Testosterone signaling pathways for reducing neuropathic pain in a rat model of spinothalamic tract lesion

Document Type : Original Article

Authors

1 Department of Pharmacology, School of Medicine, Arak University of Medical Sciences, Arak, Iran

2 Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran

3 Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran

10.22038/ijbms.2024.78491.16968

Abstract

Objective(s): Most individuals who suffer from spinal cord injury (SCI) experience neuropathic pain, which currently has no effective treatment. In this study, we examined how testosterone affects neuropathic pain resulting from SCI. 
Materials and Methods: We administered three different doses of testosterone (4, 8, 16 mg/kg, intraperitoneal) to male rats after an electrolytic lesion of the spinothalamic tract. We then conducted behavioral tests, including open field and von Frey tests, within 28 days post-SCI. On day 28 after SCI, we analyzed spinal tissue using western blot to measure the levels of ionized calcium binding adaptor molecule 1 (Iba1), glial fibrillary acidic protein (GFAP), phospho-extracellular signal-regulated kinase (p-ERK1/2), and p-P38 at the injury site. 
Results: The results showed that testosterone significantly improved both motor activity and mechanical allodynia compared to the SCI-only group. Testosterone also inhibited microglia and astrocyte activation. Furthermore, testosterone significantly decreased p-P38 and p-ERK levels.  
Conclusion: The findings indicate that testosterone may alleviate SCI-induced neuropathic pain by inhibiting the activation of astrocytes and microglia, as well as suppressing MAPK signaling pathways.

Keywords

Main Subjects


1. Hearn JH, Cross A. Mindfulness for pain, depression, anxiety, and quality of life in people with spinal cord injury: A systematic review. BMC Neurol 2020; 20:1-11.
2. Watson JC, Sandroni P. Central neuropathic pain syndromes. Mayo Clin Proc 2016; 91:372-385.
3. Hari AR, Wydenkeller S, Dokladal P, Halder P. Enhanced recovery of human spinothalamic function is associated with central neuropathic pain after SCI. Exp Neurol 2009; 216:428-430.
4. Pfyffer D, Vallotton K, Curt A, Freund P. Tissue bridges predict neuropathic pain emergence after spinal cord injury. J Neurol Neurosurg Psychiatry 2020; 91:1111-1117.
5. Clark MJ, Schopp LH, Mazurek MO, Zaniletti I, Lammy AB, Martin TA, et al. Testosterone levels among men with spinal cord injury: relationship between time since injury and laboratory values. Am J Phys Med Rehabil 2008; 87:758-767.
6. Hvarness H, Jakobsen H, Biering-Sørensen F. Men with spinal cord injury have a smaller prostate than men without. Scand J Urol Nephrol 2007; 41:120-123.
7. Wang H, Zhou W-x, Huang J-f, Zheng X-q, Tian H-j, Wang B, et al. Endocrine therapy for the functional recovery of spinal cord injury. Front Neurol 2020; 14:590570.
8. Christensen MD, Hulsebosch CE. Chronic central pain after spinal cord injury. J Neurotrauma 1997; 14:517-537.
9. Durga A, Sepahpanah F, Regozzi M, Hastings J, Crane DA. Prevalence of testosterone deficiency after spinal cord injury. PM&R 2011; 3:929-932.
10. Cunningham G, Matsumoto A, Swerdloff R. Patient’s guide to low testosterone. Chevy Chase, MD: The Hormone Foundation 2003.
11. Hussain R, Ghoumari AM, Bielecki B, Steibel J, Boehm N, Liere P, et al. The neural androgen receptor: a therapeutic target for myelin repair in chronic demyelination. Brain 2013; 136:132-146.
12. Gürer B, Kertmen H, Kasim E, Yilmaz ER, Kanat BH, Sargon MF, et al. Neuroprotective effects of testosterone on ischemia/reperfusion injury of the rabbit spinal cord. Injury 2015; 46:240-248.
13. Fargo KN, Foecking EM, Jones KJ, Sengelaub DR. Neuroprotective actions of androgens on motoneurons. Front Neuroendocrinol 2009; 30:130-141.
14. Kato Y, Shigehara K, Kawaguchi S, Izumi K, Kadono Y, Mizokami A. Efficacy of testosterone replacement therapy on pain in hypogonadal men with chronic pain syndrome: A subanalysis of a prospective randomised controlled study in Japan (EARTH study). Andrologia 2020; 52:e13768.
15. Calabrese D, Giatti S, Romano S, Porretta-Serapiglia C, Bianchi R, Milanese M, et al. Diabetic neuropathic pain: a role for testosterone metabolites. J Endocrinol 2014; 221:1-13.
16. Tennant F, Lichota L. Testosterone replacement in chronic pain patients. Pract Pain Manag 2010; 10:12-15.
17. Ren K. An improved method for assessing mechanical allodynia in the rat. Physiol Behav 1999; 67:711-716.
18. Naseri K, Saghaei E, Abbaszadeh F, Afhami M, Haeri A, Rahimi F, et al. Role of microglia and astrocyte in central pain syndrome following electrolytic lesion at the spinothalamic tract in rats. J Mol Neurosci 2013; 49:470-479.
19. Masoudi A, Dargahi L, Abbaszadeh F, Pourgholami MH, Asgari A, Manoochehri M, et al. Neuroprotective effects of astaxanthin in a rat model of spinal cord injury. Behav Brain Res 2017; 329:104-110.
20. Burke D, Fullen BM, Stokes D, Lennon O. Neuropathic pain prevalence following spinal cord injury: A systematic review and meta‐analysis. Eur J Pain 2017; 21:29-44.
21. Wang G, Thompson SM. Maladaptive homeostatic plasticity in a rodent model of central pain syndrome: thalamic hyperexcitability after spinothalamic tract lesions. J Neurosci 2008; 28:11959-11969.
22. Naseri K, Saghaei E, Abbaszadeh F, Afhami M, Haeri A, Rahimi F, et al. Role of microglia and astrocyte in central pain syndrome following electrolytic lesion at the spinothalamic tract in rats. J Mol Neurosci 2013; 49:470-479.
23. Yoon DH, Kim YS, Young W. Therapeutic time window for methylprednisolone in spinal cord injured rat. Yonsei Med J 1999; 40:313-320.
24. Cadotte DW, Fehlings MG. Spinal cord injury: A systematic review of current treatment options. Clin Orthop Relat Res 2011; 469:732-741.
25. Schaeffer V, Meyer L, Patte-Mensah C, Mensah-Nyagan AG. Progress in dorsal root ganglion neurosteroidogenic activity: Basic evidence and pathophysiological correlation. Prog Neurobiol 2010; 92:33-41.
26. Coronel MF, Labombarda F, González SL. Neuroactive steroids, nociception and neuropathic pain: A flashback to go forward. Steroids 2016; 110:77-87.
27. Brotfain E, E Gruenbaum S, Boyko M, Kutz R, Zlotnik A, Klein M. Neuroprotection by estrogen and progesterone in traumatic brain injury and spinal cord injury. Curr Neuropharmacol 2016; 14:641-653.
28. Elkabes S, Nicot AB. Sex steroids and neuroprotection in spinal cord injury: A review of preclinical investigations. Exp Neurol 2014; 259:28-37.
29. Clark MJ, Petroski GF, Mazurek MO, Hagglund KJ, Sherman AK, Lammy AB, et al. Testosterone replacement therapy and motor function in men with spinal cord injury: A retrospective analysis. Am J Phys Med Rehabil 2008; 87:281-284.
30. Choi JC, Park YH, Park SK, Lee JS, Kim J, Choi JI, et al. Testosterone effects on pain and brain activation patterns. Acta Anaesthesiol Scand 2017; 61:668-675.
31. Hains BC, Waxman SG. Activated microglia contribute to the maintenance of chronic pain after spinal cord injury. J Neurosci 2006; 26:4308-4317.
32. Hulsebosch CE. Gliopathy ensures persistent inflammation and chronic pain after spinal cord injury. Exp Neurol 2008; 214:6-9.
33. Ji R-R, Woolf CJ. Neuronal plasticity and signal transduction in nociceptive neurons: implications for the initiation and maintenance of pathological pain. Neurobiol Dis 2001; 8:1-10.
34. Ji RR, Gereau RWt, Malcangio M, Strichartz GR. MAP kinase and pain. Brain Res Rev 2009; 60:135-148.
35. Choi DC, Lee JY, Lim EJ, Baik HH, Oh TH, Yune TY. Inhibition of ROS-induced p38MAPK and ERK activation in microglia by acupuncture relieves neuropathic pain after spinal cord injury in rats. Exp Neurol 2012; 236:268-282.
36. Byers JS, Huguenard AL, Kuruppu D, Liu NK, Xu XM, Sengelaub DR. Neuroprotective effects of testosterone on motoneuron and muscle morphology following spinal cord injury. J Comp Neurol 2012; 520:2683-2696.
37. Lee JY, Choi HY, Ju B-G, Yune TY. Estrogen alleviates neuropathic pain induced after spinal cord injury by inhibiting microglia and astrocyte activation. Biochim Biophys Acta 2018; 1864:2472-2480.
38. Saghaei E, Abbaszadeh F, Naseri K, Ghorbanpoor S, Afhami M, Haeri A, et al. Estradiol attenuates spinal cord injury-induced pain by suppressing microglial activation in thalamic VPL nuclei of rats. Neurosci Res 2013; 75:316-323.
39. Naderi A, Asgari AR, Zahed R, Ghanbari A, Samandari R, Jorjani M. Estradiol attenuates spinal cord injury-related central pain by decreasing glutamate levels in thalamic VPL nucleus in male rats. Metab Brain Dis 2014; 29:763-770.
40. Coers S, Tanzer L, Jones KJ. Testosterone treatment attenuates the effects of facial nerve transection on glial fibrillary acidic protein (GFAP) levels in the hamster facial motor nucleus. Metab Brain Dis 2002; 17:55-63.
41. Day JR, Frank AT, O’Callaghan JP, Jones BC, Anderson JE. The effect of age and testosterone on the expression of glial fibrillary acidic protein in the rat cerebellum. Exp Neurol 1998; 151:343-346.
42. Ivanova T, Méndez P, Garcia‐Segura LM, Beyer C. Rapid stimulation of the PI3‐kinase/Akt signalling pathway in developing midbrain neurones by oestrogen. J Neuroendocrinol 2002; 14:73-79.
43. Honda K, Sawada H, Kihara T, Urushitani M, Nakamizo T, Akaike A, et al. Phosphatidylinositol 3‐kinase mediates neuroprotection by estrogen in cultured cortical neurons. J Neurosci Res 2000; 60:321-327.
44. Cooke BM, Breedlove SM, Jordan CL. Both estrogen receptors and androgen receptors contribute to testosterone-induced changes in the morphology of the medial amygdala and sexual arousal in male rats. Horm Behav 2003; 43:336-346.
45. Nguyen TVV, Yao M, Pike CJ. Androgens activate mitogen‐activated protein kinase signaling: Role in neuroprotection. J Neurochem 2005; 94:1639-1651.
46. Hammond J, Le Q, Goodyer C, Gelfand M, Trifiro M, LeBlanc A. Testosterone‐mediated neuroprotection through the androgen receptor in human primary neurons. J Neurochem 2001; 77:1319-1326.
47. Ray SK, Samantaray S, Smith JA, Matzelle DD, Das A, Banik NL. Inhibition of cysteine proteases in acute and chronic spinal cord injury. Neurotherapeutics 2011; 8:180-186.
48. Dominguez R, Jalali C, de Lacalle S. Morphological effects of estrogen on cholinergic neurons in vitro involves activation of extracellular signal-regulated kinases. J Neurosci 2004; 24:982-990.
49. Aronson D, Violan MA, Dufresne SD, Zangen D, Fielding RA, Goodyear LJ. Exercise stimulates the mitogen-activated protein kinase pathway in human skeletal muscle. J Clin Invest 1997; 99:1251-1257.
50. Garcia-Ovejero D, Azcoitia I, Doncarlos LL, Melcangi RC, Garcia-Segura LM. Glia-neuron crosstalk in the neuroprotective mechanisms of sex steroid hormones. Brain Res Rev 2005; 48:273-286.
51. Wilson ME, Liu Y, Wise PM. Estradiol enhances Akt activation in cortical explant cultures following neuronal injury. Mol Brain Res 2002; 102:48-54.
52. Chen Z, Gibson TB, Robinson F, Silvestro L, Pearson G, Xu B-e, et al. MAP kinases. Chem Rev 2001; 101:2449-2476.
53. Sweatt JD. The neuronal MAP kinase cascade: A biochemical signal integration system subserving synaptic plasticity and memory. J Neurochem 2001; 76:1-10.
54. Carrier N, Kabbaj M. Extracellular signal-regulated kinase 2 signaling in the hippocampal dentate gyrus mediates the antidepressant effects of testosterone. Biol Psychiatry 2012; 71:642-651.