Estriol preserves synaptic transmission in the hippocampus during autoimmune demyelinating disease

Marina O Ziehn, Andrea A Avedisian, Shannon M Dervin, Thomas J O'Dell, Rhonda R Voskuhl, Marina O Ziehn, Andrea A Avedisian, Shannon M Dervin, Thomas J O'Dell, Rhonda R Voskuhl

Abstract

Cognitive deficits occur in over half of multiple sclerosis patients, with hippocampal-dependent learning and memory commonly impaired. Data from in vivo MRI and post-mortem studies in MS indicate that the hippocampus is targeted. However, the relationship between structural pathology and dysfunction of the hippocampus in MS remains unclear. Hippocampal neuropathology also occurs in experimental autoimmune encephalomyelitis (EAE), the most commonly used animal model of MS. Although estrogen treatment of EAE has been shown to be anti-inflammatory and neuroprotective in the spinal cord, it is unknown if estrogen treatment may prevent hippocampal pathology and dysfunction. In the current study we examined excitatory synaptic transmission during EAE and focused on pathological changes in synaptic protein complexes known to orchestrate functional synaptic transmission in the hippocampus. We then determined if estriol, a candidate hormone treatment, was capable of preventing functional changes in synaptic transmission and corresponding hippocampal synaptic pathology. Electrophysiological studies revealed altered excitatory synaptic transmission and paired-pulse facilitation (PPF) during EAE. Neuropathological experiments demonstrated that there were decreased levels of pre- and post-synaptic proteins in the hippocampus, diffuse loss of myelin staining and atrophy of the pyramidal layers of hippocampal cornu ammonis 1 (CA1). Estriol treatment prevented decreases in excitatory synaptic transmission and lessened the effect of EAE on PPF. In addition, estriol treatment prevented several neuropathological alterations that occurred in the hippocampus during EAE. Cross-modality correlations revealed that deficits in excitatory synaptic transmission were significantly correlated with reductions in trans-synaptic protein binding partners known to modulate excitatory synaptic transmission. To our knowledge, this is the first report describing a functional correlate to hippocampal neuropathology in any MS model. Furthermore, a treatment was identified that prevented both deficits in synaptic function and hippocampal neuropathology.

Figures

Figure 1. Experimental design and standard clinical…
Figure 1. Experimental design and standard clinical scores in EAE
(A) Experimental design depicting the timing of placebo or estriol pellet implantation (D -7), EAE induction (D 0 & D 7), daily clinical scoring (D 7-40) and electrophysiology (D 21-40). Pertussis injections (intraperitoneal, i.p.) were given to both EAE groups on D0 and D2, as part of standard EAE induction, not shown. All mice were age-matched C57Bl/6 adult females, and were uniform randomly selected (one per day) for electrophysiological recording. On each day of electrophysiology, the right hemisphere was prepared for electrophysiology, while the left hemisphere of each mouse was prepared for pathology studies and kept until all tissue from all conditions had been collected. (B) EAE clinical scores were recorded in placebo-treated (EAE + PLAC, black star, dashed black line) or estriol-treated (EAE + ESTRIOL, gray triangle, dashed gray line) mice as well as in non-EAE induced healthy controls (NL, black square, solid black line). Placebo-treated mice with EAE exhibited a severe clinical disease course, while estriol-treated mice exhibited significantly reduced clinical severity. Data are representative of two separate experiments, and experiment has been repeated more four times. Repeated measures ANOVA with post-hoc pair-wise comparisons, revealed that placebo-treated EAE mice were significantly different from other two groups beginning at D12 of disease, * p < 0.05, n = 5 per group per experiment.
Figure 2. Excitatory synaptic transmission and paired-pulse…
Figure 2. Excitatory synaptic transmission and paired-pulse facilitation are altered during EAE
(A) Input/output (IO) curves were created by comparing fiber volley amplitude (FV Amp; mV) to field excitatory postsynaptic potential (fEPSP; mV/ms) during four different stimulation intensities of presynaptic fiber stimulation that caused fEPSP responses of 25, 50, 75 and 100% maximal fEPSP amplitude. One-way ANOVA analyses demonstrated that placebo-treated EAE (EAE+PLAC; black star, dashed black line) fEPSP responses were significantly reduced at each point along the IO curve (percentage of maximal fEPSP slope: 25, 50 75 and 100%) in comparison to healthy controls (NL; black squares, solid black line), p

Figure 3. Hippocampal PSD-95 levels are decreased…

Figure 3. Hippocampal PSD-95 levels are decreased in EAE and preserved with estriol treatment

(A-F)…

Figure 3. Hippocampal PSD-95 levels are decreased in EAE and preserved with estriol treatment
(A-F) Fluorescent images depict the stratum radiatum with the CA1 region of representative hippocampal slices from healthy (panel A-B), placebo-treated EAE (panel C-D) and estriol-treated EAE (panel E-F) mice, where PSD-95 (A, C & E; Cy5-red), and Synapsin-1 (B, D & F; Syn-1; Cy3-green) staining are shown at 60x magnification. During EAE, PSD-95 % area was significantly decreased in the CA1 region of the hippocampus, (C), compared to healthy controls (A). This decrease was prevented with estriol treatment during EAE, (E), quantified in graph G. One-way ANOVA and Bonferroni post-hoc analysis revealed statistical significance in PSD-95 % area where, * indicates p = 0.0324, n = 5 per group. Average number of presynaptic Syn-1+ puncta within the CA1 was not significantly different across conditions. These experiments were repeated twice with similar results. Scale bar 10um.

Figure 4. Presynaptic β-neurexin II and postsynaptic…

Figure 4. Presynaptic β-neurexin II and postsynaptic neuroligin-1 are decreased during EAE, but preserved with…

Figure 4. Presynaptic β-neurexin II and postsynaptic neuroligin-1 are decreased during EAE, but preserved with estriol treatment
Synaptic puncta were measured in the hippocampal CA1 region by counting immunoreactive NRXIIβ puncta (A-B) and NLG1 puncta (C-D) in healthy control, (panels i-ii), placebo-treated EAE, (panels iii-iv), and estriol-treated EAE mice (panels v-vi), respectively. Pseudo-colored confocal images depict synaptic staining (either NRXIIβ or NLG1, Cy3-red) in hippocampus at 60x magnification, while black and white images show outlined puncta from respective confocal images. One-way ANOVA revealed that groups differed significantly in quantity of NRXIIβ puncta, (p = 0.005) and Bonferroni post-hoc test indicated that placebo-treated EAE mice (EAE+PLAC, n = 5 mice) had significantly decreased (* p < 0.01) numbers of NRXIIβ puncta compared to healthy control (NL, n = 5 mice) and estriol-treated EAE mice (EAE+ESTRIOL, n = 4 mice), graph B. Simultaneously, NLG1 puncta quantities were also significantly different across conditions (one-way ANOVA, p = 0.0056) with follow up tests indicating significant difference between healthy controls (NL, n = 5 mice) and placebo-treated EAE mice (EAE+PLAC, ** p < 0.01, n = 5 mice), as well as significant difference between placebo-treated EAE and estriol-treated EAE mice (EAE+ESTRIOL, * p < 0.05, n = 5 mice), graph D. These experiments were repeated twice and similar results were found. Scale bars 10um.

Figure 5. Decreased myelin staining and increased…

Figure 5. Decreased myelin staining and increased microglial activation occur within the hippocampus during EAE,…

Figure 5. Decreased myelin staining and increased microglial activation occur within the hippocampus during EAE, and estriol treatment preserves myelin staining
(A-C) Pseudo-colored confocal fluorescent images depict myelin basic protein immunoreactivity (MBP, Cy5-red) and cells of microglial/macrophage lineage stained with Iba1 (Cy3-green) and Dapi (blue) in the CA1 region within healthy (A), placebo-treated EAE (B) and estriol-treated EAE (C) mice at 40x magnification. (D) One-way ANOVA revealed significant differences in myelin staining across conditions, p = 0.003, n = 5 mice per group, and Bonferroni post-hoc tests indicated that EAE caused a significant reduction in myelin staining within the CA1 region of the hippocampus in placebo-treated mice (EAE+PLAC, ** p < 0.01, n = 5 mice) compared to myelin staining in hippocampus of healthy control mice (NL, n = 5 mice). Estriol-treated EAE mice (EAE+ESTRIOL, n = 5 mice) had myelin levels significantly increased compared to placebo-treated EAE mice (* p < 0.05), but were still significantly different from myelin staining levels in healthy controls. Analysis was conducted on 10x magnification images where MBP % area represents total MBP immunoreactivity as a percent of total delineated CA1 area imaged. Iba1+ cells with morphology characteristic of reactive microglia were relatively low in healthy controls (A), and increased in placebo-treated EAE mice (B). The number of Iba1+ cells was not significantly different in estriol-treated EAE mice (C), however. The number of Iba1+ cells per mm3 were counted and presented as whole numbers (graph E). Data are representative of two experiments. Scale bar 10um.

Figure 6. Hippocampal CA1 atrophy occurs during…

Figure 6. Hippocampal CA1 atrophy occurs during EAE and estriol treatment prevents this

(A-C) Nissl-stained…

Figure 6. Hippocampal CA1 atrophy occurs during EAE and estriol treatment prevents this
(A-C) Nissl-stained hippocampal sections representative of normal (NL; A), placebo-treated EAE (EAE+PLAC; B) and estriol-treated EAE (EAE+ESTRIOL; C) mice depict the CA1 pyramidal layer at 40x magnification. Compared to healthy controls, placebo-treated EAE mice had significantly reduced CA1 pyramidal layer volume, quantified in D. With estriol treatment during EAE, the CA1 pyramidal volume was preserved to levels similar to those in controls. One-way ANOVA and Bonferroni post-hoc analysis revealed significant difference between three groups, (*p = 0.05, n = 5 mice per group), scale bar 10um. This finding has been confirmed by repeated experiments.

Figure 7. Decreased pre- and postsynaptic protein…

Figure 7. Decreased pre- and postsynaptic protein levels are significantly correlated with electrophysiological changes during…

Figure 7. Decreased pre- and postsynaptic protein levels are significantly correlated with electrophysiological changes during EAE
Cross-modality correlations were conducted to determine potential neuropathological substrates underlying changes in excitatory synaptic transmission and paired-pulse facilitation during EAE. (A) PSD-95% area was significantly correlated to maximal fEPSP slope throughout experiment, where the correlation coefficient rho = 0.8767, p = 0.019, n = 9 mice total. (B) The average number of presynaptic NRXIIβ puncta was also significantly correlated to postsynaptic responses (fEPSP) where rho = 0.8396, p = 0.0046, n = 9 mice total.
All figures (7)
Figure 3. Hippocampal PSD-95 levels are decreased…
Figure 3. Hippocampal PSD-95 levels are decreased in EAE and preserved with estriol treatment
(A-F) Fluorescent images depict the stratum radiatum with the CA1 region of representative hippocampal slices from healthy (panel A-B), placebo-treated EAE (panel C-D) and estriol-treated EAE (panel E-F) mice, where PSD-95 (A, C & E; Cy5-red), and Synapsin-1 (B, D & F; Syn-1; Cy3-green) staining are shown at 60x magnification. During EAE, PSD-95 % area was significantly decreased in the CA1 region of the hippocampus, (C), compared to healthy controls (A). This decrease was prevented with estriol treatment during EAE, (E), quantified in graph G. One-way ANOVA and Bonferroni post-hoc analysis revealed statistical significance in PSD-95 % area where, * indicates p = 0.0324, n = 5 per group. Average number of presynaptic Syn-1+ puncta within the CA1 was not significantly different across conditions. These experiments were repeated twice with similar results. Scale bar 10um.
Figure 4. Presynaptic β-neurexin II and postsynaptic…
Figure 4. Presynaptic β-neurexin II and postsynaptic neuroligin-1 are decreased during EAE, but preserved with estriol treatment
Synaptic puncta were measured in the hippocampal CA1 region by counting immunoreactive NRXIIβ puncta (A-B) and NLG1 puncta (C-D) in healthy control, (panels i-ii), placebo-treated EAE, (panels iii-iv), and estriol-treated EAE mice (panels v-vi), respectively. Pseudo-colored confocal images depict synaptic staining (either NRXIIβ or NLG1, Cy3-red) in hippocampus at 60x magnification, while black and white images show outlined puncta from respective confocal images. One-way ANOVA revealed that groups differed significantly in quantity of NRXIIβ puncta, (p = 0.005) and Bonferroni post-hoc test indicated that placebo-treated EAE mice (EAE+PLAC, n = 5 mice) had significantly decreased (* p < 0.01) numbers of NRXIIβ puncta compared to healthy control (NL, n = 5 mice) and estriol-treated EAE mice (EAE+ESTRIOL, n = 4 mice), graph B. Simultaneously, NLG1 puncta quantities were also significantly different across conditions (one-way ANOVA, p = 0.0056) with follow up tests indicating significant difference between healthy controls (NL, n = 5 mice) and placebo-treated EAE mice (EAE+PLAC, ** p < 0.01, n = 5 mice), as well as significant difference between placebo-treated EAE and estriol-treated EAE mice (EAE+ESTRIOL, * p < 0.05, n = 5 mice), graph D. These experiments were repeated twice and similar results were found. Scale bars 10um.
Figure 5. Decreased myelin staining and increased…
Figure 5. Decreased myelin staining and increased microglial activation occur within the hippocampus during EAE, and estriol treatment preserves myelin staining
(A-C) Pseudo-colored confocal fluorescent images depict myelin basic protein immunoreactivity (MBP, Cy5-red) and cells of microglial/macrophage lineage stained with Iba1 (Cy3-green) and Dapi (blue) in the CA1 region within healthy (A), placebo-treated EAE (B) and estriol-treated EAE (C) mice at 40x magnification. (D) One-way ANOVA revealed significant differences in myelin staining across conditions, p = 0.003, n = 5 mice per group, and Bonferroni post-hoc tests indicated that EAE caused a significant reduction in myelin staining within the CA1 region of the hippocampus in placebo-treated mice (EAE+PLAC, ** p < 0.01, n = 5 mice) compared to myelin staining in hippocampus of healthy control mice (NL, n = 5 mice). Estriol-treated EAE mice (EAE+ESTRIOL, n = 5 mice) had myelin levels significantly increased compared to placebo-treated EAE mice (* p < 0.05), but were still significantly different from myelin staining levels in healthy controls. Analysis was conducted on 10x magnification images where MBP % area represents total MBP immunoreactivity as a percent of total delineated CA1 area imaged. Iba1+ cells with morphology characteristic of reactive microglia were relatively low in healthy controls (A), and increased in placebo-treated EAE mice (B). The number of Iba1+ cells was not significantly different in estriol-treated EAE mice (C), however. The number of Iba1+ cells per mm3 were counted and presented as whole numbers (graph E). Data are representative of two experiments. Scale bar 10um.
Figure 6. Hippocampal CA1 atrophy occurs during…
Figure 6. Hippocampal CA1 atrophy occurs during EAE and estriol treatment prevents this
(A-C) Nissl-stained hippocampal sections representative of normal (NL; A), placebo-treated EAE (EAE+PLAC; B) and estriol-treated EAE (EAE+ESTRIOL; C) mice depict the CA1 pyramidal layer at 40x magnification. Compared to healthy controls, placebo-treated EAE mice had significantly reduced CA1 pyramidal layer volume, quantified in D. With estriol treatment during EAE, the CA1 pyramidal volume was preserved to levels similar to those in controls. One-way ANOVA and Bonferroni post-hoc analysis revealed significant difference between three groups, (*p = 0.05, n = 5 mice per group), scale bar 10um. This finding has been confirmed by repeated experiments.
Figure 7. Decreased pre- and postsynaptic protein…
Figure 7. Decreased pre- and postsynaptic protein levels are significantly correlated with electrophysiological changes during EAE
Cross-modality correlations were conducted to determine potential neuropathological substrates underlying changes in excitatory synaptic transmission and paired-pulse facilitation during EAE. (A) PSD-95% area was significantly correlated to maximal fEPSP slope throughout experiment, where the correlation coefficient rho = 0.8767, p = 0.019, n = 9 mice total. (B) The average number of presynaptic NRXIIβ puncta was also significantly correlated to postsynaptic responses (fEPSP) where rho = 0.8396, p = 0.0046, n = 9 mice total.

References

    1. Chiaravalloti ND, DeLuca J. Cognitive impairment in multiple sclerosis. Lancet Neurol. 2008;7:1139–1151.
    1. Benedict RHB, Cookfair D, Gavett R, et al. Validity of the minimal assessment of cognitive function in multiple sclerosis (MCFIMS) J Int Neuropsychol Soc. 2006;12(4):549–558.
    1. Thornton AE, Raz N. Memory impairment in multiple sclerosis. Neuropsychology. 1997;11:357–366.
    1. Benedict RHB, Ramasamy D, Munschauer F, et al. Memory impairment in multiple sclerosis: correlation with deep grey matter and mesial temporal atrophy. J Neurol Neurosurg Psychiatry. 2009;80:201–206.
    1. Roosendaal SD, Moraal B, Vrenken H, et al. In vivo imaging of hippocampal lesions in multiple sclerosis. J Magn Reson Imaging. 2008;27:726–731.
    1. Anderson VM, Fisniku LK, Khaleeli A, et al. Hippocampal atrophy in relapsing-remitting and primary progressive MS: a comparative study. Mult Scler. 2010;16:1083–1090.
    1. Sicotte NL, Kern KC, Giesser BS, et al. Regional hippocampal atrophy in multiple sclerosis. Brain. 2009;132:3072–3086.
    1. Geurts JJ, Bo L, Roosendaal SD, et al. Extensive hippocampal demyelination in multiple sclerosis. J Neuropathol Exp Neurol. 2007;66:819–827.
    1. Papadopoulos D, Dukes S, Patel R, et al. Substantial archaeocortical atrophy and neuronal loss in multiple sclerosis. Brain Pathol. 2009;19:238–253.
    1. Dutta R, Chang A, Doud MK, et al. Demyelination causes synaptic alterations in hippocampi from Multiple Sclerosis patients. Ann Neurol. 2011;69:445–454.
    1. Rasmussen S, Wang Y, Kivisakk P, et al. Persistent activation of microglia is associated with neuronal dysfunction of callosal projecting pathways and multiple sclerosis-like lesions in relapsing remitting experimental autoimmune encephalomyelitis. Brain. 2007;130:2816–2829.
    1. Crawford DK, Mangiardi M, Song B, et al. Oestrogen receptor beta ligand: a novel treatment to enhance endogenous functional remyelination. Brain. 2010;133:2999–3016.
    1. MacKenzie-Graham A, Tiwari-Woodruff SK, Sharma G, et al. Purkinje cell loss in experimental autoimmune encephalomyelitis. Neuroimage. 2009;48:637–651.
    1. Centonze D, Muzio L, Rossi S, et al. Inflammation triggers synaptic alteration and degeneration in experimental autoimmune encephalomyelitis. J Neurosci. 2009;29:3442–3452.
    1. Gold SM, Voskuhl RR. Estrogen and testosterone therapies in multiple sclerosis. Prog Brain Res. 2009;175:239–251.
    1. Confavreux C, Hutchinson M, Hours MM, et al. Rate of pregnancy-related relapse in multiple sclerosis. Pregnancy in Multiple Sclerosis Group. New Eng J of Med. 1998;339:285–291.
    1. Sicotte NL, Liva SM, Kluth R, et al. Treatment of multiple sclerosis with the pregnancy hormone estriol. Ann Neurol. 2002;52:421–428.
    1. Kim S, Liva SM, Dalal MA, et al. Estriol ameliorates autoimmune demyelinating disease: implications for multiple sclerosis. Neurology. 1999;52(6):1230–1238.
    1. Bebo BF, Jr, Fyfe-Johnson A, Adlard K, et al. Low-dose estrogen therapy ameliorates experimental autoimmune encephalomyelitis in two difference inbred mouse strains. J Immunol. 2001;166(3):2080–2089.
    1. Morales LBJ, Loo KK, Liu HB, et al. Treatment with an estrogen receptor {alpha} ligand Is neuroprotective in experimental autoimmune encephalomyelitis. J Neurosci. 2006;26:6823–6823.
    1. Tiwari-Woodruff S, Morales LBJ, Lee R, et al. Differential neuroprotective and anti-inflammatory effects of estrogen receptor (ER)α and ERβ ligand treatment. PNAS. 2007;104(37):14813–14818.
    1. Vetego E, Benedusi V, Maggi A. Estrogen anti-inflammatory activity in the brain: A therapeutic opportunity for menopause and neurodegenerative diseases. Front Neuroendocrinol. 2008;29(4):507–519.
    1. Stein DG, Hoffman SW. Estrogen and progesterone as neuroprotective agents in the treatment of acute brain injuries. Pediatr Rehabil. 2003;6(1):13–22.
    1. Wise PM, Dubal DB, Rau SW, et al. Are estrogens protective or risk factors in brain injury and neurodegeneration? Reevaluation after the Women’s Health Initiative. Endocr Rev. 2005;26(3):308–312.
    1. Behl C, Skutella T, Lezoualch F, et al. Neuroprotection against oxidative stress by estrogens: structure-activity relationship. Mol Pharmacol. 1997;51(4):535–541.
    1. Murphy DD, Cole NB, Greenberger V, et al. Estradiol increases dendritic spine density by reducing GABA neurotransmission in hippocampal neurons. J Neurosci. 1998;18:2550–2559.
    1. Jover T, Tanaka H, Calderone A, et al. Estrogen protects against global ischemia-induced neuronal death and prevents activation of apoptotic signaling cascades in the hippocampal CA1. J Neurosci. 2002;22:2115–2124.
    1. Kretz O, Fester L, Wehrenberg U, et al. Hippocampal synapses depend on hippocampal estrogen synthesis. J Neurosci. 2004;24(26):5913–5921.
    1. Toran-Allerand CD. Novel sites and mechanism of oestrogen action in the brain. Novartis Found Symp. 2000;230:56–73.
    1. Kramar EA, Chen LY, Brandon NJ, et al. Cytoskeletal changes underlie estrogen’s acute effects on synaptic transmission and plasticity. J Neurosci. 2009;29(41):12982–12993.
    1. Yun SH, Park KA, Kwon S, et al. Estradiol enhances long-term potentiation in hippocampal slices from aged apoE4-TR mice. Hippocampus. 2007;17(12):1153–1157.
    1. Woolley CS. Effects of estrogen in the CNS. Curr Opin Neurobiol. 1999;9(3):349–354.
    1. Woolley CS. Acute effects of estrogen on neuronal physiology. Annu Rev Pharmacol Toxicol. 2007;47:657–680.
    1. Ziehn MO, Avedisian AA, Tiwari-Woodruff S, et al. Hippocampal CA1 atrophy and synaptic loss during experimental autoimmune encephalomyelitis, EAE. Lab Invest. 2010;90:774–786.
    1. Pettinelli C, McFarlin DE. Adoptive transfer of experimental allergic encephalomyelitis in SJL/J mice after in vitro activation of lymph node cells by myelin basic protein: requirement for Lyt 1+ 2- T lymphocytes. J Immunol. 1981;127:1420–1423.
    1. Cuthbert PC, Stanford LA, Coba MP, et al. Synapse-associated protein 102/dlgh3 couples the NMDA receptor to specific plasticity pathways and learning strategies. J Neurosci. 2007;27(10):2673–2682.
    1. Komiyama NH, Watabe AM, Carlisle HJ, et al. SynGAP regulates ERK/MAPK signaling, synaptic plasticity, and learning in the complex with postsynaptic density 95 and NMDA receptor. J Neurosci. 2002;22(22):9721–9732.
    1. Wiltgen BJ, Royle GA, Gray EE, et al. A role for calcium-permeable AMPA receptors in synaptic plasticity and learning. PLoS One. 2010;5(9):e12818. pii.
    1. Paxinos G, Franklin KBJ. The Mouse Brain in Stereological Coordinates. 2nd ed. San Diego, CA: Academic Press; 2001.
    1. Gundersen HJ, Jensen EB. The efficiency of systematic sampling in stereology and its prediction. J Microsc. 1987;147:229–263.
    1. Cruz-Orive LM. Precision of Cavalieri sections and slices with local errors. J Microsc. 1999;193:182–198.
    1. Spence RD, Hamby ME, Umeda E, et al. Neuroprotection mediated through estrogen receptor-alpha in astrocytes. Proc Natl Acad Sci USA. 2011;108(21):8867–8872.
    1. Arezzo JC, Litwak MS, Zotova EG. Correlation and dissociation of electrophysiology and histopathology in the assessment of toxic neuropathy. Toxicol Pathol. 2011;39:46–51.
    1. Aubert S, Wendling F, Regis J, McGonigal A, Figarella-Branger D, Peragut JC, Girard N, Chauvel P, Bartolomei F. Local and remote epileptogenicity in focal cortical dysplasias and neurodevelopmental tumours. Brain. 2009;132:3072–3086.
    1. Lehmann HC, Zhang J, Mori S, et al. Diffusion tensor imaging to assess axonal regeneration in peripheral nerves. Exp Neurol. 2010;223(1):238–244.
    1. Palaszynski KM, Liu H, Loo KK, et al. Estriol treatment ameliorates disease in males with experimental autoimmune encephalomyelitis: implications for multiple sclerosis. J Neuroimmunol. 2004;149(1-2):84–89.
    1. Futai K, Kim MJ, Hashikawa T, et al. Retrograde modulation of presynaptic release probability through signaling mediated by PSD-95-neuroligin. Nat Neurosci. 2007;10(2):186–195.
    1. Kuzumaki N, Ikegami D, Imai S, et al. Enhanced IL-1beta production in response to the activation of hippocampal glial cells impairs neurogenesis in aged mice. Synapse. 2010;64:721–728.
    1. Voskuhl RR, Peterson RS, Song B, et al. Reactive astrocytes form scar-like perivascular barriers to leukocytes during adaptive immune inflammation of the CNS. J Neurosci. 2009;29:11511–11522.
    1. Debanne D, Guerineau NC, Gahwiler BH, et al. Paired-pulse facilitation and depression at unitary synapses in rat hippocampus: quantal fluctuation affects subsequent release. J Physiol. 1996;15(Pt 1):163–176.
    1. Dobrunz LE, Stevens CF. Heterogeneity of release probability, facilitation and depletion at central synapses. Neuron. 1997;18:995–1008.
    1. Zucker RS, Regehr WG. Short-term synaptic plasticity. Annu Rev Physiol. 2002;64:355–405.
    1. Conti R, Lisman J. The high variance of AMPA receptor- and NMDA receptor-mediated responses at single hippocampal synapses: evidence for multi-quantal release. Proc Natl Acad Sci. 2003;100:4885–4890.
    1. Dean C, Dresbach T. Neuroligins and neurexins: linking cell adhesion, synapse formation and cognitive function. Trends in Neurosci. 2006;29(1):21–29.
    1. Kim E, Sheng M. PDZ domain proteins of synapses. Nat Rev Neurosci. 2004;5(10):771–781.
    1. Han K, Kim E. Synaptic adhesion molecules and PSD-95. Prog Neurobiol. 2008;84(3):263–283.
    1. Nguyen T, Sudhof TC. Binding properties of neuroligin 1 and neurexin 1beta reveal function as heterophilic cell adhesion molecules. J Biol Chem. 1997;272:26032–26039.
    1. Dean C, Scholl FG, Choih J, et al. Neurexin mediates the assembly of presynaptic terminals. Nat Neusci. 2003;6(7):708–716.
    1. Levinson JN, Chery N, Huang K, et al. Neuroligins mediate excitatory and inhibitory synapse formation: involvement of PSD-95 and neurexin-1beta in neuroligin-induced synaptic specificity. J Biol Chem. 2005;280(17):17312–17319.
    1. Jelks KB, Wylie R, Floyd CL, et al. Estradiol targets synaptic proteins to induce glutamatergic synapse formation in cultured hippocampal neurons: critical role of estrogen receptor-alpha. J Neurosci. 2007;27:6903–6913.
    1. Teyler TJ, Vardaris RM, Lewis D, et al. Gonadal steroids: effects on excitability of hippocampal pyramidal cells. Science. 1980;209(4460):1017–1018.
    1. Smejkalova T, Woolley CS. Estradiol acutely potentiates hippocampal excitatory synaptic transmission through a presynaptic mechanism. J Neurosci. 2010;30(48):16137–16148.
    1. Foy MR, Xu J, Xie X, et al. 17β-estradiol enhances NMDA receptor-mediated EPSPs and long-term potentiation. J Neurophysiol. 1999;81:925–929.
    1. Li C, Brake WG, Romeo RD, et al. Estrogen alters hippocampal dendritic spine shape and enhances synaptic protein immunoreactivity and spatial memory in female mice. Proc Natl Acad Sci. 2004;101(7):2185–2190.
    1. Kang HS, Lee CK, Kim JR, et al. Gene expression analysis of the pro-oestrous-stage rat uterus reveals neuroligin 2 as a novel steroid-regulated gene. Reprod Fertil Dev. 2004;16(8):763–772.
    1. Garcia-Segura LM, Azcoitia I, DonCarlos LL. Neuroprotection by estradiol. Prog Neurobiol. 2001;63(1):29–60.
    1. Carroll JC, Rosario ER, Chang L, et al. Progesterone and estrogen regulate Alzheimer-like neuropathology in female 3xTg-AD mice. J. Neurosci. 2007;27:13357–13365.
    1. Pike CJ, Carroll JC, Rosario ER, et al. Protective actions of sex steroid hormones in Alzheimer’s disease. Front Neuroendocrinol. 2009;30(2):239–258.
    1. Voskuhl RR. “Estrogens in the Treatment of MS” Multiple Sclerosis Therapeutics. 3rd Edition. United Kingdom: Informa Healthcare; 2007. pp. 645–657.
    1. Enmark E, Gustafsson JA. Oestrogen receptors – an overview. J Intern Med. 1999;246(2):133–138.
    1. Mehra RD, Sharma K, Nyakas C, et al. Estrogen receptor alpha and beta immunoreactive neurons in normal adult and aged female rat hippocampus: a qualitative and quantitative study. Brain Res. 2005;1056(1):22–35.
    1. Pfaff DW, Ribiero AC. Theoretical consequences of fluctuating versus constant liganding of oestrogen receptor-alpha in neurons. J Neuroendocrinol. 2010;22(6):486–491.
    1. Azcoitia I, Santos-Galindo M, Arevalo MA, et al. Role of astroglia in the neuroplastic and neuroprotective actions of estradiol. Eur J Neurosci. 2010;32(12):1995–2002.
    1. Arevalo MA, Santos-Galindo M, Bellini MJ, et al. Actions of estrogens on glial cells: implications for neuroprotection. Biochim Biophys Acta. 2010;1800(10):1106–1112.
    1. Mitterling KL, Spencer JL, Dziedzic N, et al. Cellular and subcellular localization of estrogen and progestin receptor immunoreactivities in the mouse hippocampus. J Comp Neurol. 2010;518(14):2729–2743.

Source: PubMed

3
Tilaa