Inhibition of soluble tumor necrosis factor ameliorates synaptic alterations and Ca2+ dysregulation in aged rats

Diana M Sama, Hafiz Mohmmad Abdul, Jennifer L Furman, Irina A Artiushin, David E Szymkowski, Stephen W Scheff, Christopher M Norris, Diana M Sama, Hafiz Mohmmad Abdul, Jennifer L Furman, Irina A Artiushin, David E Szymkowski, Stephen W Scheff, Christopher M Norris

Abstract

The role of tumor necrosis factor α (TNF) in neural function has been investigated extensively in several neurodegenerative conditions, but rarely in brain aging, where cognitive and physiologic changes are milder and more variable. Here, we show that protein levels for TNF receptor 1 (TNFR1) are significantly elevated in the hippocampus relative to TNF receptor 2 (TNFR2) in aged (22 months) but not young adult (6 months) Fischer 344 rats. To determine if altered TNF/TNFR1 interactions contribute to key brain aging biomarkers, aged rats received chronic (4-6 week) intracranial infusions of XPro1595: a soluble dominant negative TNF that preferentially inhibits TNFR1 signaling. Aged rats treated with XPro1595 showed improved Morris Water Maze performance, reduced microglial activation, reduced susceptibility to hippocampal long-term depression, increased protein levels for the GluR1 type glutamate receptor, and lower L-type voltage sensitive Ca(2+) channel (VSCC) activity in hippocampal CA1 neurons. The results suggest that diverse functional changes associated with brain aging may arise, in part, from selective alterations in TNF signaling.

Conflict of interest statement

Competing Interests: The authors have read the journal's policy and have the following conflicts: David E. Szymkowski is the Senior Director of Bio-therapeutics at Xencor: the company that manufactures the anti-TNF biologic XPro1595 used in our study. Dr. Szymkowski's role on this project was to provide council on dosing and to provide a background on XPro1595's unique mechanism of action. Dr. Szymkowski played no role in data collection or statistical comparisons. Because the neuroscience field is outside his area of expertise, he did not provide any council or shade any interpretation of XPro1595's effects on neurologic function. For these reasons, the authors that Dr. Szymkowski's status as an author for this study is appropriate and in no way compromised or biased the experimental results or interpretations. Dr. Hafiz Mohmmad Abdul is now employed by Proteostasis Therapeutics, Inc. Dr. Mohmmad Abdul took this position after the data in our article were collected and interpreted. Please note that Proteostasis Therapeutics Inc had no role whatsoever in any portion of this study. The corresponding author of this article, Dr. Chris Norris, currently serves as an Academic Editor at PLoS ONE. This does not alter the authors' adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. Increased TNFR1/TNFR2 ratio during aging.
Figure 1. Increased TNFR1/TNFR2 ratio during aging.
A, Representative Western blots for TNFR1 and TNFR2 in hippocampal membrane fractions from young (6 mos; n = 6) and aged (22 mos; n = 10) male Fischer rats, with the Na/K-ATPase loading control shown below. B, There was no significant change in TNFR1 protein levels with age (p = 0.67); however, there was a significant decrease in TNFR2 with age (* p<0.05).
Figure 2. XPro1595 alters Iba-1 levels.
Figure 2. XPro1595 alters Iba-1 levels.
XPro1595 (0.08 mg/kg/day) was administered to the hippocampus over a six week period using Alzet osmotic pumps. Immunolabeling revealed a reduction in inflammatory phenotype in microglia (A, Iba-1) and, to a lesser extent, in astrocytes (D, GFAP) of XPro1595 treated aged rats. CA1 = CA1 stratum pyramidale. Representative Western blots and mean ± SEM protein levels are for Iba-1 (B and C) and GFAP (E and F) in hippocampal fractions of vehicle and XPro1595-treated rats. β-actin and GAPDH served as loading controls for Iba-1 and GFAP, respectively. Results revealed a marked, significant reduction in Iba-1 protein levels (* p<0.05) and a smaller, nonsignificant reduction in GFAP levels in the XPro1595 group (p = 0.45).
Figure 3. XPro1595 improves learning and synaptic…
Figure 3. XPro1595 improves learning and synaptic measures. A,
For behavioral assays, XPro1595 (0.08 mg/kg/day) was administered to the hippocampus over a six week period. Performance (mean ± SEM) in the spatial version of the Morris Water Maze was measured by path length (cm), and normalized (%) to block one performance levels for each rat. Each group showed significant improvement by block 6 (p<0.01). However, XPro1595-, but not vehicle-treated, rats showed significant improvement as early as block 2 and 3, suggesting that TNF-blockade accelerates learning rates during aging. B, Mean EPSP slope amplitudes (vertical SEM bars) were plotted against mean FV amplitudes (horizontal SEM bars) to generate CA3-CA1 synaptic strength curves. Each average curve was fit with sigmoidal equations and compared across treatment groups using Z tests. Aged rats that received intraventricular infusions of XPro1595 over a four week period (0.08 mg/kg/day) exhibited a slight, but significant increase in maximal EPSP amplitude, relative to the vehicle group (z = 2.17). Other curve parameters (i.e. slope and half-maximal FV) were not affected by XPro1595. C, In a twin pulse paradigm (50 ms interpulse interval), the EPSP slope corresponding to pulse 2 (S2) was expressed as a percentage (mean ± SEM) of the pulse 1 (S1) EPSP slope to obtain measures of paired-pulse facilitation (PPF). No treatment effect was observed for PPF (p = 0.27). D, Time plot showing normalized mean ± SEM EPSP slope amplitudes collected before (1) and after (2) the delivery of prolonged 1 Hz stimulation (bar, 900 consecutive pulses). The inset shows representative CA1 EPSP waveforms averaged in the pre- (1) and post-1 Hz (2) periods for vehicle and XPro1595 groups. Scale bars indicate 0.5 mV vertical by 2 ms horizontal. E, Bar graph shows the amount of LTD, expressed as a percentage of the pre-1 Hz baseline (mean ± SEM) in each treatment condition. The results revealed significant LTD in vehicle, but not in XPro1595-treated rats (* p<0.05, repeated measures ANOVA).
Figure 4. Effects of XPro1595 on glutamate…
Figure 4. Effects of XPro1595 on glutamate receptor protein levels. A,
Representative Western blots are shown for AMPA receptor (GluR1 and GluR2) and NMDA receptor subtypes (NR1, NR2A, and NR2B) in hippocampal membrane fractions from aged (22 month) rats treated for four weeks (intraventricular delivery) with vehicle or XPro1595 (0.08 mg/kg/day). The Na/K-ATPase loading control is shown below. B, XPro1595 treatment resulted in a selective increase in GluR1 levels (* p<0.05).
Figure 5. XPro1595 reduces L-VSCC activity in…
Figure 5. XPro1595 reduces L-VSCC activity in CA1 neurons of aged rats. A,
Left panel, Cartoon illustration of a partially dissociated hippocampal slice in which CA1 is “unzipped” along stratum pyramidale to expose CA1 neurons for patch clamp recording. Right panel, Photomicrograph showing a glass micropipette tip patched onto a CA1 pyramidal neuron in an “unzipped” slice. B, Three representative L-VSCC current traces and the average ensemble current (bottom trace) in cell attached patches from aged rats treated for six weeks (intrahippocampal delivery) with vehicle or XPro1595 (0.08 mg/kg/day). Traces were taken from 45–50 consecutive step depolarizations (150 ms duration) from −70 to +10 mV. C, Mean ± SEM current (I) densities (pA/μm2) for each treatment group are shown. XPro1595 significantly reduced VSCC I density nearly three-fold (* p<0.05). D–E, Representative Western blots (D) and mean+SEM protein levels (E) for the major pore-forming L-VSCC subunits (CaV1.2 and CaV1.3) from hippocampal membrane fractions. The Na/K-ATPase served as loading control. XPro1595 did not significantly alter either CaV1.2 (p = 0.17) or CaV1.3 (p = 0.63) levels.

References

    1. McCoy MK, Tansey MG. TNF signaling inhibition in the CNS: implications for normal brain function and neurodegenerative disease. J Neuroinflammation. 2008;5:45.
    1. Gavilan MP, Revilla E, Pintado C, Castano A, Vizuete ML, et al. Molecular and cellular characterization of the age-related neuroinflammatory processes occurring in normal rat hippocampus: potential relation with the loss of somatostatin GABAergic neurons. J Neurochem. 2007;103:984–996.
    1. Terao A, Apte-Deshpande A, Dousman L, Morairty S, Eynon BP, et al. Immune response gene expression increases in the aging murine hippocampus. J Neuroimmunol. 2002;132:99–112.
    1. Tha KK, Okuma Y, Miyazaki H, Murayama T, Uehara T, et al. Changes in expressions of proinflammatory cytokines IL-1beta, TNF-alpha and IL-6 in the brain of senescence accelerated mouse (SAM) P8. Brain Res. 2000;885:25–31.
    1. Dickson DW, Lee SC, Mattiace LA, Yen SH, Brosnan C. Microglia and cytokines in neurological disease, with special reference to AIDS and Alzheimer's disease. Glia. 1993;7:75–83.
    1. Macdonald NJ, Decorti F, Pappas TC, Taglialatela G. Cytokine/neurotrophin interaction in the aged central nervous system. J Anat 197 Pt. 2000;4:543–551.
    1. Barnum CJ, Tansey MG. The duality of TNF signaling outcomes in the brain: potential mechanisms? Exp Neurol. 2011;229:198–200.
    1. Foster TC. Calcium homeostasis and modulation of synaptic plasticity in the aged brain. Aging Cell. 2007;6:319–325.
    1. Thibault O, Gant JC, Landfield PW. Expansion of the calcium hypothesis of brain aging and Alzheimer's disease: minding the store. Aging Cell. 2007;6:307–317.
    1. Pickering M, O'Connor JJ. Pro-inflammatory cytokines and their effects in the dentate gyrus. Prog Brain Res. 2007;163:339–354.
    1. Furukawa K, Mattson MP. The transcription factor NF-kappaB mediates increases in calcium currents and decreases in NMDA- and AMPA/kainate-induced currents induced by tumor necrosis factor-alpha in hippocampal neurons. J Neurochem. 1998;70:1876–1886.
    1. Park KM, Yule DI, Bowers WJ. Tumor necrosis factor-alpha potentiates intraneuronal Ca2+ signaling via regulation of the inositol 1,4,5-trisphosphate receptor. J Biol Chem. 2008;283:33069–33079.
    1. Thibault O, Landfield PW. Increase in single L-type calcium channels in hippocampal neurons during aging. Science. 1996;272:1017–1020.
    1. Paredes D, Acosta S, Gemma C, Bickford PC. Role of TNFalpha Induced Inflammation in Delay Eyeblink Conditioning in Young and Aged Rats. Aging Dis. 2010;1:191–198.
    1. Zalevsky J, Secher T, Ezhevsky SA, Janot L, Steed PM, et al. Dominant-negative inhibitors of soluble TNF attenuate experimental arthritis without suppressing innate immunity to infection. J Immunol. 2007;179:1872–1883.
    1. Steed PM, Tansey MG, Zalevsky J, Zhukovsky EA, Desjarlais JR, et al. Inactivation of TNF signaling by rationally designed dominant-negative TNF variants. Science. 2003;301:1895–1898.
    1. Grell M, Wajant H, Zimmermann G, Scheurich P. The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci U S A. 1998;95:570–575.
    1. McAlpine FE, Lee JK, Harms AS, Ruhn KA, Blurton-Jones M, et al. Inhibition of soluble TNF signaling in a mouse model of Alzheimer's disease prevents pre-plaque amyloid-associated neuropathology. Neurobiol Dis. 2009;34:163–177.
    1. McCoy MK, Martinez TN, Ruhn KA, Szymkowski DE, Smith CG, et al. Blocking soluble tumor necrosis factor signaling with dominant-negative tumor necrosis factor inhibitor attenuates loss of dopaminergic neurons in models of Parkinson's disease. J Neurosci. 2006;26:9365–9375.
    1. Norris CM, Foster TC. MK-801 improves retention in aged rats: implications for altered neural plasticity in age-related memory deficits. Neurobiol Learn Mem. 1999;71:194–206.
    1. Foster TC, Sharrow KM, Masse JR, Norris CM, Kumar A. Calcineurin links Ca2+ dysregulation with brain aging. J Neurosci. 2001;21:4066–4073.
    1. Mathis DM, Furman JL, Norris CM. J Vis Exp; 2011. Preparation of Acute Hippocampal Slices from Rats and Transgenic Mice for the Study of Synaptic Alterations during Aging and Amyloid Pathology.
    1. Blalock EM, Chen KC, Vanaman TC, Landfield PW, Slevin JT. Epilepsy-induced decrease of L-type Ca2+ channel activity and coordinate regulation of subunit mRNA in single neurons of rat hippocampal ‘zipper’ slices. Epilepsy Res. 2001;43:211–226.
    1. Gray R, Fisher R, Spruston N, Johnston D. New York: Wiley; 1990. Preparations of the vertebrate central nervous system in vitro. pp. 3–23.
    1. Norris CM, Blalock EM, Chen KC, Porter NM, Thibault O, et al. Hippocampal ‘zipper’ slice studies reveal a necessary role for calcineurin in the increased activity of L-type Ca(2+) channels with aging. Neurobiol Aging. 2010;31:328–338.
    1. Thibault O, Pancani T, Landfield PW, Norris CM. Biochim Biophys Acta 1822: 546-549; 2012. Reduction in neuronal L-type calcium channel activity in a double knock-in mouse model of Alzheimer's disease. pp. 546–549.
    1. Sakmann B, Neher E. Geometric parameters of pipettes and membrane patches. In: Sakmann B, Neher E, editors. Single Channel Recording. New York: Plenum; 1983. pp. 37–51.
    1. Ohlsson H, Edlund T. Sequence-specific interactions of nuclear factors with the insulin gene enhancer. Cell. 1986;45:35–44.
    1. Marchetti L, Klein M, Schlett K, Pfizenmaier K, Eisel UL. Tumor necrosis factor (TNF)-mediated neuroprotection against glutamate-induced excitotoxicity is enhanced by N-methyl-D-aspartate receptor activation. Essential role of a TNF receptor 2-mediated phosphatidylinositol 3-kinase-dependent NF-kappa B pathway. J Biol Chem. 2004;279:32869–32881.
    1. Grell M, Douni E, Wajant H, Lohden M, Clauss M, et al. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell. 1995;83:793–802.
    1. McAlpine FE, Tansey MG. Neuroinflammation and tumor necrosis factor signaling in the pathophysiology of Alzheimer's disease. J Inflamm Res. 2008;1:29–39.
    1. Van Eldik LJ, Thompson WL, Ralay Ranaivo H, Behanna HA, Martin Watterson D. Glia proinflammatory cytokine upregulation as a therapeutic target for neurodegenerative diseases: function-based and target-based discovery approaches. Int Rev Neurobiol. 2007;82:277–296.
    1. Mrak RE, Griffin WS. Glia and their cytokines in progression of neurodegeneration. Neurobiol Aging. 2005;26:349–354.
    1. Barnes CA, Rao G, Foster TC, McNaughton BL. Region-specific age effects on AMPA sensitivity: electrophysiological evidence for loss of synaptic contacts in hippocampal field CA1. Hippocampus. 1992;2:457–468.
    1. Foster TC, Kumar A. Susceptibility to induction of long-term depression is associated with impaired memory in aged Fischer 344 rats. Neurobiol Learn Mem. 2007;87:522–535.
    1. Norris CM, Korol DL, Foster TC. Increased susceptibility to induction of long-term depression and long-term potentiation reversal during aging. J Neurosci. 1996;16:5382–5392.
    1. Norris CM, Halpain S, Foster TC. Reversal of age-related alterations in synaptic plasticity by blockade of L-type Ca2+ channels. J Neurosci. 1998;18:3171–3179.
    1. Norris CM, Scheff SW. Recovery of afferent function and synaptic strength in hippocampal CA1 following traumatic brain injury. J Neurotrauma. 2009;26:2269–2278.
    1. Vouimba RM, Foy MR, Foy JG, Thompson RF. 17beta-estradiol suppresses expression of long-term depression in aged rats. Brain Res Bull. 2000;53:783–787.
    1. Hsu KS, Huang CC, Liang YC, Wu HM, Chen YL, et al. Alterations in the balance of protein kinase and phosphatase activities and age-related impairments of synaptic transmission and long-term potentiation. Hippocampus. 2002;12:787–802.
    1. Thibault O, Hadley R, Landfield PW. Elevated postsynaptic [Ca2+]i and L-type calcium channel activity in aged hippocampal neurons: relationship to impaired synaptic plasticity. J Neurosci. 2001;21:9744–9756.
    1. Campbell LW, Hao SY, Thibault O, Blalock EM, Landfield PW. Aging changes in voltage-gated calcium currents in hippocampal CA1 neurons. J Neurosci. 1996;16:6286–6295.
    1. Brewer LD, Dowling AL, Curran-Rauhut MA, Landfield PW, Porter NM, et al. Estradiol reverses a calcium-related biomarker of brain aging in female rats. J Neurosci. 2009;29:6058–6067.
    1. Veng LM, Mesches MH, Browning MD. Age-related working memory impairment is correlated with increases in the L-type calcium channel protein alpha1D (Cav1.3) in area CA1 of the hippocampus and both are ameliorated by chronic nimodipine treatment. Brain Res Mol Brain Res. 2003;110:193–202.
    1. Chen KC, Blalock EM, Thibault O, Kaminker P, Landfield PW. Expression of alpha 1D subunit mRNA is correlated with L-type Ca2+ channel activity in single neurons of hippocampal “zipper” slices. Proc Natl Acad Sci U S A. 2000;97:4357–4362.
    1. Herman JP, Chen KC, Booze R, Landfield PW. Up-regulation of alpha1D Ca2+ channel subunit mRNA expression in the hippocampus of aged F344 rats. Neurobiol Aging. 1998;19:581–587.
    1. Foster TC. Regulation of synaptic plasticity in memory and memory decline with aging. Prog Brain Res. 2002;138:283–303.
    1. Foster TC, Norris CM. Age-associated changes in Ca(2+)-dependent processes: relation to hippocampal synaptic plasticity. Hippocampus. 1997;7:602–612.
    1. Rosenzweig ES, Barnes CA. Impact of aging on hippocampal function: plasticity, network dynamics, and cognition. Prog Neurobiol. 2003;69:143–179.
    1. Kumar A, Foster TC. Shift in induction mechanisms underlies an age-dependent increase in DHPG-induced synaptic depression at CA3 CA1 synapses. J Neurophysiol. 2007;98:2729–2736.
    1. Billard JM. Long-term depression in the hippocampal CA1 area of aged rats, revisited: contribution of temporal constraints related to slice preparation. PLoS One. 2010;5:e9843.
    1. Kumar A, Foster TC. Intracellular calcium stores contribute to increased susceptibility to LTD induction during aging. Brain Res. 2005;1031:125–128.
    1. Jouvenceau A, Dutar P. A role for the protein phosphatase 2B in altered hippocampal synaptic plasticity in the aged rat. J Physiol Paris. 2006;99:154–161.
    1. Foy MR, Baudry M, Foy JG, Thompson RF. 17beta-estradiol modifies stress-induced and age-related changes in hippocampal synaptic plasticity. Behav Neurosci. 2008;122:301–309.
    1. Mulkey RM, Malenka RC. Mechanisms underlying induction of homosynaptic long-term depression in area CA1 of the hippocampus. Neuron. 1992;9:967–975.
    1. Dudek SM, Bear MF. Homosynaptic long-term depression in area CA1 of hippocampus and effects of N-methyl-D-aspartate receptor blockade. Proc Natl Acad Sci U S A. 1992;89:4363–4367.
    1. Foster TC, Sharrow KM, Kumar A, Masse J. Interaction of age and chronic estradiol replacement on memory and markers of brain aging. Neurobiol Aging. 2003;24:839–852.
    1. Mulkey RM, Endo S, Shenolikar S, Malenka RC. Involvement of a calcineurin/inhibitor-1 phosphatase cascade in hippocampal long-term depression. Nature. 1994;369:486–488.
    1. Genoux D, Haditsch U, Knobloch M, Michalon A, Storm D, et al. Protein phosphatase 1 is a molecular constraint on learning and memory. Nature. 2002;418:970–975.
    1. Deyo RA, Straube KT, Disterhoft JF. Nimodipine facilitates associative learning in aging rabbits. Science. 1989;243:809–811.
    1. Wang JQ, Arora A, Yang L, Parelkar NK, Zhang G, et al. Phosphorylation of AMPA receptors: mechanisms and synaptic plasticity. Mol Neurobiol. 2005;32:237–249.
    1. Kessels HW, Malinow R. Synaptic AMPA receptor plasticity and behavior. Neuron. 2009;61:340–350.
    1. Beattie EC, Stellwagen D, Morishita W, Bresnahan JC, Ha BK, et al. Control of synaptic strength by glial TNFalpha. Science. 2002;295:2282–2285.
    1. Stellwagen D, Beattie EC, Seo JY, Malenka RC. Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-alpha. J Neurosci. 2005;25:3219–3228.
    1. Disterhoft JF, Moyer, Thompson LT. The calcium rationale in aging and Alzheimer's disease. Evidence from an animal model of normal aging. Ann N Y Acad Sci. 1994;747:382–406.
    1. Khachaturian ZS. The role of calcium regulation in brain aging: reexamination of a hypothesis. Aging (Milano) 1989;1:17–34.
    1. Landfield PW, Pitler TA. Prolonged Ca2+-dependent afterhyperpolarizations in hippocampal neurons of aged rats. Science. 1984;226:1089–1092.
    1. Stutzmann GE. The pathogenesis of Alzheimers disease is it a lifelong “calciumopathy”? Neuroscientist. 2007;13:546–559.
    1. Bezprozvanny I, Mattson MP. Neuronal calcium mishandling and the pathogenesis of Alzheimer's disease. Trends Neurosci. 2008;31:454–463.
    1. Chavis P, Fagni L, Lansman JB, Bockaert J. Functional coupling between ryanodine receptors and L-type calcium channels in neurons. Nature. 1996;382:719–722.
    1. Sukhareva M, Smith SV, Maric D, Barker JL. Functional properties of ryanodine receptors in hippocampal neurons change during early differentiation in culture. J Neurophysiol. 2002;88:1077–1087.
    1. Kumar A, Foster TC. Enhanced long-term potentiation during aging is masked by processes involving intracellular calcium stores. J Neurophysiol. 2004;91:2437–2444.
    1. Moyer, Disterhoft JF. Nimodipine decreases calcium action potentials in rabbit hippocampal CA1 neurons in an age-dependent and concentration-dependent manner. Hippocampus. 1994;4:11–17.
    1. McMonagle-Strucko K, Fanelli RJ. Enhanced acquisition of reversal training in a spatial learning task in rats treated with chronic nimodipine. Pharmacol Biochem Behav. 1993;44:827–835.
    1. Tancredi V, D'Arcangelo G, Grassi F, Tarroni P, Palmieri G, et al. Tumor necrosis factor alters synaptic transmission in rat hippocampal slices. Neurosci Lett. 1992;146:176–178.
    1. Cunningham AJ, Murray CA, O'Neill LA, Lynch MA, O'Connor JJ. Interleukin-1 beta (IL-1 beta) and tumour necrosis factor (TNF) inhibit long-term potentiation in the rat dentate gyrus in vitro. Neurosci Lett. 1996;203:17–20.
    1. Albensi BC, Mattson MP. Evidence for the involvement of TNF and NF-kappaB in hippocampal synaptic plasticity. Synapse. 2000;35:151–159.
    1. Aloe L, Fiore M, Probert L, Turrini P, Tirassa P. Overexpression of tumour necrosis factor alpha in the brain of transgenic mice differentially alters nerve growth factor levels and choline acetyltransferase activity. Cytokine. 1999;11:45–54.
    1. Golan H, Levav T, Mendelsohn A, Huleihel M. Involvement of tumor necrosis factor alpha in hippocampal development and function. Cereb Cortex. 2004;14:97–105.
    1. Davare MA, Hell JW. Increased phosphorylation of the neuronal L-type Ca(2+) channel Ca(v)1.2 during aging. Proc Natl Acad Sci U S A. 2003;100:16018–16023.
    1. Norris CM, Blalock EM, Chen KC, Porter NM, Landfield PW. Calcineurin enhances L-type Ca(2+) channel activity in hippocampal neurons: increased effect with age in culture. Neuroscience. 2002;110:213–225.
    1. Gant JC, Chen KC, Norris CM, Kadish I, Thibault O, et al. Disrupting function of FK506-binding protein 1b/12.6 induces the Ca(2)+-dysregulation aging phenotype in hippocampal neurons. J Neurosci. 2011;31:1693–1703.
    1. Kantrow SP, Gierman JL, Jaligam VR, Zhang P, Piantadosi CA, et al. Regulation of tumor necrosis factor cytotoxicity by calcineurin. FEBS Lett. 2000;483:119–124.
    1. Fernandez AM, Fernandez S, Carrero P, Garcia-Garcia M, Torres-Aleman I. Calcineurin in reactive astrocytes plays a key role in the interplay between proinflammatory and anti-inflammatory signals. J Neurosci. 2007;27:8745–8756.
    1. Zhang JM, Li H, Liu B, Brull SJ. Acute topical application of tumor necrosis factor alpha evokes protein kinase A-dependent responses in rat sensory neurons. J Neurophysiol. 2002;88:1387–1392.
    1. Bouwmeester T, Bauch A, Ruffner H, Angrand PO, Bergamini G, et al. A physical and functional map of the human TNF-alpha/NF-kappa B signal transduction pathway. Nat Cell Biol. 2004;6:97–105.
    1. Giordano A, Avellino R, Ferraro P, Romano S, Corcione N, et al. Rapamycin antagonizes NF-kappaB nuclear translocation activated by TNF-alpha in primary vascular smooth muscle cells and enhances apoptosis. Am J Physiol Heart Circ Physiol. 2006;290:H2459–2465.
    1. Furman JL, Artiushin IA, Norris CM. Disparate effects of serum on basal and evoked NFAT activity in primary astrocyte cultures. Neurosci Lett. 2010;469:365–369.
    1. Macdonald NJ, Delderfield SM, Zhang W, Taglialatela G. Tumour necrosis factor-alpha- vs. growth factor deprivation-promoted cell death: distinct converging pathways. Aging Cell. 2003;2:245–256.
    1. Hu W, Ralay Ranaivo H, Roy SM, Behanna HA, Wing LK, et al. Development of a novel therapeutic suppressor of brain proinflammatory cytokine up-regulation that attenuates synaptic dysfunction and behavioral deficits. Bioorg Med Chem Lett. 2007;17:414–418.
    1. Sama MA, Mathis DM, Furman JL, Abdul HM, Artiushin IA, et al. Interleukin-1beta-dependent signaling between astrocytes and neurons depends critically on astrocytic calcineurin/NFAT activity. J Biol Chem. 2008;283:21953–21964.
    1. Griffin R, Nally R, Nolan Y, McCartney Y, Linden J, et al. The age-related attenuation in long-term potentiation is associated with microglial activation. J Neurochem. 2006;99:1263–1272.
    1. Kotilinek LA, Westerman MA, Wang Q, Panizzon K, Lim GP, et al. Cyclooxygenase-2 inhibition improves amyloid-beta-mediated suppression of memory and synaptic plasticity. Brain. 2008;131:651–664.
    1. Floden AM, Li S, Combs CK. Beta-amyloid-stimulated microglia induce neuron death via synergistic stimulation of tumor necrosis factor alpha and NMDA receptors. J Neurosci. 2005;25:2566–2575.
    1. Vitkovic L, Bockaert J, Jacque C. “Inflammatory” cytokines: neuromodulators in normal brain? J Neurochem. 2000;74:457–471.

Source: PubMed

3
Subscribe