Elsevier

Brain Research

Volume 1283, 4 August 2009, Pages 177-185
Brain Research

Research Report
Protective effects of progesterone administration on axonal pathology in mice with experimental autoimmune encephalomyelitis

https://doi.org/10.1016/j.brainres.2009.04.057Get rights and content

Abstract

Experimental autoimmune encephalomyelitis (EAE), an induced model of Multiple Sclerosis presents spinal cord demyelination, axonal pathology and neuronal dysfunction. Previous work has shown that progesterone attenuated the clinical severity, demyelination and neuronal dysfunction of EAE mice (Garay et al., J. Steroid Biochem. Mol. Biol., 2008). Here we studied if progesterone also prevented axonal damage, a main cause of neurological disability. To this end, some axonal parameters were compared in EAE mice pretreated with progesterone a week before immunization with MOG40–54 and in a group of steroid-free EAE mice. On day 16th after EAE induction, we determined in both groups and in control mice: a) axonal density in semithin sections of the spinal cord ventral funiculus; b) appearance of amyloid precursor protein (APP) immunopositive spheroids as an index of damaged axons; c) levels of the growth associated protein GAP43 mRNA and immunopositive cell bodies, as an index of aberrant axonal sprouting. Steroid-naive EAE mice showed decreased axonal density, shrunken axons, abundance of irregular vesicular structures, degenerating APP+ axons, increased expression of GAP43 mRNA and immunoreactive protein in motoneurons. Instead, EAE mice receiving progesterone treatment showed increased axonal counts, high proportion of small diameter axons, reduced APP+ profiles, and decreased GAP43 expression. In conclusion, progesterone enhanced axonal density, decreased axonal damage and prevented GAP43 hyperexpression in the spinal cord of EAE mice. Thus, progesterone also exerts protective effects on the axonal pathology developing in EAE mice.

Introduction

Experimental autoimmune encephalomyelitis (EAE) is an immune-mediated disease that exhibits characteristics in common with Multiple Sclerosis (MS) (Ayers et al., 2004; Kornek et al., 2006; Wujek et al., 2002). EAE can be induced in different species by administration of myelin proteins, including myelin oligodendrocyte glycoprotein (MOG) (Evron et al., 1984, Keith, 1978). The spinal cord of EAE mice presents low expression of the central myelin proteins MOG, myelin basic protein (MBP) and proteolipid protein (PLP). Neuropathology includes loss of axons, neuronal dysfunction, infiltration of inflammatory cells, microglial activation, astrocytosis, proliferation of oligodendrocyte precursor cells, and neurological deficits (Ayers et al., 2004, Garay et al., 2005, Kim et al., 2006, McQualter and Bernard, 2007, Sun et al., 2003).

A major contributing factor to clinical symptoms of MS and EAE is the axonal damage that worsens with disease progression (Blamire et al., 2007, Dutta et al., 2007, Rovaris et al., 2005, Onuki et al., 2001, Wujek et al., 2002). Axonal pathology includes swelling, deficits in transport, degeneration and disruption of the normal axonal cytoarchitecture, changes of neurofilament phosphorylation, sodium channel distribution and development of amyloid precursor protein (APP) immunostaining (Ayers et al., 2004, Kim et al., 2006, Herrero-Herranz et al., 2008, Papadopoulos et al., 2006; Penkova and Hidalgo, 2003). There is a strong belief that restoration of myelin sheaths in EAE—a factor of critical importance for MS—will greatly preserve axonal survival (Zawadzka and Franklin, 2007). Therefore, testing neuroprotective agents that could preserve axonal integrity of EAE mice may be of potential applications in humans.

There is substantial background supporting a role for progesterone as a regulator of myelin protein expression and prevention of axonal damage both in the peripheral as well as the central nervous system (Azcoitia et al., 2003, De Nicola et al., 2006, Melcangi et al., 2005; Stein et al., 2008). When given to ovariectomized female and male animals, progesterone-treated rats show reduced axonal injury (seen via diminished APP immunoreactivity) when compared to controls (O'Connor et al., 2007). Therefore, it seems that under pathological circumstances or hormone deprivation, demyelination and axonal damage can be prevented by treatment or application of progesterone and/or its reduced derivatives.

Progesterone also induces strong immunosuppression during human pregnancy that may prevent relapses of MS (Confavreux et al., 1998, Druckmann and Druckmann, 2005, Hughes, 2004). This beneficial effect of pregnancy extends to several rodents species (Evron et al., 1984, Keith, 1978). Based on these demonstrations, the European multicentric trial POPART-MUS is currently enrolling post-partum women with MS who receive a mixture of estrogen/progestin to simulate steroid levels achieved during pregnancy, in an attempt to avoid the incidence of post-partum relapses (Confavreux et al., 1998, El-Etr et al., 2005). This clinical trial is supported by demonstrations that estrogens and progesterone protected and/or attenuate EAE development (Bebo et al., 2001, Elloso et al., 2005, Garay et al., 2007, Garay et al., 2008; Kim et al., 2006; Offner, 2004).

Previous studies have shown that progesterone pre-treatment of EAE mice attenuated the clinical outcome, inflammatory cell infiltration and demyelination of the spinal cord (Garay et al., 2007, Garay et al., 2008). Here we studied if progesterone pre-treatment also prevented axonal pathology. Our experimental approach intended to simulate the inhibitory effects of high progestin levels of pregnancy on relapses of MS and the prevention of post-partum relapses by steroid administration.

Section snippets

Results

Toluidine-blue staining of spinal cord sections showed that axonal pathology was a prominent feature of mice with induced EAE, as expected from early reports (Ayers et al., 2004, Kim et al., 2006, Papadopoulos et al., 2006; Penkova and Hidalgo, 2003). Fig. 1 (upper photomicrographs) compared a typical control mouse (a), EAE mouse (b) and EAE mouse receiving progesterone (c). A prominent axonal loss occurred in EAE mice, which showed areas of reduced staining intensity, shrunken axons and

Discussion

The present investigation shows that progesterone pre-treatment enhanced axonal density, decreased axonal damage and prevented GAP43 hyperexpression in the spinal cord of EAE female mice. The use of females was based on demonstrations that immune reactivity is greater in females than males and that men are less susceptible to the disease than women (Bebo et al., 2001, Confavreux et al., 1998, El-Etr et al., 2005). Our paradigm simulates the situation in pregnant women with MS, in which relapses

EAE induction

Adult female C57Bl/6 mice were immunized using a MOG40–54 peptide (200 μg per mouse) emulsified in complete Freund's adjuvant (Sigma, St Louis. MO) containing 0.6 mg Mycobacterium tuberculosis (Instituto Malbran, Argentina). The animals also received i.p. injections of Pertussis toxin (400 ng) (Sigma, St Louis) immediately after the immunization and another boost on the day after. Control mice received the emulsion without the MOG40–54 peptide and the Pertussis toxin. The animals developed EAE

Acknowledgments

We thank the following institutions for financial support: FONCYT (PICT 2004 25610 and PICT 2007-01044), the National Research Council of Argentina (CONICET, PIP 5542 and PIP 112-200801-00542), and the University of Buenos Aires (M016). The assistance of Mariana Lopez Ravasio with the preparation of semithin sections is gratefully acknowledged.

References (55)

  • Herrero-HerranzE. et al.

    Pattern of axonal injury in murine myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis: implications for multiple sclerosis

    Neurobiol. Dis.

    (2008)
  • HughesM.D.

    Multiple sclerosis and pregnancy

    Neurol. Clin.

    (2004)
  • KeithA.B.

    Effect of pregnancy on experimental allergic encephalomyelitis in guinea pigs and rats

    J. Neurol. Sci.

    (1978)
  • KimJ.H. et al.

    Detecting axon damage in spinal cord from a mouse model of multiple sclerosis

    Neurobiol. Dis.

    (2006)
  • KornekB. et al.

    Multiple sclerosis and chronic autoimmune encephalomyelitis: a comparative quantitative study of axonal injury in active, inactive, and remyelinated lesions

    Am. J. Pathol.

    (2000)
  • LinkerR.A. et al.

    Mechanisms of axonal degeneration in EAE—lessons from CNTF and MHC I knockout mice

    J. Neurol. Sci.

    (2005)
  • MelcangiR.C. et al.

    Peripheral nerves: a target for the action of neuroactive steroids

    Brain Res. Brain Res. Rev.

    (2005)
  • O'ConnorC.A. et al.

    Effects of progesterone on neurologic and morphologic outcome following diffuse traumatic brain injury in rats

    Exp. Neurol.

    (2007)
  • PapadopoulosD. et al.

    Axon loss is responsible for chronic neurological deficit following inflammatory demyelination in the rat

    Exp. Neurol.

    (2006)
  • PettusE.H. et al.

    Progesterone treatment inhibits the inflammatory agents that accompany traumatic brain injury

    Brain Res.

    (2005)
  • SteinD.G.

    Progesterone exerts neuroprotective effects after brain injury

    Brain Res. Rev.

    (2008)
  • BeboB.F. et al.

    Low-dose estrogen therapy ameliorates experimental autoimmune encephalomyelitis in two different inbred mouse strains

    J. Immunol.

    (2001)
  • BlamireA.M. et al.

    Axonal damage in the spinal cord of multiple sclerosis patients detected by magnetic resonance spectroscopy

    Magn. Reson. Med.

    (2007)
  • CirizaI. et al.

    Reduced progesterone metabolites protect rat hippocampal neurones from kainic acid excitotoxicity in vivo

    J. Neuroendocrinol.

    (2004)
  • ColemanM.

    Axon degeneration mechanisms: commonality amid diversity

    Nat. Rev. Neurosci.

    (2005)
  • ConfavreuxC. et al.

    Rate of pregnancy-related relapse in multiple sclerosis

    New Engl. J. Med.

    (1998)
  • CurtisR. et al.

    Up-regulation of GAP-43 and growth of axons in rat spinal cord after compression

    J. Neurocytol.

    (1993)
  • Cited by (83)

    • Developmental expression of genes involved in progesterone synthesis, metabolism and action during the post-natal cerebellar myelination

      2021, Journal of Steroid Biochemistry and Molecular Biology
      Citation Excerpt :

      After several washes with phosphate buffer 0.1 M, pH 7.4, tissue blocks were postfixed in 1% OsO4 in 0.1 M phosphate buffer pH 7.4 for 1 h. Afterwards, tissue blocks were dehydrated and flat-embedded in Durcupan (Fluka Chemic AG, Sweden). Semithin sections (0.5 μm) were cut with an ultramicrotome (Reichert Jung Ultracut E) and stained with toluidine blue [48]. Homogenates from whole cerebellum from mice at P5 (n = 5), P18 (n = 7) and P35 (n = 7) were prepared by sonication in ice-cold lysis buffer (50 mM Tris–HCl, 150 mM NaCl, 2 mM EDTA, 1 mM Na3VO4, and 1% TritonX100, pH 7.

    • Regulatory B Cells: Dark Horse in Pregnancy Immunotherapy?

      2021, Journal of Molecular Biology
      Citation Excerpt :

      Progesterone, on the other hand, has been implicated in the promotion of a Th2-dominant immune profile by suppressing B-cell antigen expression and increasing production of protective asymmetric antibodies during pregnancy [70]. A close link between progesterone and Breg function is suggested in parallel studies in cancer and autoimmune diseases where it was demonstrated that progesterone treatment leads to inhibition of T-cell proliferation, production bias of anti-inflammatory cytokines over pro-inflammatory cytokines, and increase in IL-10 production concurrent with expansion of B cells [71–73]. Interestingly, a recent study featured progesterone in the induction of higher CD83 expression in B cells, a proposed novel pathway for establishing fetal tolerance [74].

    View all citing articles on Scopus
    View full text