264
Views
51
CrossRef citations to date
0
Altmetric
Review

MAPK, β-amyloid and synaptic dysfunction: the role of RAGE

, , &
Pages 1635-1645 | Published online: 09 Jan 2014

References

  • Gandy S. The role of cerebral amyloid β accumulation in common forms of Alzheimer disease. J. Clin. Invest.115(5), 1121–1129 (2005).
  • Kametani F. ε-secretase: reduction of amyloid precursor protein ε-site cleavage in Alzheimer’s disease. Curr. Alzheimer Res.5(2), 165–171 (2008).
  • Eisenhauer PB, Johnson RJ, Wells JM, Davies TA, Fine RE. Toxicity of various amyloid β peptide species in cultured human blood–brain barrier endothelial cells: increased toxicity of dutch-type mutant. J. Neurosci. Res.60(6), 804–810 (2000).
  • Johnson LV, Leitner WP, Rivest AJ et al. The Alzheimer’s Aβ-peptide is deposited at sites of complement activation in pathologic deposits associated with aging and age-related macular degeneration. Proc. Natl Acad. Sci. USA99(18), 11830–11835 (2002).
  • Stromer T, Serpell LC. Structure and morphology of the Alzheimer’s amyloid fibril. Microsc. Res. Tech.67(3–4), 210–217 (2005).
  • Cirrito JR, May PC, O’Dell MA et al.In vivo assessment of brain interstitial fluid with microdialysis reveals plaque-associated changes in amyloid-β metabolism and half-life. J. Neurosci.23(26), 8844–8853 (2003).
  • Cirrito JR, Yamada KA, Finn MB et al. Synaptic activity regulates interstitial fluid amyloid-β levels in vivo. Neuron48(6), 913–922 (2005).
  • Cirrito JR, Kang JE, Lee J et al. Endocytosis is required for synaptic activity-dependent release of amyloid-β in vivo. Neuron58(1), 42–51 (2008).
  • Dubois B, Feldman HH, Jacova C et al. Research criteria for the diagnosis of Alzheimer’s disease: revising the NINCDS-ADRDA criteria. Lancet Neurol.6(8), 734–746 (2007).
  • Hsia AY, Masliah E, McConlogue L et al. Plaque-independent disruption of neural circuits in Alzheimer’s disease mouse models. Proc. Natl Acad. Sci. USA96(6), 3228–3233 (1999).
  • Chapman PF, White GL, Jones MW et al. Impaired synaptic plasticity and learning in aged amyloid precursor protein transgenic mice. Nat. Neurosci.2(3), 271–276 (1999).
  • Li QX, Maynard C, Cappai R et al. Intracellular accumulation of detergent-soluble amyloidogenic A β fragment of Alzheimer’s disease precursor protein in the hippocampus of aged transgenic mice. J. Neurochem.72(6), 2479–2487 (1999).
  • Larson J, Lynch G, Games D, Seubert P. Alterations in synaptic transmission and long-term potentiation in hippocampal slices from young and aged PDAPP mice. Brain Res.840(1–2), 23–35 (1999).
  • Giacchino J, Criado JR, Games D, Henriksen S. In vivo synaptic transmission in young and aged amyloid precursor protein transgenic mice. Brain Res.876(1–2), 185–190 (2000).
  • Wirths O, Multhaup G, Czech C et al. Intraneuronal Aβ accumulation precedes plaque formation in β-amyloid precursor protein and presenilin-1 double-transgenic mice. Neurosci Lett.306(1–2), 116–120 (2001).
  • Oddo S, Caccamo A, Shepherd JD et al. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron39(3), 409–421 (2003).
  • Ingelsson M, Fukumoto H, Newell KL et al. Early Aβ accumulation and progressive synaptic loss, gliosis, and tangle formation in AD brain. Neurology62(6), 925–931 (2004).
  • Robbins TW, Elliott R, Sahakian BJ. Neuropsychology – dementia and affective disorders. Br. Med. Bull.52(3), 627–643 (1996).
  • Bartus RT. On neurodegenerative diseases, models, and treatment strategies: lessons learned and lessons forgotten a generation following the cholinergic hypothesis. Exp. Neurol.163(2), 495–529 (2000).
  • Auld DS, Kornecook TJ, Bastianetto S, Quirion R. Alzheimer’s disease and the basal forebrain cholinergic system: relations to β-amyloid peptides, cognition, and treatment strategies. Prog. Neurobiol.68(3), 209–245 (2002).
  • Bartus RT, Dean RL 3rd. Pharmaceutical treatment for cognitive deficits in Alzheimer’s disease and other neurodegenerative conditions: exploring new territory using traditional tools and established maps. Psychopharmacology (Berl.), 202(1–3), 15–36 (2009).
  • Selkoe DJ. Alzheimer’s disease is a synaptic failure. Science298(5594), 789–791 (2002).
  • Arancio O, Zhang HP, Chen X et al. RAGE potentiates Aβ-induced perturbation of neuronal function in transgenic mice. EMBO J.23(20), 4096–4105 (2004).
  • Origlia N, Righi M, Capsoni S et al. Receptor for advanced glycation end product-dependent activation of p38 mitogen-activated protein kinase contributes to amyloid-β-mediated cortical synaptic dysfunction. J. Neurosci.28(13), 3521–3530 (2008).
  • Braak H, Braak E. Entorhinal–hippocampal interaction in mnestic disorders. Hippocampus3, 239–246 (1993).
  • Witter MP. Organization of the entorhinal–hippocampal system: a review of current anatomical data. Hippocampus3, 33–44 (1993).
  • Suzuki WA, Amaral DG. Functional neuroanatomy of the medial temporal lobe memory system. Cortex40(1), 220–222 (2004).
  • Braak H, Braak E. Demonstration of amyloid deposits and neurofibrillary changes in whole brain sections. Brain Pathol.1(3), 213–216 (1991).
  • Bliss TV, Collingridge GL. A synaptic model of memory: long-term potentiation in the hippocampus. Nature361(6407), 31–39 (1993).
  • Whitlock JR, Heynen AJ, Shuler MG, Bear MF. Learning induces long-term potentiation in the hippocampus. Science313(5790), 1093–1097 (2006).
  • Lambert MP, Barlow AK, Chromy BA et al. Diffusible, nonfibrillar ligands derived from Aβ1–42 are potent central nervous system neurotoxins. Proc. Natl Acad. Sci. USA95(11), 6448–6453 (1998).
  • Chen QS, Wei WZ, Shimahara T, Xie CW. Alzheimer amyloid β-peptide inhibits the late phase of long-term potentiation through calcineurin-dependent mechanisms in the hippocampal dentate gyrus. Neurobiol. Learn. Mem.77(3), 354–371 (2002).
  • Walsh DM, Klyubin I, Fadeeva JV et al. Naturally secreted oligomers of amyloid β protein potently inhibit hippocampal long-term potentiation in vivo. Nature416(6880), 535 (2002).
  • Zhao D, Watson JB, Xie CW. Amyloid β prevents activation of calcium/calmodulin-dependent protein kinase II and AMPA receptor phosphorylation during hippocampal long-term potentiation. J. Neurophysiol.92(5), 2853–2858 (2004).
  • Wang Q, Walsh DM, Rowan MJ, Selkoe DJ, Anwyl R. Block of long-term potentiation by naturally secreted and synthetic amyloid β-peptide in hippocampal slices is mediated via activation of the kinases c-Jun N-terminal kinase, cyclin-dependent kinase 5, and p38 mitogen-activated protein kinase as well as metabotropic glutamate receptor type 5. J. Neurosci.24(13), 3370–3378 (2004).
  • Walsh DM, Townsend M, Podlisny MB et al. Certain inhibitors of synthetic amyloid β-peptide (Aβ) fibrillogenesis block oligomerization of natural Aβ and thereby rescue long-term potentiation. J. Neurosci.25(10), 2455–2462 (2005).
  • Shankar GM, Li S, Mehta TH et al. Amyloid-β protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat. Med.14(8), 837–842 (2008).
  • Snyder EM, Nong Y, Almeida CG et al. Regulation of NMDA receptor trafficking by amyloid-β. Nat. Neurosci.8(8), 1051–1058 (2005).
  • Tyszkiewicz JP, Yan Z. β-Amyloid peptides impair PKC-dependent functions of metabotropic glutamate receptors in prefrontal cortical neurons. J. Neurophysiol.93(6), 3102–3111 (2005).
  • Hsieh H, Boehm J, Sato C et al. AMPAR removal underlies Aβ-induced synaptic depression and dendritic spine loss. Neuron52(5), 831–843 (2006).
  • Parameshwaran K, Sims C, Kanju P et al. Amyloid β-peptide Aβ(1–42) but not Aβ(1–40) attenuates synaptic AMPA receptor function. Synapse61(6), 367–374 (2007).
  • Yan SD, Chen X, Fu J et al. RAGE and amyloid-β peptide neurotoxicity in Alzheimer’s disease. Nature382(6593), 685–691 (1996).
  • Yan SD, Yan SF, Chen X et al. Non-enzymatically glycated tau in Alzheimer’s disease induces neuronal oxidant stress resulting in cytokine gene expression and release of amyloid β-peptide. Nat. Med.1(7), 693–699 (1995).
  • Schmidt AM, Yan SD, Yan SF, Stern DM. The biology of the receptor for advanced glycation end products and its ligands. Biochim. Biophys. Acta1498(2–3), 99–111 (2000).
  • Schmidt AM, Yan SD, Yan SF, Stern DM. The multiligand receptor RAGE as a progression factor amplifying immune and inflammatory responses. J. Clin. Invest.108(7), 949–955 (2001).
  • Yan SD, Bierhaus A, Nawroth PP, Stern DM. RAGE and Alzheimer’s disease: a progression factor for amyloid-β-induced cellular perturbation? J. Alzheimers Dis.16(4), 833–843 (2009)
  • Yeh CH, Sturgis L, Haidacher J et al. Requirement for p38 and p44/p42 mitogen-activated protein kinases in RAGE-mediated nuclear factor-kB transcriptional activation and cytokine secretion. Diabetes50(6), 1495–1504 (2001).
  • Li JH, Wang W, Huang XR et al. Advanced glycation end products induce tubular epithelial-myofibroblast transition through the RAGE-ERK1/2 MAP kinase signaling pathway. Am. J. Pathol.164(4), 1389–1397 (2004).
  • Chen X, Walker DG, Schmidt AM, Arancio O, Lue LF, Yan SD. RAGE: a potential target for Aβ-mediated cellular perturbation in Alzheimer’s disease. Curr. Mol. Med.7(8), 735–742 (2007).
  • Bucciarelli LG, Wendt T, Rong L et al. RAGE is a multiligand receptor of the immunoglobulin superfamily: implications for homeostasis and chronic disease. Cell. Mol. Life Sci.59(7), 1117–1128 (2002).
  • Ishihara K, Tsutsumi K, Kawane S, Nakajima M, Kasaoka T. The receptor for advanced glycation end-products (RAGE) directly binds to ERK by a D-domain-like docking site. FEBS Lett.550(1–3), 107–113 (2003).
  • Lue LF, Walker DG, Brachova L et al. Involvement of microglial receptor for advanced glycation end products (RAGE) in Alzheimer’s disease: identification of a cellular activation mechanism. Exp. Neurol.171(1), 29–45 (2001).
  • Deane R, Du Yan S, Submamaryan RK et al. RAGE mediates amyloid-β peptide transport across the blood-brain barrier and accumulation in brain. Nat. Med.9(7), 907–913 (2003).
  • Mucke L, Yu GQ, McConlogue L et al. Astroglial expression of human α(1)-antichymotrypsin enhances Alzheimer-like pathology in amyloid protein precursor transgenic mice. Am. J. Pathol.157(6), 2003–2010 (2000).
  • Simm A, Casselmann C, Schubert A et al. Age associated changes of AGE-receptor expression: RAGE upregulation is associated with human heart dysfunction. Exp. Gerontol.39(3), 407–413 (2004).
  • Ding Q, Keller JN. Evaluation of rage isoforms, ligands, and signaling in the brain. Biochim. Biophys. Acta1746(1), 18–27 (2005).
  • Chaney MO, Stine WB, Kokjohn TA et al. RAGE and amyloid β interactions: atomic force microscopy and molecular modelling. Biochim. Biophys. Acta1741(1–2), 199–205 (2005).
  • Thomas GM, Huganir RL. MAPK cascade signalling and synaptic plasticity. Nat. Rev. Neurosci.5(3), 173–183 (2004).
  • Costa M, Marchi M, Cardarelli F et al. Dynamic regulation of ERK2 nuclear translocation and mobility in living cells. J. Cell Sci.119(23), 4952–4963 (2006).
  • Marchi M, D’Antoni A, Formentini I et al. The N-terminal domain of ERK1 accounts for the functional differences with ERK2. PLoS One3(12), e3873 (2008).
  • Selcher JC, Nekrasova T, Paylor R, Landreth GE, Sweatt JD. Mice lacking the ERK1 isoform of MAP kinase are unimpaired in emotional learning. Learn. Mem.8(1), 11–19 (2001).
  • Mazzucchelli C, Vantaggiato C, Ciamei A et al. Knockout of ERK1 MAP kinase enhances synaptic plasticity in the striatum and facilitates striatal-mediated learning and memory. Neuron34(5), 807–820 (2002).
  • Aouadi M, Binetruy B, Caron L, Le Marchand-Brustel Y, Bost F. Role of MAPKs in development and differentiation: lessons from knockout mice. Biochimie88(9), 1091–1098 (2006).
  • Sweatt JD. Mitogen-activated protein kinases in synaptic plasticity and memory. Curr. Opin. Neurobiol.14(3), 311–317 (2004).
  • Costa RM, Silva AJ. Molecular and cellular mechanisms underlying the cognitive deficits associated with neurofibromatosis 1. J. Child. Neurol.17(8), 622–651 (2002).
  • Kanterewicz BI, Urban NN, McMahon DB et al. The extracellular signal-regulated kinase cascade is required for NMDA receptor-independent LTP in area CA1 but not area CA3 of the hippocampus. J. Neurosci.20(9), 3057–3066 (2000).
  • Coogan AN, O’Leary DM, O’Connor JJ. P42/44 MAP kinase inhibitor PD98059 attenuates multiple forms of synaptic plasticity in rat dentate gyrus in vitro. J. Neurophysiol.81(1), 103–110 (1999).
  • Huang YY, Martin KC, Kandel ER. Both protein kinase A and mitogen-activated protein kinase are required in the amygdala for the macromolecular synthesis-dependent late phase of long-term potentiation. J. Neurosci.20(17), 6317–6325 (2000).
  • Schafe GE, Atkins CM, Swank MW et al. Activation of ERK/MAP kinase in the amygdala is required for memory consolidation of pavlovian fear conditioning. J. Neurosci.20(21), 8177–8187 (2000).
  • Di Cristo G, Berardi N, Cancedda L et al. Requirement of ERK activation for visual cortical plasticity. Science292(5525), 2337–2340 (2001).
  • English JD, Sweatt JD. Activation of p42 mitogen-activated protein kinase in hippocampal long term potentiation. J. Biol. Chem.271(40), 24329–24332 (1996).
  • Impey S, Obrietan K, Wong ST et al. Cross talk between ERK and PKA is required for Ca2+ stimulation of CREB-dependent transcription and ERK nuclear translocation. Neuron21(4), 869–883 (1998).
  • Pages G, Guerin S, Grall D et al. Defective thymocyte maturation in p44 MAP kinase (Erk 1) knockout mice. Science286(5443), 1374–1377 (1999).
  • Samuels IS, Karlo JC, Faruzzi AN et al. Deletion of ERK2 mitogen-activated protein kinase identifies its key roles in cortical neurogenesis and cognitive function. J. Neurosci.28(27), 6983–6995 (2008).
  • Zhu JJ, Qin Y, Zhao M, Van Aelst L, Malinow R. Ras and Rap control AMPA receptor trafficking during synaptic plasticity. Cell110(4), 443–455 (2002).
  • Webster B, Hansen L, Adame A et al. Astroglial activation of extracellular-regulated kinase in early stages of Alzheimer disease. J. Neuropathol. Exp. Neurol.65(2), 142–151 (2006).
  • Dineley KT, Westerman M, Bui D et al. B-amyloid activates the mitogen-activated protein kinase cascade via hippocampal a7 nicotinic acetylcholine receptors: in vitro and in vivo mechanisms related to Alzheimer’s disease. J. Neurosci.21(12), 4125–4133 (2001).
  • Ferreira A, Lu Q, Orecchio L, Kosik KS. Selective phosphorylation of adult tau isoforms in mature hippocampal neurons exposed to fibrillar Aβ. Mol. Cell. Neurosci.9(3), 220–234 (1997).
  • Rapoport M, Ferreira A. PD98059 prevents neurite degeneration induced by fibrillar β-amyloid in mature hippocampal neurons. J. Neurochem.74(1), 125–133 (2000).
  • Chong YH, Shin YJ, Lee EO et al. ERK1/2 activation mediates Aβ oligomer-induced neurotoxicity via caspase-3 activation and tau cleavage in rat organotypic hippocampal slice cultures. J. Biol. Chem.281(29), 20315–20325 (2006).
  • Zhuang S, Schnellmann RG. A death-promoting role for extracellular signal-regulated kinase. J. Pharmacol. Exp. Ther.319(3), 991–997 (2006).
  • Stanciu M, Wang Y, Kentor R et al. Persistent activation of ERK contributes to glutamate-induced oxidative toxicity in a neuronal cell line and primary cortical neuron cultures. J. Biol. Chem.275(16), 12200–12206 (2000).
  • Townsend M, Mehta T, Selkoe DJ. Soluble Aβ inhibits specific signal transduction cascades common to the insulin receptor pathway. J. Biol. Chem.282(46), 33305–33312 (2007).
  • Sturchler E, Galichet A, Weibel M, Leclerc E, Heizmann CW. Site-specific blockade of RAGE-Vd prevents amyloid-β oligomer neurotoxicity. J. Neurosci.28(20), 5149–5158 (2008).
  • Goedert M, Cuenda A, Craxton M, Jakes R, Cohen P. Activation of the novel stress-activated protein kinase SAPK4 by cytokines and cellular stresses is mediated by SKK3 (MKK6); comparison of its substrate specificity with that of other SAP kinases. EMBO J.16(12), 3563–3571 (1997).
  • Cuenda A, Cohen P, Buee-Scherrer V, Goedert M. Activation of stress-activated protein kinase-3 (SAPK3) by cytokines and cellular stresses is mediated via SAPKK3 (MKK6); comparison of the specificities of SAPK3 and SAPK2 (RK/p38). EMBO J.16(2), 295–305 (1997).
  • Kuma Y, Sabio G, Bain J et al. BIRB796 inhibits all p38 MAPK isoforms in vitro and in vivo. J. Biol. Chem.280(20), 19472–19479 (2005).
  • Cohen PS, Schmidtmayerova H, Dennis J et al. The critical role of p38 MAP kinase in T cell HIV-1 replication. Mol. Med.3(5), 339–346 (1997).
  • Kyriakis JM, Avruch J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol. Rev.81(2), 807–869 (2001).
  • Cuenda A, Rousseau S. p38 MAP-kinases pathway regulation, function and role in human diseases. Biochim. Biophys. Acta1773(8), 1358–1375 (2007).
  • Bolshakov VY, Carboni L, Cobb MH, Siegelbaum SA, Belardetti F. Dual MAP kinase pathways mediate opposing forms of long-term plasticity at CA3–CA1 synapses. Nat. Neurosci.3(11), 1107–1112 (2000).
  • Xia Z, Dickens M, Raingeaud J, Davis RJ, Greenberg ME. Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science270(5240), 1326–1331 (1995).
  • Kawasaki H, Morooka T, Shimohama S et al. Activation and involvement of p38 mitogen-activated protein kinase in glutamate-induced apoptosis in rat cerebellar granule cells. J. Biol. Chem.272(30), 18518–18521 (1997).
  • Li Y, Liu L, Barger SW, Griffin WS. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J. Neurosci.23(5), 1605–1611 (2003).
  • Kim SH, Smith CJ, Van Eldik LJ. Importance of MAPK pathways for microglial proinflammatory cytokine IL-1β production. Neurobiol. Aging25(4), 431–439 (2004).
  • Culbert AA, Skaper SD, Howlett DR et al. MAPK-activated protein kinase 2 deficiency in microglia inhibits proinflammatory mediator release and resultant neurotoxicity. Relevance to neuroinflammation in a transgenic mouse model of Alzheimer disease. J. Biol. Chem.281(33), 23658–23667 (2006).
  • Zhu X, Mei M, Lee HG et al. P38 activation mediates amyloid-β cytotoxicity. Neurochem. Res.30(6–7), 791–796 (2005).
  • Troy CM, Rabacchi SA, Xu Z et al. β-amyloid-induced neuronal apoptosis requires c-Jun N-terminal kinase activation. J. Neurochem.77(1), 157–164 (2001).
  • Coogan AN, O’Neill LA, O’Connor JJ. The P38 mitogen-activated protein kinase inhibitor SB203580 antagonizes the inhibitory effects of interleukin-1β on long-term potentiation in the rat dentate gyrus in vitro. Neuroscience93(1), 57–69 (1999).
  • Kelly A, Vereker E, Nolan Y et al. Activation of p38 plays a pivotal role in the inhibitory effect of lipopolysaccharide and interleukin-1 β on long term potentiation in rat dentate gyrus. J. Biol. Chem.278(21), 19453–19462 (2003).
  • Lali FV, Hunt AE, Turner SJ, Foxwell BM. The pyridinyl imidazole inhibitor SB203580 blocks phosphoinositide-dependent protein kinase activity, protein kinase B phosphorylation, and retinoblastoma hyperphosphorylation in interleukin-2-stimulated T cells independently of p38 mitogen-activated protein kinase. J. Biol. Chem.275, 7395–7402 (2000).
  • Bennett BL, Sasaki DT, Murray BW et al. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc. Natl Acad. Sci. USA98, 13681–13686 (2001).
  • Munoz L, Ranaivo HR, Roy SM et al. A novel p38 α MAPK inhibitor suppresses brain proinflammatory cytokine up-regulation and attenuates synaptic dysfunction and behavioral deficits in an Alzheimer’s disease mouse model. J. Neuroinflam.4(21), 1–14 (2007).
  • Yan SD, Schmidt AM, Anderson GM et al. Enhanced cellular oxidant stress by the interaction of advanced glycation end products with their receptors/binding proteins. J. Biol. Chem.269(13), 9889–9897 (1994).
  • Pulverer BJ, Kyriakis JM, Avruch J, Nikolakaki E, Woodgett JR. Phosphorylation of c-jun mediated by MAP kinases. Nature353(6345), 670–674 (1991).
  • Derijard B, Hibi M, Wu IH et al. JNK1: a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain. Cell76(6), 1025–1037 (1994).
  • Yang D, Tournier C, Wysk M et al. Targeted disruption of the MKK4 gene causes embryonic death, inhibition of c-Jun NH2-terminal kinase activation, and defects in AP-1 transcriptional activity. Proc. Natl Acad. Sci. USA94(7), 3004–3009 (1997).
  • Kallunki T, Su B, Tsigelny I et al. JNK2 contains a specificity-determining region responsible for efficient c-Jun binding and phosphorylation. Genes Dev.8(24), 2996–3007 (1994).
  • Gupta S, Barrett T, Whitmarsh AJ et al. Selective interaction of JNK protein kinase isoforms with transcription factors. EMBO J.15(11), 2760–2770 (1996).
  • Chang JS, Wendt T, Qu W et al. Oxygen deprivation triggers upregulation of early growth response-1 by the receptor for advanced glycation end products. Circ. Res.102(8), 905–913 (2008).
  • Chen JT, Lu DH, Chia CP et al. Impaired long-term potentiation in c-Jun N-terminal kinase 2-deficient mice. J. Neurochem.93(2), 463–473 (2005).
  • Carletti R, Tacconi S, Bettini E, Ferraguti F. Stress activated protein kinases, a novel family of mitogen-activated protein kinases, are heterogeneously expressed in the adult rat brain and differentially distributed from extracellular-signal-regulated protein kinases. Neuroscience69(4), 1103–1110 (1995).
  • Kumagae Y, Zhang Y, Kim OJ, Miller CA. Human c-Jun N-terminal kinase expression and activation in the nervous system. Brain Res. Mol. Brain Res.67(1), 10–17 (1999).
  • Mielke K, Herdegen T. JNK and p38 stresskinases – degenerative effectors of signal-transduction-cascades in the nervous system. Prog. Neurobiol.61(1), 45–60 (2000).
  • Curran BP, Murray HJ, O’Connor JJ. A role for c-Jun N-terminal kinase in the inhibition of long-term potentiation by interleukin-1β and long-term depression in the rat dentate gyrus in vitro. Neuroscience118(2), 347–357 (2003).
  • Li XM, Li CC, Yu SS et al. JNK1 contributes to metabotropic glutamate receptor-dependent long-term depression and short-term synaptic plasticity in the mice area hippocampal CA1. Eur. J. Neurosci.25(2), 391–396 (2007).
  • Xu X, Raber J, Yang D, Su B, Mucke L. Dynamic regulation of c-Jun N-terminal kinase activity in mouse brain by environmental stimuli. Proc. Natl Acad. Sci. USA94(23), 12655–12660 (1997).
  • Kuan CY, Burke RE. Targeting the JNK signaling pathway for stroke and Parkinson’s diseases therapy. Curr. Drug Targets CNS Neurol. Disord.4(1), 63–67 (2005).
  • Silva RM, Kuan CY, Rakic P, Burke RE. Mixed lineage kinase-c-jun N-terminal kinase signaling pathway: a new therapeutic target in Parkinson’s disease. Mov. Disord.20(6), 653–664 (2005).
  • Braak H, Braak E. Morphological criteria for the recognition of Alzheimer’s disease and the distribution pattern of cortical changes related to this disorder. Neurobiol. Aging15(3), 355–380 (1994).
  • Pei JJ, Braak E, Braak H et al. Localization of active forms of C-jun kinase (JNK) and p38 kinase in Alzheimer’s disease brains at different stages of neurofibrillary degeneration. J. Alzheimers Dis.3(1), 41–48 (2001).
  • Shoji M, Iwakami N, Takeuchi S et al. JNK activation is associated with intracellular β-amyloid accumulation. Brain Res. Mol. Brain Res.85(1–2), 221–233 (2000).
  • Morishima Y, Gotoh Y, Zieg J et al. B-amyloid induces neuronal apoptosis via a mechanism that involves the c-Jun N-terminal kinase pathway and the induction of Fas ligand. J. Neurosci.21(19), 7551–7560 (2001).
  • Jang JH, Surh YJ. β-Amyloid induces oxidative DNA damage and cell death through activation of c-Jun N terminal kinase. Ann. NY Acad. Sci.973, 228–236 (2002).
  • Wei W, Norton DD, Wang X, Kusiak JW. Aβ 17–42 in Alzheimer’s disease activates JNK and caspase-8 leading to neuronal apoptosis. Brain125(Pt 9), 2036–2043 (2002).
  • Fogarty MP, Downer EJ, Campbell V. A role for c-Jun N-terminal kinase 1 (JNK1), but not JNK2, in the β-amyloid-mediated stabilization of protein p53 and induction of the apoptotic cascade in cultured cortical neurons. Biochem. J.371(Pt 3), 789–798 (2003).
  • Minogue AM, Schmid AW, Fogarty MP et al. Activation of the c-Jun N-terminal kinase signaling cascade mediates the effect of amyloid-β on long term potentiation and cell death in hippocampus: a role for interleukin-1b? J. Biol. Chem.278(30), 27971–27980 (2003).
  • Bell KA, O’Riordan KJ, Sweatt JD, Dineley KT. MAPK recruitment by β-amyloid in organotypic hippocampal slice cultures depends on physical state and exposure time. J. Neurochem.91(2), 349–361 (2004).
  • Taguchi A, Blood DC, del Toro G et al. Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature405(6784), 354–360 (2000).
  • Chang L, Jones Y, Ellisman MH, Goldstein LS, Karin M. JNK1 is required for maintenance of neuronal microtubules and controls phosphorylation of microtubule-associated proteins. Dev Cell.4(4), 521–533 (2003).
  • Bianchi R, Giambanco I, Donato R. S100B/RAGE-dependent activation of microglia via NF-κB and AP-1 coregulation of COX-2 expression by S100B, IL-1β and TNF-α. Neurobiol. Aging (2008).
  • Minogue AM, Lynch AM, Loane DJ, Herron CE, Lynch MA. Modulation of amyloid-β-induced and age-associated changes in rat hippocampus by eicosapentaenoic acid. J. Neurochem.103(3), 914–926 (2007).
  • Birks J. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Database Syst. Rev.1, CD005593 (2006).
  • Lipton SA. Paradigm shift in NMDA receptor antagonist drug development: molecular mechanism of uncompetitive inhibition by memantine in the treatment of Alzheimer’s disease and other neurologic disorders. J. Alzheimers Dis.6(6 Suppl.), S61–S74 (2004).
  • Klyubin I, Wang Q, Reed MN et al. Protection against Aβ-mediated rapid disruption of synaptic plasticity and memory by memantine. Neurobiol. Aging (2009) (Epub ahead of print).
  • Lazarov O, Marr RA. Neurogenesis and Alzheimer’s disease: at the crossroads. Exp. Neurol. DOI: 10.1016/j.expneurol.2009.08.009 (2009) (Epub ahead of print).
  • Wang JM, Irwin RW, Liu L, Chen S, Brinton RD. Regeneration in a degenerating brain: potential of allopregnanolone as a neuroregenerative agent. Curr. Alzheimer Res.4(5), 510–517 (2007).
  • Brinton RD, Wang JM. Therapeutic potential of neurogenesis for prevention and recovery from Alzheimer’s disease: alloprenanaolone as a proof of concept neurogenic agent. Curr. Alzheimer Res.3(3), 185–190 (2006).
  • Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid β-peptide. Nat. Rev. Mol. Cell Biol.8(2), 101–112 (2007).
  • Tomita T. At the frontline of Alzheimer’s disease treatment: γ-secretase inhibitor/modulator mechanism. Naunyn Schmiedebergs Arch. Pharmacol.377(4–6), 295–300 (2008).
  • Puzzo D, Privitera L, Leznik E et al. Picomolar amyloid-β positively modulates synaptic plasticity and memory in hippocampus. J. Neurosci.28(53), 14537–14545 (2008).
  • Federoff HJ. Development of vaccination approaches for the treatment of neurological diseases. J. Comp. Neurol.515(1), 4–14 (2009).
  • Giuffrida ML, Caraci F, Pignataro B et al. β-amyloid monomers are neuroprotective. J. Neurosci.29(34), 10582–10587 (2009).
  • Atwood CS, Obrenovich ME, Liu T et al. Amyloid-β: a chameleon walking two worlds: a review of the trophic and toxic properties of amyloid-β. Brain Res. Brain Res. Rev.46(1), 118–120 (2003).
  • Rowan MJ, Klyubin I, Wang Q, Anwyl R. Mechanisms of the inhibitory effects of amyloid β-protein on synaptic plasticity. Exp. Gerontol.39(11–12), 1661–1667 (2004).
  • Zhu X, Rottkamp CA, Hartzler A et al. Activation of MKK6, an upstream activator of p38, in Alzheimer’s disease. J. Neurochem.79(2), 311–318 (2001).
  • Giovannini MG, Scali C, Prosperi C et al. β-amyloid-induced inflammation and cholinergic hypofunction in the rat brain in vivo: involvement of the p38MAPK pathway. Neurobiol. Dis.11, 257–274 (2002).
  • Hensley K, Floyd RA, Zheng NY et al. P38 kinase is activated in the Alzheimer’s disease brain. J. Neurochem.72, 2053–2058 (1999).
  • Sun A, Liu M, Nguyen XV, Bing G. P38 MAP kinase is activated at early stages in Alzheimer’s disease brain. Exp. Neurol.183, 394–405 (2003).
  • Swatton JE, Sellers LA, Faull RL, Holland A, Iritani S, Bahn S. Increased MAP kinase activity in Alzheimer’s and Down syndrome but not in schizophrenia human brain. Eur. J. Neurosci.19, 2711–2719 (2004).
  • Savage MJ, Lin YG, Ciallella JR, Flood DG, Scott RW. Activation of c-Jun N-terminal kinase and p38 in an Alzheimer’s disease model is associated with amyloid deposition. J. Neurosci.22, 3376–3385 (2002).
  • Ferrer I. Stress kinases involved in tau phosphorylation in Alzheimer’s disease, tauopathies and APP transgenic mice. Neurotox. Res.6, 469–475 (2004).
  • Webster J, Andrews R, Shen J et al. Small-molecule inhibitors of the receptor for advanced glycation end-products (RAGE) are an effective therapy in animal models of Alzheimer’s disease. Program and abstracts of the ICAD 2008: Alzheimer’s Association International Conference on Alzheimer’s Disease. Chicago, IL, USA, 26–31 July 2008.
  • Sabbagh MN, Bell J, Agro A, Aisen P, Galasko D. An oral antagonist of the receptor for advanced glycation end-products (RAGE) is safe and well-tolerated in the treatment of Alzheimer’s disease: Results from a Phase II study. Program and abstracts of the ICAD 2008: Alzheimer’s Association International Conference on Alzheimer’s Disease. Chicago, IL, USA, 26–31 July 2008

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.