dor é um fenômeno dinâmico que sofre influência de diversos mecanismos de controle excitatórios e inibitórios. Diferenças na percepção dolorosa relacionadas ao sexo podem estar associadas à hiperalgesia em mulheres, mas também à hipoatividade do sistema inibitório de dor no sexo feminino. O objetivo da presente revisão, que mostra algumas relações entre hormônios gonadais, sistema nervoso central e dor é prover pontos de referência para a discussão de um dos aspectos mais intrigantes da fisiopatologia da dor: a presença de diferenças no estímulo doloroso relacionadas ao sexo.

Palavras-chave

DOR, HORMÔNIOS

Abstract

Sex is an important factor in painful experience modulation. Large volume of evidence shows that experience is different for males and females, as well as the answer to some classes of analgesics. Laboratory experiments suggest that women have a lower pain threshold than men related to pain from noxious stimuli such as heat, cold, pressure and electrical stimulation. Pain is a dynamic phenomenon under the influence of various mechanisms of excitatory and inhibitory control. The differences in pain perception related to sex may be associated with hyperalgesia in women, but also to the hypoactivity of the inhibitory system of pain in females. The purpose of this review besides showing some relationship for gonadal hormones, central nervous system and pain is to provide reference points for the discussion of one of the most intriguing aspects of the pathophysiology of pain: the differences in the presence of painful stimuli related to gender.

Keywords

Sex Factors, Pain, Gonadal Steroid Hormones

References

Hurley RW, Adams MCB. Sex, gender and pain: an overview of a complex field. Anesth Analg. 2008;107:309-317.

Greenspan JD, Craft RM, LeResche L. Studying sex and gender differences in pain and analgesia: A consensus report. Pain. 2007;132:26-45.

Unruh AM. Gender variations in clinical pain experience. Pain. 1996;65:123-167.

Riley JL, Robinson ME, Wise EA. Sex differences in the perception of noxious experimental stimuli: a meta-analysis. Pain. 1998;74:181-187.

Aloisi AM, Ceccarelli I, Herdegen T. Gonadectomy and persistent pain differently affect hippocampal c-Fos expression in male and female rats. Neurosci Lett. 2006;281:29-32.

Turk DC, Meichenbaum DH, Berman WH. Application of biofeedback for the regulation of pain: a critical review. Psychol Bull. 1979;86:1322-1338.

McEwen BS. Permanence of brain sex differences and structural plasticity of the adult brain. Proc Natl Acad Sci USA. 1999;96:7128-7130.

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

Tousignant-Laflamme Y, Marchand S. Excitatory and inhibitory pain mechanisms during the menstrual cycle in healthy women. Pain. 2009;146:47-55.

Fillingim RB, Gear RW. Sex differences in opioid analgesia: clinical and experimental findings. Eur J Pain. 2004;8:413-425.

Gaumond I, Arsenault P, Marchand S. Specificity of female and male sex hormones on excitatory and inhibitory phases of formalin-induced nociceptive responses. Brain Res. 2005;1052:105-111.

Thompson AD, Angelotti T, Nag S. Sex-specific modulation of spinal nociception by alpha-adrenoceptors: differential regulation by estrogen and testosterone. Neuroscience. 2008;153:1268-1277.

Hagiwara H, Kimura F, Mitsushima D. Formalin-induced nociceptive behavior and c-Fos experssion in middle-aged female rats. Physiol Behav. 2010;100:101-104.

Berkley KJ. Sex differences in pain. Behav Brain Sci. 1997;20:371-380.

Fillingim RB, Gear RW. Sex differences in opioid analgesia: clinical and experimental findings. Eur J Pain. 2004;8:413-425.

Wolf f, Ross K, Anderson J. Aspects of fibromyalgia in the general population: sex, pain threshold, and fibromyalgia symptoms. J Rheumatol. 1995;22:151-156.

Keogh E, McCraken LM, Eccleston C. Do men and women differ in their response to interdisciplinary chronic pain management?. Pain. 2005;114:37-46.

Triadafilopoulos G, Finlayson M, Grellet C. Bowel dysfunction in postmenopausal women. Women Health. 1998;27:55-66.

Cervero F, Laird JM. Role of ion channels in mechanisms controlling gastrointestinal pain pathways. Curr Opin Pharmacol. 2003;3:608-612.

Howard FM. Chronic pelvic pain. Obstet Gynecol. 2003;101:594-611.

Vicent K. Chronic pelvic pain in women. Postgrad Med J. 2009;85:24-29.

Bingefors K, Isacson D. Epidemiology, co-morbidity, and impact on health-related quality of life of self-reported headache and musculoskeletal pain: a gender perspective. Eur J Pain. 2004;8:435-450.

Isacson D, Bingefors K. Epidemiology of analgesic use: a gender perspective. Eur J Anaesthesiol Suppl. 2002;26:5-15.

Brody S, Carlstrom K, Lagrelius A. Serum sex hormone binding globulin (SHBG), testosterone/SHBG index, endometrial pathology and bone mineral density in postmenopausal women. Acta Obstet Gynecol Scand. 1987;66:357-360.

Katzenellenbogen BS, Katzenellenbogen JA. Estrogen receptor transcription and transactivation, estrogen receptor alpha and estrogen receptor β: regulation by selective estrogen receptor modulators and importance in breast cancer. Breast Cancer Res. 2000;2:335-344.

Vasudevan N, Kia HK, Inoue S. Isoform specificity for oestrogen receptor and thyroid hormone receptor genes and their interactions on the NR2D gene promoter. J Neuroendocrinol. 2002;14:836-842.

Bai Y, Giguere V. Isoform-selective interactions between estrogen receptors and steroid receptor coactivators promoted by estradiol and ErbB-2 signaling in living cells. Mol Endocrinol. 2003;17:589-599.

Arnold AP, Breedlove SM. Organizational and activational effects of sex steroids on brain and behavior: a reanalysis. Horm Behav. 1985;19:469-498.

Micevych P, Sinchak K, Mills RH. The luteinizing hormone surge is preceded by an estrogen-induced increase of hypothalamic progesterone in ovariectomized and adrenalectomized rats. Neuroendocrinology. 2003;78:29-35.

Sinchak K, Mills RH, Tao L. Estrogen induces de novo progesterone synthesis in astrocytes. Dev Neurosci. 2003;25:343-348.

Amateu SK, Alt JJ, Stamps CL. Brain estradiol content in newborn rats: sex differences, regional heterogeneity, and possible de novo synthesis by the female telencephalon. Endocrinology. 2004;145:2906-2917.

Hojo Y, Hattori TA, Enami T. Adult male rat hippocampus synthesizes estradiol from pregnenolone by cytochromes P45017 alpha and P450 aromatase localized in neurons. Proc Natl Acad Sci USA. 2004;101:865-870.

Kretz O, Fester L, Wehrenberg U. Hippocampal synapses depend on hippocampal estrogen synthesis. J Neurosci. 2004;24:5913-5921.

Bennett HL, Gustafsson JA, Keast JR. Estrogen receptor expression in lumbosacral dorsal root ganglion cells innervating the female rat urinary bladder. Auton Neurosci. 2003;105:90-100.

Evrard HC. Estrogen synthesis in the spinal dorsal horn: a new central mechanism for the hormonal regulation of pain. Am J Physiol Regul Integr Comp Physiol. 2006;291:R291-299.

Muller-Lissner SA, Fumagalli I, Bardhan KD. Tagaserod, a 5HT(4) receptor partial agonist, relieves symptoms in irritable bowel syndrome patients with abdominal pain, bloating and constipation. Aliment Pharmacol Ther. 2001;15:1655-1666.

Simpson ER. Sources of estrogen and their importance. J Steroid Biochem Mol Biol. 2003;86:225-230.

Foster TC. Interaction of rapid signal transduction cascades and gene expression in mediating estrogen effects on memory over the life span. Front Neuroencocrinol. 2005;26:51-64.

Robichaud M, Debonnel D. Oestrogen and testosterone modulate the firing activity of dorsal raphe nucleus serotonergic neurones in both male and female rats. J Neuroendocrinol. 2005;17:179-185.

Paul SM, Purdy RH. Neuroactive Steroids. FASEB J. 1992;6:2311-2322.

Rupprecht R, Holsboer F. Neuroactive steroids: mechanisms of action and neuropsychopharmacological perspectives. Trends Neurosci. 1999;22:410-416.

Rupprecht R, di Michele F, Hermann B. Neuroactive steroids: molecular mechanisms of action and implications for neuropsychopharmacology. Brain Res Rev. 2001;37:59-67.

Teyler TJ, Vardaris RM, Lewis D. Gonadal steroids: effects on excitability of hippocampal pyramidal cells. Science. 1980;209:1017-1018.

Beyenburg S, Stoffel-Wagner B, Bauer J. Neuroactive steroids and seizure susceptibility. Epilepsy Res. 2001;44:141-153.

Shibuya K, Takata N, Hojo Y. Hippocampal cytochrome P450s synthesize brain neurosteroids which are paracrine neuromodulators of synaptic signal transduction. Biochim Biophys Acta. 2003;1619:301-316.

Mellon SH. Neurosteroids: biochemistry, modes of action, and clinical relevance. J Clin Endocrinol Metab. 1994;78:1003-1008.

Balthazart J, Ball GF. Fast regulation of steroid biosynthesis: a further piece in the neurosteroid puzzle. Trends Neurosci. 2000;23:57-58.

Lanlua P, Decorti F, Gangula PRR. Female steroid hormones modulate receptors for nerve growth factor in rat dorsal root ganglia. Biol Reprod. 2001;64:331-338.

Rudick CN, Woolley CS. Estradiol induces a phasic c-fos response in the hippocampal CA1 and CA3 regions of adult female rats. Hippo-campus. 2000;10:274-283.

Toran-Alleran CD, Singh M, Sétáló G Jr. Novel mechanisms of estrogen action in the brain: new players and an old story. Front Neuroendocrinol. 1999;20:97-121.

Foy MR, Xu J, Xie X. 17 beta-estradiol enhances NMDA receptor-mediated EPSPs and long-term potentiation. J Neurophysiol. 1999;81:925-929.

McEwen BS, Alves SE. Estrogen actions in the central nervous system. Endocr Rev. 1999;20:279-307.

Silberstein SD. Hormone related headache. Med Clin North Am. 2001;85:1017-1035.

Zuloaga DG, Puts DA, Jordan CL. The role of androgen receptors in the masculinization of brain and behavior: what we've learned from the testicular feminization mutation. Horm Behav. 2008;53:613-626.

Aloisi AM, Affaitati G, Ceccarelli I. Estradiol and testosterone differently affect visceral pain-related behavioural responses in male and female rats. Eur J Pain. 2010;14:602-607.

Affaitati G, Ceccarelli I, Fiorenzani P. Sex differences in the analgesic effects of ICI 182,780 and flutamide on ureteral calculosis in rats. Horm Behav. 2011;59(1):9-13.

Sanoja R, Cervero F. Estrogen-dependent changes in visceral afferent sensitivity. Auton Neurosci. 2010;153:84-89.

Sanoja R, Cervero F. Estrogen-dependent abdominal hyperalgesia induced by ovariectomy in adult mice: a model of functional abdominal pain. Pain. 2005;118:243-252.

Sanoja R, Cervero F. Estrogen modulation of ovariectomy-induced hyperalgesia in adult mice. Eur J Pain. 2008;12:573-581.

Dong DX, Mann MK, Kumar U. Sex-related differences in MNDA-evoked rat masseter muscle afferente discharge result from estrogen-mediated modulation of peripheral NMDA receptor activity. Neuroscience. 2007;146:822-832.

Tang B, Ji Y, Traub RJ. Estrogen alters spinal NMDA receptor activity via a PKA signaling pathway in a visceral pain model in the rat. Pain. 2008;137:540-549.

Dina OA, Gear EW, Messing RO. Severity of alcohol-induced painful peripheral neuropathy in female rats: role of estrogen and protein kinase (A and Ce). Neuroscience. 2007;145:350-356.

Ceccarelli I, Fiorezani P, Massafra C. Repeated nociceptive stimulation induces different behavioral and neuronal responses in intact and gonadectomized female rats. Brain Res. 2006;1106:142-149.

Mannino CA, South SM, Quinones-Jenab V. Estradiol replacement in ovariectomized rats is antihyperalgesic in formalin test. J Pain. 2007;8:334-342.

Nag S, Mokha SS. Testosterone is essential for alpha-2-adrenoceptor-induced antinociception in the trigeminal region of the male rat. Neurosci Lett. 2009;467:48-52.

Wu W, Bi Y, Kou X. 17-beta Estradiol enhaced allodynia if inflamatory temporomandibular joint through upregulation of hippocampal TRPV1 in ovariectomized rats. J Neurosci. 2010;26:8710-8719.

Stoffel EC, Ulibarri CM, Folk JE. Gonadal hormone modulation of mu, kappa and delta opioid antinociception in male and female rats. J Pain. 2005;6:261-274.

Mitrovic I, Mitrovic MM, Bader S. Contribuition of GIRK-2 mediated postsynaptic signaling to opiate and alpha -2 adrenergic analgesia and analgesic sex differences. Proc Natl Acad Sci USA. 2003;100:271-276.

Claiborne J, Nag S, Mokha SS. Activation of opioid receptor like-1 receptor in the spinal cord produces sex-specific antinociception in the rat: estrogen attenuates antinociception in the female, whereas testosterone is required for the expression of antinociception in the male. J Neurosci. 2006;26:13048-13053.

Li L, Fan X, Warner M. Ablation of estrogen receptor alpha or beta eliminates sex differences in mechanical pain threshold in norma and inflamed mice. Pain. 2009;143:37-40.

Zubieta JK, Smith YR, Bueller JA. mu-opioid receptor-mediated antinociceptive responses differ in men and women. J Neurosci. 2002;22:5100-5107.

Zubieta JK, Dannals RF, Frost JJ. Gender and age influences on human brain mu-opioid receptor binding measured by PET. Am J Psychiatry. 1999;156:842-848.

Wiesenfeld-Hallin Z. Sex differences in pain perception. Gender Med. 2005;2:137-145.

Cicero TJ, Nock B, OConnor L. Role of steroids in sex differences in morphine-induced analgesia: activational and organizational effects. J Pharmacol Exp Ther. 2002;300:695-701.

Krzanowska EK, Ogawa S, Pfaf DW. Reversal of sex differences in morphine analgesia elicited from the ventrolateral periaqueductal gray in rats by neonatal hormone manipulations. Brain Res. 2002;929:1-9.

Cook CD, Barret AC, Roach AL. Sex-related differences in the antinociceptive effects of opioids: importance of rat genotype, nociceptive stimulus intensity, and efficacy at the mu opioid receptor. Psycho-pharmacology. 2000;150:430-442.

Gear RW, Miaskowiski C, Gordon NC. The kappa opioid nalbuphine produces gender- and dose-dependent analgesia and antianalgesia in patients with postoperative pain. Pain. 1999;83:339-345.

Dahan A, Kest B, Waxman AR. Sex-specific responses to opiates: animal and human studies. Anesth Analg. 2008;107:83-95.

Kepler KL, Standifer KM, Paul D. Gender effects and central opioid analgesia. Pain. 1991;45:87-94.

Wise EA, Price DD, Myers CD. Gender role expectations of pain: relationship to experimental pain perception. Pain. 2002;96::335-342.

Jensen IB, Bergstrom G, Ljungquist T, Bodin L, Nygren AL. A randomized controlled component analysis of a behavioural medicine rehabilitation program for chronic spinal pain: are the effects dependent on gender?. Pain. 2001;91:65-78.

Krogstad BS, Jokstad A, Dahl BL. The reporting of pain, somatic complaints, and anxiety in a group of patients with TMD before and 2 years after treatment: sex differences. J Orofac Pain. 1996;10:263-269.

Hansen FR, Bendix T, Skov P. Intensive, dynamic back-muscle exercises, conventional physiotherapy, or placebo-control treatment of low-back-pain-a randomized, observer-blind trial. Spine. 1993;18:98-106.

Klatzkin RR, Mechlin B, Girdler SS. Menstrual cycle does not influence gender differences in experimental pain sensitivity. Eur J Pain. 2010;14:77-82.

Okifuji A, Turk DC. Sex hormones and pain in regularly menstruating women with fibromyalgia syndrome. J Pain. 2006;11:851-859.

Fillingim RB, Maxiner W. Gender differences in the responses to noxious stimuli. Pain Forum. 1995;4:221-308.

Chesterton LS, Barlas P, Foster NE. Gender differences in pressure pain threshold in healthy humans. Pain. 2003;101:259-266.

Kowalczyk WJ, Sullivan MA, Evans SM. Sex differences and hormonal influences on response to mechanical pressure pain in humans. J Pain. 2010;11:330-342.

Nisenblat V, Engel-Yeger B, Ohel G. The association between supra-physiological levels of estradiol and response patterns to experimental pain. Eur J Pain. 2010;14:840-846.

LeBars D, Dickenson AH, Besson JM. Diffuse noxious inhibitory controls (DNIC): Efffects on dorsal horn convergent neurons in the rat. Pain. 1979;6:283-304.

Edwards RR. Individual differences in endogenous pain modulation as a risk factor for chronic pain. Neurology. 2005;65:437-443.

Tousignant-Laflamme Y, Pagé S, Goffaux P. An experimental model to measure excitatory and inhibitory pain mechanisms in humans. Brain Res. 2008;1230:73-79.

France CR, Suchowiecki S. A comparison of diffuse noxious inhibitory controls in men and women. Pain. 1999;81:77-84.

Baad-Hansen L, Pousen HF, Jensen HM. Lack of sex differences in modulation of experimental intraoral pain by diffuse noxious inhibitory controls (DNIC). Pain. 2005;116:359-365.

Serrao M, Rossi P, Sandrini G. Effects of diffuse noxious inhibitory controls on temporal summation of the RIII reflex in humans. Pain. 2004;112:353-360.

Ge HY, Madeleine P, Arendt-Nielsen L. Sex differences in temporal characteristics of descending inhibitory control: an evaluation using repeated bilateral experimental induction of muscle pain. Pain. 2004;110:72-78.

Ge HY, Madeleine P, Arendt-Nielsen L. Gender differences in pain modulation evoked by repeated injections of glutamate into the human trapezius muscle. Pain. 2005;113:134-140.

Staud R, Robinson ME, Vierck Jr CJ. Diffuse noxious inhibitory controls (DNIC) attenuate temporal summation of second pain in normal males but not in normal females or fibromyalgia patients. Pain. 2003;101:167-174.

Cepeda MS, Carr DB. Women experience more pain and require more morphine than men to achieve a similar degree of analgesia. Anesth Analg. 2003;97:1464-1468.

Miller PL, Ernst AA. Sex differences in analgesia: a randomized trial of mu versus k opioid agonists. South Med J. 2004;97:35-41.

Gear RW, Miaskowisk C, Gordon NC. Kappa-opioids produce significantly greater analgesia in women than in men. Nat Med. 1996;2:1248-1250.

Gear RW, Gordon NC, Heller PH. Gender difference in analgesic response to the kappa-opioid pentazocine. Neurosci Lett. 1996;205:207-209.

Mogil JS, Wilson SG, Chesler EJ. The melanocortin-1 receptor gene mediates female-specific mechanisms of analgesia in mice and humans. Proc Natl Acad Sci USA. 2003;100:4867-4872.

Loyd DR, Murphy AZ. The role of the periaqueductal gray in the modulation of pain in males and females: are the anatomy and physiology really that different?. Neural Plast. 2009;2009.

Smith YR, Stohler CS, Nichols TE. Pronociceptive and antinociceptive effects of estradiol through endogenous opioid neurotransmission in women. J Neurosci. 2006;26:5777-57785.

5dd693900e88256c7713f286 rba Articles
Links & Downloads

Braz J Anesthesiol

Share this page
Page Sections