Articles

Effect of Progesterone Therapy on TNF-α and iNOS Gene Expression in Spinal Cord Injury Model

Abstract

Spinal cord injury (SCI) as a destructive crash result in neurons degeneration. The SCI lead to the onset of biochemical and molecular cascades such as inflammation that in turn has a key role in neurodegeneration development. The previous studies demonstrated the role of TNF-α and iNOS genes in intensifying the process after SCI. As a consequence, these genes overexpression intensify the inflammation and neuron degeneration process. In the present study, 32 male Wistar rats were chased and divided into four groups of eight. The SCI were induced in three groups and another group used as a sham. The progesterone hormone used as a therapeutic agent in rats with SCI. The results showed that injection of 10 μg/kg/12h progesterone hormone reduced the TNF-α and iNOS gene expression significantly and confirmed the role of progesterone in the reduction of inflammation. Also, the numbers of intact neurons in progesterone group were higher than other groups that demonstrated the protective effects of progesterone on neuron death. The BBB test was performed and demonstrated that progesterone is an effective factor to the improvement of locomotor response. These results of the study confirmed the anti-inflammatory activity of progesterone hormone and suggested that it can be used as a therapeutic factor for SCI.

Balentine J. Pathology of experimental spinal cord trauma. I. The necrotic lesion as a function of vascular injury. Laboratory investigation; a journal of technical methods and pathology. 1978;39(3):236-53.

Cortez SC, McIntosh T, Noble L. Experimental fluid percussion brain injury: vascular disruption and neuronal and glial alterations. Brain research. 1989;482(2):271-82.

Priestley J, Ramer M, King V, et al. Stimulating regeneration in the damaged spinal cord. Journal of Physiology-Paris. 2002;96(1):123-33.

Tator CH. Update on the pathophysiology and pathology of acute spinal cord injury. Brain Pathology. 1995;5(4):407-13.

McIlwain DL, Hoke VB. The role of the cytoskeleton in cell body enlargement, increased nuclear eccentricity and chromatolysis in axotomized spinal motor neurons. BMC neuroscience. 2005;6(1):19.

Beattie MS, Farooqui AA, BRESNAHAN JC. Review of current evidence for apoptosis after spinal cord injury. Journal of neurotrauma. 2000;17(10):915-25.

Amar AP, Levy ML. Pathogenesis and pharmacological strategies for mitigating secondary damage in acute spinal cord injury. Neurosurgery. 1999;44(5):1027-39.

D'Souza S, Alinauskas K, McCrea E, et al. Differential susceptibility of human CNS-derived cell populations to TNF-dependent and independent immune-mediated injury. The Journal of neuroscience. 1995;15(11):7293-300.

Sipe KJ, Srisawasdi D, Dantzer R, et al. An endogenous 55 kDa TNF receptor mediates cell death in a neural cell line. Molecular brain research. 1996;38(2):222-32.

BETHEA JR, NAGASHIMA H, ACOSTA MC, et al. Systemically administered interleukin-10 reduces tumor necrosis factor-alpha production and significantly improves functional recovery following traumatic spinal cord injury in rats. Journal of neurotrauma. 1999;16(10):851-63.

Lee YB, Yune TY, Baik SY, et al. Role of tumor necrosis factor-α in neuronal and glial apoptosis after spinal cord injury. Experimental neurology. 2000;166(1):190-5.

Beckman JS, Koppenol WH. Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and ugly. American Journal of Physiology-Cell Physiology. 1996;271(5):C1424-C37.

Brosnan CF, Lee SC, Liu J. Regulation of inducible nitric oxide synthase expression in human glia: implications for inflammatory central nervous system diseases. Biochemical Society transactions. 1997;25(2):679-83.

Coyle P. The neuroimmunology of multiple sclerosis. Advances in neuroimmunology. 1996;6(2):143-54.

Parkinson JF, Mitrovic B, Merrill J. The role of nitric oxide in multiple sclerosis. Journal of molecular medicine. 1997;75(3):174-86.

Yune TY, Chang MJ, Kim SJ, et al. Increased production of tumor necrosis factor-α induces apoptosis after traumatic spinal cord injury in rats. Journal of neurotrauma. 2003;20(2):207-19.

Wada K, Chatzipanteli K, Busto R, et al. Role of nitric oxide in traumatic brain injury in the rat. Journal of neurosurgery. 1998;89(5):807-18.

De Nicola AF. Steroid hormones and neuronal regeneration. Advances in neurology. 1993;59:199.

Schumacher M, Akwa Y, Guennoun R, et al. Steroid synthesis and metabolism in the nervous system: trophic and protective effects. Journal of neurocytology. 2000;29(5-6):307-26.

Garcia-Segura LM, Azcoitia I, DonCarlos LL. Neuroprotection by estradiol. Progress in neurobiology. 2001;63(1):29-60.

Stein DG. Brain damage, sex hormones and recovery: a new role for progesterone and estrogen? Trends in neurosciences. 2001;24(7):386-91.

Thomas AJ, Nockels RP, Pan HQ, et al. Progesterone is neuroprotective after acute experimental spinal cord trauma in rats. Spine. 1999;24(20):2134.

Roof RL, Hoffman SW, Stein DG. Progesterone protects against lipid peroxidation following traumatic brain injury in rats. Molecular and Chemical Neuropathology. 1997;31(1):1-11.

Ogata T, Nakamura Y, Tsuji K, et al. Steroid hormones protect spinal cord neurons from glutamate toxicity. Neuroscience. 1993;55(2):445-9.

Cutler SM, Pettus EH, Hoffman SW, et al. Tapered progesterone withdrawal enhances behavioral and molecular recovery after traumatic brain injury. Experimental neurology. 2005;195(2):423-9.

Pettus EH, Wright DW, Stein DG, et al. Progesterone treatment inhibits the inflammatory agents that accompany traumatic brain injury. Brain research. 2005;1049(1):112-9.

Pan D, Liu W, Yang X, et al. Inhibitory effect of progesterone on inflammatory factors after experimental traumatic brain injury. Biomedical and Environmental Sciences. 2007;20(5):432.

He J, Evans C-O, Hoffman SW, et al. Progesterone and allopregnanolone reduce inflammatory cytokines after traumatic brain injury. Experimental neurology. 2004;189(2):404-12.

Hubbard P. The response of novel NG2 glia to spinal cord injury. Spinal Res. 2003;1:100-7.

Hubbard P, Berry M, Butt A. NG2 glia (oligodendrocyte progenitor cells) in the adult rat spinal cord. Journal of Anatomy. 2002;200(2):213.

Labombarda F, Gonzalez S, Deniselle MCG, et al. Progesterone increases the expression of myelin basic protein and the number of cells showing NG2 immunostaining in the lesioned spinal cord. Journal of neurotrauma. 2006;23(2):181-92.

Nishiyama A, Chang A, Trapp BD. NG2+ glial cells: a novel glial cell population in the adult brain. Journal of Neuropathology & Experimental Neurology. 1999;58(11):1113-24.

Labombarda F, González SL, Lima A, et al. Effects of progesterone on oligodendrocyte progenitors, oligodendrocyte transcription factors, and myelin proteins following spinal cord injury. Glia. 2009;57(8):884-97.

Allen AR. Surgery of experimental lesion of spinal cord equivalent to crush injury of fracture dislocation of spinal column: a preliminary report. Journal of the American Medical Association. 1911;57(11):878-80.

Anderson DK, Means ED, Waters TR, et al. Microvascular perfusion and metabolism in injured spinal cord after methylprednisolone treatment. Journal of neurosurgery. 1982;56(1):106-13.

Tator CH, Fehlings MG. Review of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanisms. Journal of neurosurgery. 1991;75(1):15-26.

Hausmann O. Post-traumatic inflammation following spinal cord injury. Spinal cord. 2003;41(7):369-78.

Conti A, Cardali S, Genovese T, et al. Role of inflammation in the secondary injury following experimental spinal cord trauma. Journal of neurosurgical sciences. 2003;47(2):89.

Conti A, Aguennouz MH, La Torre D, et al. Expression of the tumor necrosis factor receptor-associated factors 1 and 2 and regulation of the nuclear factor-kB antiapoptotic activity in human gliomas. Journal of neurosurgery. 2005;103(5):873-81.

La Rosa G, Cardali S, Genovese T, et al. Inhibition of the nuclear factor-κB activation with pyrrolidine dithiocarbamate attenuating inflammation and oxidative stress after experimental spinal cord trauma in rats. Journal of Neurosurgery: Spine. 2004;1(3):311-21.

Xu J, Fan G, Chen S, et al. Methylprednisolone inhibition of TNF-α expression and NF-kB activation after spinal cord injury in rats. Molecular brain research. 1998;59(2):135-42.

Bartholdi D, Schwab ME. Expression of pro‐inflammatory cytokine and chemokine mRNA upon experimental spinal cord injury in mouse: An in situ hybridization study. European Journal of Neuroscience. 1997;9(7):1422-38.

Yakovlev A, Faden A. Sequential expression of c-fos protooncogene, TNF-alpha, and dynorphin genes in spinal cord following experimental traumatic injury. Molecular and Chemical Neuropathology. 1994;23(2-3):179-90.

Zendedel A, Johann S, Mehrabi S, et al. Activation and Regulation of NLRP3 Inflammasome by Intrathecal Application of SDF-1a in a Spinal Cord Injury Model. Molecular neurobiology. 2015:1-13.

Habgood M, Bye N, Dziegielewska K, et al. Changes in blood–brain barrier permeability to large and small molecules following traumatic brain injury in mice. European Journal of Neuroscience. 2007;25(1):231-8.

Pineau I, Lacroix S. Proinflammatory cytokine synthesis in the injured mouse spinal cord: multiphasic expression pattern and identification of the cell types involved. Journal of Comparative Neurology. 2007;500(2):267-85.

Pan JZ, Ni L, Sodhi A, et al. Cytokine activity contributes to induction of inflammatory cytokine mRNAs in spinal cord following contusion. Journal of neuroscience research. 2002;68(3):315-22.

Lammertse DP. Update on pharmaceutical trials in acute spinal cord injury. The journal of spinal cord medicine. 2003;27(4):319-25.

Hermann GE, Rogers RC, Bresnahan JC, et al. Tumor necrosis factor-α induces cFOS and strongly potentiates glutamate-mediated cell death in the rat spinal cord. Neurobiology of disease. 2001;8(4):590-9.

Genovese T, Melani A, Esposito E, et al. Selective adenosine A (2a) receptor agonists reduce the apoptosis in an experimental model of spinal cord trauma. Journal of biological regulators and homeostatic agents. 2009;24(1):73-86.

Cantarella G, Di Benedetto G, Scollo M, et al. Neutralization of tumor necrosis factor-related apoptosis-inducing ligand reduces spinal cord injury damage in mice. Neuropsychopharmacology. 2010;35(6):1302-14.

Inukai T, Uchida K, Nakajima H, et al. Tumor necrosis factor-alpha and its receptors contribute to apoptosis of oligodendrocytes in the spinal cord of spinal hyperostotic mouse (twy/twy) sustaining chronic mechanical compression. Spine. 2009;34(26):2848-57.

Drew PD, Chavis JA. Female sex steroids: effects upon microglial cell activation. Journal of neuroimmunology. 2000;111(1):77-85.

Bethea JR, Castro M, Keane RW, et al. Traumatic spinal cord injury induces nuclear factor-κB activation. The Journal of neuroscience. 1998;18(9):3251-60.

Sheldahl L, Marriott L, Bryant D, et al. Neuroprotective effects of estrogen and selective estrogen receptor modulators begin at the plasma membrane. Minerva endocrinologica. 2007;32(2):87.

De Bosscher K, Berghe WV, Haegeman G. Cross-talk between nuclear receptors and nuclear factor κB. Oncogene. 2006;25(51):6868-86.

Safe S, Kim K. Non-classical genomic estrogen receptor (ER)/specificity protein and ER/activating protein-1 signaling pathways. Journal of molecular endocrinology. 2008;41(5):263-75.

Du S, Rubin A, Klepper S, et al. Calcium influx and activation of calpain I mediate acute reactive gliosis in injured spinal cord. Experimental neurology. 1999;157(1):96-105.

Files
IssueVol 54, No 6 (2016) QRcode
SectionArticles
Keywords
Spinal cord injury Tumor necrosis alpha Inducible nitric oxide synthase Progesterone Iran

Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
How to Cite
1.
Farahabadi A, Akbari M, Amini Pishva A, Zendedel A, Arabkheradmand A, Beyer C, Dashti N, Hassanzadeh G. Effect of Progesterone Therapy on TNF-α and iNOS Gene Expression in Spinal Cord Injury Model. Acta Med Iran. 2016;54(6):345-351.