全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Calpain Dysregulation in Alzheimer’s Disease

DOI: 10.5402/2012/728571

Full-Text   Cite this paper   Add to My Lib

Abstract:

Alzheimer’s disease (AD) is characterized by the presence of senile plaques and neurofibrillary tangles in the neocortex and hippocampus of AD patients. In addition, a marked decrease in synaptic contacts has been detected in these affected brain areas. Due to its prevalence in the aging population, this disease has been the focus of numerous studies. The data obtained from those studies suggest that the mechanisms leading to the formation of the hallmark lesions of AD might be linked. One of such mechanisms seems to be the dysregulation of calcium homeostasis that results in the abnormal activation of calpains. Calpains are a family of Ca2+-dependent cysteine proteases that play a key role in multiple cell functions including cell development, differentiation and proliferation, axonal guidance, growth cone motility, and cell death, among others. In this paper, we briefly reviewed data on the structure of these proteases and their regulation under normal conditions. We also summarized data underscoring the participation of calpains in the neurodegenerative mechanisms associated with AD. 1. Introduction Alzheimer’s disease (AD) is the most common cause of dementia in the aging population. This disease develops over time and leads to significant cognitive deficits affecting memory, insight, judgment, abstraction, and language functions [1]. AD affects more than 5 million people in the United States and this number is projected to rise to 35 million by 2050 [2, 3]. This estimate underscores both the scope of this health care issue for the society as a whole and the need for the development of therapeutic options for these patients. The diagnosis of this neurodegenerative disease relies on the presence of senile plaques and neurofibrillary tangles in affected brain areas at autopsy. These AD hallmark lesions are the results of the pathological deposition of proteins normally present throughout the brain. Senile plaques are composed of extracellular deposits of beta-amyloid (Aβ) derived by proteolytic cleavage from the amyloid precursor protein (APP) [4–10]. Neurofibrillary tangles, on the other hand, are intracellular bundles of self-assembled tau proteins [11–38]. The formation of both senile plaques and neurofibrillary tangles is associated with progressive and irreversible degeneration of neuronal processes and the loss of synaptic connections [39–50]. Initially, multiple studies focused on defining the characteristics of AD and on the analysis of the composition of senile plaques and neurofibrillary tangles. More recently, data have been obtained on the

References

[1]  M. J. Chiu, T. F. Chen, P. K. Yip, M. S. Hua, and L. Y. Tang, “Behavioral and psychologic symptoms in different types of dementia,” Journal of the Formosan Medical Association, vol. 105, no. 7, pp. 556–562, 2006.
[2]  L. E. Hebert, P. A. Scherr, J. L. Bienias, D. A. Bennett, and D. A. Evans, “Alzheimer disease in the US population: prevalence estimates using the 2000 census,” Archives of Neurology, vol. 60, no. 8, pp. 1119–1122, 2003.
[3]  I. Mebane-Sims, “Alzheimer’s disease facts and figures,” Alzheimer’s Dement, vol. 5, pp. 234–270, 2009.
[4]  G. G. Glenner and C. W. Wong, “Alzheimer's disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein,” Biochemical and Biophysical Research Communications, vol. 120, no. 3, pp. 885–890, 1984.
[5]  C. Haass and D. J. Selkoe, “Cellular processing of β-amyloid precursor protein and the genesis of amyloid β-peptide,” Cell, vol. 75, no. 6, pp. 1039–1042, 1993.
[6]  B. A. Yankner and M. M. Mesulam, “β-amyloid and the pathogenesis of Alzheimer's disease,” The New England Journal of Medicine, vol. 325, no. 26, pp. 1849–1857, 1991.
[7]  J. H. K. Tam and S. H. Pasternak, “Amyloid and Alzheimer’s disease: inside and out,” Canadian Journal of Neurological Sciences, vol. 39, pp. 286–289, 2012.
[8]  D. J. Selkoe, “Toward a comprehensive theory for Alzheimer's disease. Hypothesis: Alzheimer's disease is caused by the cerebral accumulation and cytotoxicity of amyloid β-protein,” Annals of the New York Academy of Sciences, vol. 924, pp. 17–25, 2000.
[9]  T. E. Golde, “The Aβ hypothesis: leading us to rationally-designed therapeutic strategies for the treatment or prevention of Alzheimer disease,” Brain Pathology, vol. 15, no. 1, pp. 84–87, 2005.
[10]  S. Sinha and I. Lieberburg, “Cellular mechanisms of β-amyloid production and secretion,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 20, pp. 11049–11053, 1999.
[11]  H. M. Wisniewski, H. K. Narang, and R. D. Terry, “Neurofibrillary tangles of paired helical filaments,” Journal of the Neurological Sciences, vol. 27, no. 2, pp. 173–181, 1976.
[12]  M. Goedert, C. M. Wischik, R. A. Crowther, J. E. Walker, and A. Klug, “Cloning and sequencing of the cDNA encoding a core protein of the paired helical filament of Alzheimer disease: identification as the microtubule-associated protein tau,” Proceedings of the National Academy of Sciences of the United States of America, vol. 85, no. 11, pp. 4051–4055, 1988.
[13]  M. Goedert, M. G. Spillantini, R. Jakes, D. Rutherford, and R. A. Crowther, “Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer's disease,” Neuron, vol. 3, no. 4, pp. 519–526, 1989.
[14]  M. Goedert, R. Jakes, R. A. Crowther et al., “The abnormal phosphorylation of tau protein at Ser-202 in Alzheimer disease recapitulates phosphorylation during development,” Proceedings of the National Academy of Sciences of the United States of America, vol. 90, no. 11, pp. 5066–5070, 1993.
[15]  A. del C. Alonso, I. Grundke-Iqbal, and K. Iqbal, “Alzheimer's disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules,” Nature Medicine, vol. 2, no. 7, pp. 783–787, 1996.
[16]  I. Grundke-Iqbal, K. Iqbal, M. Quinlan, Y.-C. Tung, M. S. Zaidi, and H. M. Wisniewski, “Microtubule-associated protein tau. A component of Alzheimer paired helical filaments,” The Journal of Biological Chemistry, vol. 261, no. 13, pp. 6084–6089, 1986.
[17]  I. Grundke-Iqbal, A. W. Vorbrodt, K. Iqbal, Y. C. Tung, G. P. Wang, and H. M. Wisniewski, “Microtubule-associated polypeptides tau are altered in Alzheimer paired helical filaments,” Molecular Brain Research, vol. 4, no. 1, pp. 43–52, 1988.
[18]  I. Grundke-Iqbal, K. Iqbal, Y.-C. Tung, M. Quinlan, H. M. Wisniewski, and L. I. Binder, “Abnormal phosphorylation of the microtubule-associated protein τ (tau) in Alzheimer cytoskeletal pathology,” Proceedings of the National Academy of Sciences of the United States of America, vol. 83, no. 13, pp. 44913–4917, 1986.
[19]  A. L. Guillozet, S. Weintraub, D. C. Mash, and M. M. Mesulam, “Neurofibrillary tangles, amyloid, and memory in aging and mild cognitive impairment,” Archives of Neurology, vol. 60, no. 5, pp. 729–736, 2003.
[20]  K. Iqbal, A. del C. Alonso, S. Chen et al., “Tau pathology in Alzheimer disease and other tauopathies,” Biochimica et Biophysica Acta, vol. 1739, no. 2, pp. 198–210, 2005.
[21]  K. S. Kosik, C. L. Joachim, and D. J. Selkoe, “Microtubule-associated protein tau (τ) is a major antigenic component of paired helical filaments in Alzheimer disease,” Proceedings of the National Academy of Sciences of the United States of America, vol. 83, no. 11, pp. 4044–4048, 1986.
[22]  K. S. Kosik and S. M. Greemberg, “Tau protein and Alzheimer’s disease,” in Alzheimer’s Disease, R. Terry, R. Katzman, and K. L. Bick, Eds., pp. 335–344, Raven Press Ltd, New York, NY, USA, 1994.
[23]  H. Braak, E. Braak, and E. M. Mandelkow, “A sequence of cytoskeleton changes related to the formation of neurofibrillary tangles and neuropil threads,” Acta Neuropathologica, vol. 87, no. 6, pp. 554–567, 1994.
[24]  V. M.-Y. Lee, B. J. Balin, L. Otvos, and J. Q. Trojanowski, “A68: a major subunit of paired helical filaments and derivatized forms of normal tau,” Science, vol. 251, no. 4994, pp. 675–678, 1991.
[25]  V. M.-Y. Lee, M. Goedert, and J. Q. Trojanowski, “Neurodegenerative tauopathies,” Annual Review of Neuroscience, vol. 24, pp. 1121–1159, 2001.
[26]  P. V. Arriagada, J. H. Growdon, E. T. Hedley-Whyte, and B. T. Hyman, “Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer's disease,” Neurology, vol. 42, no. 3, pp. 631–639, 1992.
[27]  M. Rapoport, H. N. Dawson, L. I. Binder, M. P. Vitek, and A. Ferreira, “Tau is essential to β-amyloid-induced neurotoxicity,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 9, pp. 6364–6369, 2002.
[28]  E. D. Roberson, K. Scearce-Levie, J. J. Palop et al., “Reducing endogenous tau ameliorates amyloid β-induced deficits in an Alzheimer's disease mouse model,” Science, vol. 316, no. 5825, pp. 750–754, 2007.
[29]  M. G. Spillantini and M. Goedert, “Tau protein pathology in neurodegenerative diseases,” Trends in Neurosciences, vol. 21, no. 10, pp. 428–433, 1998.
[30]  J. G. Wood, S. S. Mirra, N. J. Pollock, and L. I. Binder, “Neurofibrillary tangles of Alzheimer disease share antigenic determinants with the axonal microtubule-associated protein tau (τ),” Proceedings of the National Academy of Sciences of the United States of America, vol. 83, no. 11, pp. 4040–4043, 1986.
[31]  H. Braak and E. Braak, “Neuropathological stageing of Alzheimer-related changes,” Acta Neuropathologica, vol. 82, no. 4, pp. 239–259, 1991.
[32]  B. T. Hyman, J. C. Augustinack, and M. Ingelsson, “Transcriptional and conformational changes of the tau molecule in Alzheimer's disease,” Biochimica et Biophysica Acta, vol. 1739, no. 2, pp. 150–157, 2005.
[33]  D. H. Geschwind, “Tau phosphorylation, tangles, and neurodegeneration: the chicken or the egg?” Neuron, vol. 40, no. 3, pp. 457–460, 2003.
[34]  C. Ballatore, V. M. Y. Lee, and J. Q. Trojanowski, “Tau-mediated neurodegeneration in Alzheimer's disease and related disorders,” Nature Reviews Neuroscience, vol. 8, no. 9, pp. 663–672, 2007.
[35]  M. Morris, S. Maeda, K. Vossel, and L. Mucke, “The many faces of tau,” Neuron, vol. 70, no. 3, pp. 410–426, 2011.
[36]  P. Novak, M. Prcina, and E. Kontsekova, “Tauons and prions: infamous cousins?” Alzheimer's Disease, vol. 26, pp. 413–430, 2011.
[37]  A. Calignon, M. Polydoro, M. Suarez-Calvet, et al., “Propagation of tau pathology in a model of early Alzheimer’s disease,” Neuron, vol. 73, pp. 685–697, 2012.
[38]  L. Liu, V. Drouet, J. W. Wu, et al., “Trans-synaptic spread of tau pathology in vivo,” PlosOne, vol. 7, Article ID e31303, 2012.
[39]  R. D. Terry, E. Masliah, D. P. Salmon et al., “Physical basis of cognitive alterations in Alzheimer's disease: synapse loss is the major correlate of cognitive impairment,” Annals of Neurology, vol. 30, no. 4, pp. 572–580, 1991.
[40]  S. T. DeKosky and S. W. Scheff, “Synapse loss in frontal cortex biopsies in Alzheimer's disease: correlation with cognitive severity,” Annals of Neurology, vol. 27, no. 5, pp. 457–464, 1990.
[41]  P. Tiraboschi, L. A. Hansen, M. Alford, E. Masliah, L. J. Thal, and J. Corey-Bloom, “The decline in synapses and cholinergic activity is asynchronous in Alzheimer's disease,” Neurology, vol. 55, no. 9, pp. 1278–1283, 2000.
[42]  D. J. Selkoe, “Alzheimer's disease is a synaptic failure,” Science, vol. 298, no. 5594, pp. 789–791, 2002.
[43]  P. J. Yao, “Synaptic frailty and clathrin-mediated synaptic vesicle trafficking in Alzheimer's disease,” Trends in Neurosciences, vol. 27, no. 1, pp. 24–29, 2004.
[44]  J. E. Hamos, L. J. DeGennaro, and D. A. Drachman, “Synaptic loss in Alzheimer's disease and other dementias,” Neurology, vol. 39, no. 3, pp. 355–361, 1989.
[45]  E. Marcello, R. Epis, C. Saraceno, and M. Di Luca, “Synaptic dysfunction in Alzheimer’s disease,” in Synaptic Plasticity, M. R. Kreutz and C. Sala, Eds., pp. 573–601, Springer, Weinheim, Germany, 2012.
[46]  C. Bertoni-Freddari, P. Fattoretti, M. Pieroni, W. Meier-Ruge, and J. Ulrich, “Enlargement of synaptic size as a compensative reaction in aging and dementia,” Pathology Research and Practice, vol. 188, no. 4-5, pp. 612–615, 1992.
[47]  M. C. Gastard, J. C. Troncoso, and V. E. Koliatsos, “Caspase activation in the limbic cortex of subjects with early Alzheimer's disease,” Annals of Neurology, vol. 54, no. 3, pp. 393–398, 2003.
[48]  W. G. Honer, “Pathology of presynaptic proteins in Alzheimer's disease: more than simple loss of terminals,” Neurobiology of Aging, vol. 24, no. 8, pp. 1047–1062, 2003.
[49]  K. Hsiao, P. Chapman, S. Nilsen et al., “Correlative memory deficits, Aβ elevation, and amyloid plaques in transgenic mice,” Science, vol. 274, no. 5284, pp. 99–102, 1996.
[50]  D. L. King and G. W. Arendash, “Maintained synaptophysin immunoreactivity in Tg2576 transgenic mice during aging: correlations with cognitive impairment,” Brain Research, vol. 926, no. 1-2, pp. 58–68, 2002.
[51]  D. E. Goll, V. F. Thompson, H. Li, W. Wei, and J. Cong, “The calpain system,” Physiological Reviews, vol. 83, no. 3, pp. 731–801, 2003.
[52]  Y. Ono and H. Sorimachi, “Calpains- an elaborate proteolytic system,” Biochimica et Biophysica Acta, vol. 1824, pp. 224–236, 2012.
[53]  P. S. Vosler, C. S. Brennan, and J. Chen, “Calpain-mediated signaling mechanisms in neuronal injury and neurodegeneration,” Molecular Neurobiology, vol. 38, no. 1, pp. 78–100, 2008.
[54]  R. A. Nixon, “The calpains in aging and aging-related diseases,” Ageing Research Reviews, vol. 2, no. 4, pp. 407–418, 2003.
[55]  K. Suzuki and H. Sorimachi, “A novel aspect of calpain activation,” FEBS Letters, vol. 433, no. 1-2, pp. 1–4, 1998.
[56]  T. Moldoveanu, C. M. Hosfield, D. Lim, J. S. Elce, Z. Jia, and P. L. Davies, “A Ca2+ switch aligns the active site of calpain,” Cell, vol. 108, no. 5, pp. 649–660, 2002.
[57]  C. M. Hosfield, J. S. Elce, P. L. Davies, and Z. Jia, “Crystal structure of calpain reveals the structural basis for Ca2+-dependent protease activity and a novel mode of enzyme activation,” EMBO Journal, vol. 18, no. 24, pp. 6880–6889, 1999.
[58]  S. Strobl, C. Fernandez-Catalan, M. Braun et al., “The crystal structure of calcium-free human m-calpain suggests an electrostatic switch mechanism for activation by calcium,” Proceedings of the National Academy of Sciences of the United States of America, vol. 97, no. 2, pp. 588–592, 2000.
[59]  P. Tompa, Y. Emori, H. Sorimachi, K. Suzuki, and P. Friedrich, “Domain III of calpain is a Ca2+-regulated phospholipid-binding domain,” Biochemical and Biophysical Research Communications, vol. 280, no. 5, pp. 1333–1339, 2001.
[60]  H. Sorimachi and K. Suzuki, “The structure of calpain,” Journal of Biochemistry, vol. 129, no. 5, pp. 653–664, 2001.
[61]  E. Carafoli and M. Molinari, “Calpain: a protease in search of a function?” Biochemical and Biophysical Research Communications, vol. 247, no. 2, pp. 193–203, 1998.
[62]  T. Hamakubo, R. Kannagi, T. Murachi, and A. Matus, “Distribution of calpains I and II in rat brain,” Journal of Neuroscience, vol. 6, no. 11, pp. 3103–3111, 1986.
[63]  L. S. Perlmutter, R. Siman, C. Gall, P. Seubert, M. Baudry, and G. Lynch, “The ultrastructural localization of calcium-activated protease 'calpain' in rat brain,” Synapse, vol. 2, no. 1, pp. 79–88, 1988.
[64]  T. Murachi, M. Hatanaka, and T. Hamakubo, “Calpains and neuropeptide metabolism,” in Neuropeptides and Their Peptidases, T. E. Chichester, Ed., pp. 202–228,, Ellis Horwood, Chichester, UK, 1987.
[65]  D. E. Croall, C. A. Slaughter, H. S. Wortham, C. M. Skelly, L. DeOgny, and C. R. Moomaw, “Polyclonal antisera specific for the proenzyme form of each calpain,” Biochimica et Biophysica Acta, vol. 1121, no. 1-2, pp. 47–53, 1992.
[66]  S. Fischer, J. Vandekerckhove, C. Ampe, P. Traub, and K. Weber, “Protein-chemical identification of the major cleavage sites of the Ca2+ proteinase on murine vimentin, the mesenchymal intermediate filament protein,” Biological Chemistry Hoppe-Seyler, vol. 367, no. 11, pp. 1147–1152, 1986.
[67]  A. S. Harris, D. E. Croall, and J. S. Morrow, “The calmodulin-binding site in α-fodrin is near the calcium-dependent protease-I cleavage site,” The Journal of Biological Chemistry, vol. 263, no. 30, pp. 15754–15761, 1988.
[68]  P. R. Stabach, C. D. Cianci, S. B. Glantz, Z. Zhang, and J. S. Morrow, “Site-directed mutagenesis of αII spectrin at codon 1175 modulates its μ-calpain susceptibility,” Biochemistry, vol. 36, no. 1, pp. 57–65, 1997.
[69]  S. A. Coolican and D. R. Hathaway, “Effect of L-α-phosphatidylinositol on a vascular smooth muscle Ca2+-dependent protease. Reduction of the Ca2+ requirement for autolysis,” The Journal of Biological Chemistry, vol. 259, no. 19, pp. 11627–11630, 1984.
[70]  T. C. Saido, K. Mizuno, and K. Suzuki, “Proteolysis of protein kinase C by calpain: effect of acidic phospholipids,” Biomedica Biochimica Acta, vol. 50, no. 4–6, pp. 485–489, 1991.
[71]  T. C. Saido, M. Shibata, T. Takenawa, H. Murofushi, and K. Suzuki, “Positive regulation of μ-calpain action by polyphosphoinositides,” The Journal of Biological Chemistry, vol. 267, no. 34, pp. 24585–24590, 1992.
[72]  R. Kiss, D. Kovács, P. Tompa, and A. Perczel, “Local structural preferences of calpastatin, the intrinsically unstructured protein inhibitor of calpain,” Biochemistry, vol. 47, no. 26, pp. 6936–6945, 2008.
[73]  V. F. Thompson and D. E. Goll, “Purification of m-calpain, m-calpain, and calpastatin from animal tissues,” in Methods in Molecular Biology. Calpain Methods and Protocols, J. S. Elce and N. J. Totowa, Eds., vol. 144, pp. 3–6, Humana Press, 2000.
[74]  M. V. Rao, P. S. Mohan, C. M. Peterhoff et al., “Marked calpastatin (CAST) depletion in Alzheimer's disease accelerates cytoskeleton disruption and neurodegeneration: neuroprotection by CAST overexpression,” Journal of Neuroscience, vol. 28, no. 47, pp. 12241–12254, 2008.
[75]  Y. Emori, H. Kawasaki, S. Imajoh, K. Imahori, and K. Suzuki, “Endogenous inhibitor for calcium-dependent cysteine protease contains four internal repeats that could be responsible for its multiple reactive sites,” Proceedings of the National Academy of Sciences of the United States of America, vol. 84, no. 11, pp. 3590–3594, 1987.
[76]  M. Maki, E. Takano, H. Mori, R. Kannagi, T. Murachi, and M. Hatanaka, “Repetitive region of calpastatin is a functional unit of the proteinase inhibitor,” Biochemical and Biophysical Research Communications, vol. 143, no. 1, pp. 300–308, 1987.
[77]  M. Maki, E. Takano, H. Mori, A. Sato, T. Murachi, and M. Hatanaka, “All four internally repetitive domains of pig calpastatin possess inhibitory activities against calpains I and II,” FEBS Letters, vol. 223, no. 1, pp. 174–180, 1987.
[78]  S. Barnoy, M. Maki, and N. S. Kosower, “Overexpression of calpastatin inhibits L8 myoblast fusion,” Biochemical and Biophysical Research Communications, vol. 332, no. 3, pp. 697–701, 2005.
[79]  M. Higuchi, M. Tomioka, J. Takano et al., “Distinct mechanistic roles of calpain and caspase activation in neurodegeneration as revealed in mice overexpressing their specific inhibitors,” The Journal of Biological Chemistry, vol. 280, no. 15, pp. 15229–15237, 2005.
[80]  A. Maekawa, J. K. Lee, T. Nagaya et al., “Overexpression of calpastatin by gene transfer prevents troponin I degradation and ameliorates contractile dysfunction in rat hearts subjected to ischemia/reperfusion,” Journal of Molecular and Cellular Cardiology, vol. 35, no. 10, pp. 1277–1284, 2003.
[81]  A. Glading, P. Chang, D. A. Lauffenburger, and A. Wells, “Epidermal growth factor receptor activation of calpain is required for fibroblast motility and occurs via an ERK/MAP kinase signaling pathway,” The Journal of Biological Chemistry, vol. 275, no. 4, pp. 2390–2398, 2000.
[82]  H. Shiraha, A. Glading, J. Chou, Z. Jia, and A. Wells, “Activation of m-calpain (calpain II) by epidermal growth factor is limited by protein kinase A phosphorylation of m-calpain,” Molecular and Cellular Biology, vol. 22, no. 8, pp. 2716–2727, 2002.
[83]  S. D. Smith, Z. Jia, K. K. Huynh, A. Wells, and J. S. Elce, “Glutamate substitutions at a PKA consensus site are consistent with inactivation of calpain by phosphorylation,” FEBS Letters, vol. 542, no. 1–3, pp. 115–118, 2003.
[84]  M. Averna, R. De Tullio, F. Salamino, R. Minafra, S. Pontremoli, and E. Melloni, “Age-dependent degradation of calpastatin in kidney of hypertensive rats,” The Journal of Biological Chemistry, vol. 276, no. 42, pp. 38426–38432, 2001.
[85]  M. Benuck, M. Banay-Schwartz, T. DeGuzman, and A. Lajtha, “Changes in brain protease activity in aging,” Journal of Neurochemistry, vol. 67, no. 5, pp. 2019–2029, 1996.
[86]  H. Manya, M. Inomata, T. Fujimori et al., “Klotho protein deficiency leads to overactivation of μ-calpain,” The Journal of Biological Chemistry, vol. 277, no. 38, pp. 35503–35508, 2002.
[87]  J. A. Sloane, J. D. Hinman, M. Lubonia, W. Hollander, and C. R. Abraham, “Age-dependent myelin degeneration and proteolysis of oligodendrocyte proteins is associated with the activation of calpain-1 in the rhesus monkey,” Journal of Neurochemistry, vol. 84, no. 1, pp. 157–168, 2003.
[88]  K. I. Saito, J. S. Elce, J. E. Hamos, and R. A. Nixon, “Widespread activation of calcium-activated neutral proteinase (calpain) in the brain in Alzheimer disease: a potential molecular basis for neuronal degeneration,” Proceedings of the National Academy of Sciences of the United States of America, vol. 90, no. 7, pp. 2628–2632, 1993.
[89]  T. Saitoh, E. Masliah, L. W. Jin, G. M. Cole, T. Wieloch, and I. P. Shapiro, “Protein kinases and phosphorylation in neurologic disorders and cell death,” Laboratory Investigation, vol. 64, no. 5, pp. 596–616, 1991.
[90]  A. Ferreira and E. H. Bigio, “Calpain-mediated tau cleavage: a mechanism leading to neurodegeneration shared by multiple tauopathies,” Molecular Medicine, vol. 17, no. 7-8, pp. 676–685, 2011.
[91]  C. Peterson and J. E. Goldman, “Alterations in calcium content and biochemical processes in cultured skin fibroblasts from aged and Alzheimer donors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 83, no. 8, pp. 2758–2762, 1986.
[92]  J. O. Karlsson, K. Blennow, B. Holmberg et al., “Increased proteolytic activity in erythrocytes from patients with Alzheimer's disease,” Dementia, vol. 3, no. 4, pp. 200–204, 1992.
[93]  G. N. Patrick, L. Zukerberg, M. Nikolic, S. de la Monte, P. Dikkes, and L.-H. Tsai, “Conversion of p35 to p25 deregulates Cdk5 activity and promotes neurodegeneration,” Nature, vol. 402, no. 6762, pp. 615–622, 1999.
[94]  T. Tsuji, S. Shimohama, J. Kimura, and K. Shimizu, “M-calpain (calcium-activated neutral proteinase) in alzheimer's disease brains,” Neuroscience Letters, vol. 248, no. 2, pp. 109–112, 1998.
[95]  B. L. Kelly, R. Vassar, and A. Ferreira, “β-amyloid-induced dynamin 1 depletion in hippocampal neurons: a potential mechanism for early cognitive decline in Alzheimer disease,” The Journal of Biological Chemistry, vol. 280, no. 36, pp. 31746–31753, 2005.
[96]  B. L. Kelly and A. Ferreira, “β-amyloid-induced dynamin 1 degradation is mediated by N-methyl-D-aspartate receptors in hippocampal neurons,” The Journal of Biological Chemistry, vol. 281, no. 38, pp. 28079–28089, 2006.
[97]  B. L. Kelly and A. Ferreira, “Beta-amyloid disrupted synaptic vesicle endocytosis in cultured hippocampal neurons,” Neuroscience, vol. 147, no. 1, pp. 60–70, 2007.
[98]  K. M. Abdel-Hamid and K. G. Baimbridge, “The effects of artificial calcium buffers on calcium responses and glutamate-mediated excitotoxicity in cultured hippocampal neurons,” Neuroscience, vol. 81, no. 3, pp. 673–687, 1997.
[99]  J. C. Dodart, K. R. Bales, K. S. Gannon et al., “Immunization reverses memory deficits without reducing brain Aβ burden in Alzheimer's disease model,” Nature Neuroscience, vol. 5, no. 5, pp. 452–457, 2002.
[100]  C. Rovira, N. Arbez, and J. Mariani, “Aβ(25–35) and Aβ(1–40) act on different calcium channels in CA1 hippocampal neurons,” Biochemical and Biophysical Research Communications, vol. 296, no. 5, pp. 1317–1321, 2002.
[101]  Z. Qiu and D. L. Gruol, “Interleukin-6, β-amyloid peptide and NMDA interactions in rat cortical neurons,” Journal of Neuroimmunology, vol. 139, no. 1-2, pp. 51–57, 2003.
[102]  J. E. Huettner and B. P. Bean, “Block of N-methyl-D-aspartate-activated current by the anticonvulsant MK-801: selective binding to open channels,” Proceedings of the National Academy of Sciences of the United States of America, vol. 85, no. 4, pp. 1307–1311, 1988.
[103]  R. Bullock, “Efficacy and safety of memantine in moderate-to-severe Alzheimer disease: the evidence to date,” Alzheimer Disease and Associated Disorders, vol. 20, no. 1, pp. 23–29, 2006.
[104]  R. J. Fanelli, R. T. McCarthy, and J. Chisholm, “Neuropharmacology of nimodipine: from single channels to behavior,” Annals of the New York Academy of Sciences, vol. 747, pp. 336–350, 1994.
[105]  A. M. Nicholson and A. Ferreira, “Cholesterol and neuronal susceptibility to beta-amyloid toxicity,” Trends in Cognitive Sciences, vol. 5, pp. 35–56, 2011.
[106]  M. Kivipelto, E. L. Helkala, T. H?nninen et al., “Midlife vascular risk factors and late-life mild cognitive impairment: a population-based study,” Neurology, vol. 56, no. 12, pp. 1683–1689, 2001.
[107]  A. Solomon, M. Kivipelto, B. Wolozin, J. Zhou, and R. A. Whitmer, “Midlife serum cholesterol and increased risk of Alzheimer's and vascular dementia three decades later,” Dementia and Geriatric Cognitive Disorders, vol. 28, no. 1, pp. 75–80, 2009.
[108]  E. M. L. Bastiaanse, L. D. E. Atsma, M. M. C. Kuijpers, and A. Van der Laarse, “The effect of sarcolemmal cholesterol content on intracellular calcium ion concentration in cultured cardiomyocytes,” Archives of Biochemistry and Biophysics, vol. 313, no. 1, pp. 58–63, 1994.
[109]  H. Hartmann, A. Eckert, and W. E. Muller, “Apolipoprotein E and cholesterol affect neuronal calcium signalling: the possible relationship to β-amyloid neurotoxicity,” Biochemical and Biophysical Research Communications, vol. 200, no. 3, pp. 1185–1192, 1994.
[110]  M. Kawahara and Y. Kuroda, “Intracellular calcium changes in neuronal cells induced by Alzheimer's β-amyloid protein are blocked by estradiol and cholesterol,” Cellular and Molecular Neurobiology, vol. 21, no. 1, pp. 1–13, 2001.
[111]  A. M. Nicholson and A. Ferreira, “Increased membrane cholesterol might render mature hippocampal neurons more susceptible to β-Amyloid-induced calpain activation and tau toxicity,” Journal of Neuroscience, vol. 29, no. 14, pp. 4640–4651, 2009.
[112]  A. M. Nicholson, D. N. R. Methner, and A. Ferreira, “Membrane cholesterol modulates β-amyloid-dependent Tau cleavage by inducing changes in the membrane content and localization of N-methyl-D-aspartic acid receptors,” The Journal of Biological Chemistry, vol. 286, no. 2, pp. 976–986, 2011.
[113]  T. Vaisid, N. S. Kosower, A. Katzav, J. Chapman, and S. Barnoy, “Calpastatin levels affect calpain activation and calpain proteolytic activity in APP transgenic mouse model of Alzheimer's disease,” Neurochemistry International, vol. 51, no. 6-7, pp. 391–397, 2007.
[114]  K. Blomgren, U. Hallin, A. L. Andersson et al., “Calpastatin is up-regulated in response to hypoxia and is a suicide substrate to calpain after neonatal cerebral hypoxia-ischemia,” The Journal of Biological Chemistry, vol. 274, no. 20, pp. 14046–14052, 1999.
[115]  R. L. Mellgren, M. T. Mericle, and R. D. Lane, “Proteolysis of the calcium-dependent protease inhibitor by myocardial calcium-dependent protease,” Archives of Biochemistry and Biophysics, vol. 246, no. 1, pp. 233–239, 1986.
[116]  E. Adamec, P. Mohan, J. P. Vonsattel, and R. A. Nixon, “Calpain activation in neurodegenerative diseases: confocal immunofluorescence study with antibodies specifically recognizing the active form of calpain 2,” Acta Neuropathologica, vol. 104, no. 1, pp. 92–104, 2002.
[117]  F. Grynspan, W. R. Griffin, A. Cataldo, S. Katayama, and R. A. Nixon, “Active site-directed antibodies identify calpain II as an early- appearing and pervasive component of neurofibrillary pathology in Alzheimer's disease,” Brain Research, vol. 763, no. 2, pp. 145–158, 1997.
[118]  S. Shimohama, T. Suenaga, W. Araki, Y. Yamaoaka, K. Shimizu, and J. Kimura, “Presence of calpain II immunoreactivity in senile plaques in Alzheimer's disease,” Brain Research, vol. 558, no. 1, pp. 105–108, 1991.
[119]  S. S. Sisodia, S. H. Kim, and G. Thinakaran, “Function and dysfunction of the presenilins,” American Journal of Human Genetics, vol. 65, no. 1, pp. 7–12, 1999.
[120]  R. Vassar, B. D. Bennett, S. Babu-Khan et al., “β-secretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE,” Science, vol. 286, no. 5440, pp. 735–741, 1999.
[121]  D. Edbauer, E. Winkler, J. T. Regula, B. Pesold, H. Steiner, and C. Haass, “Reconstitution of γ-secretase activity,” Nature Cell Biology, vol. 5, no. 5, pp. 486–488, 2003.
[122]  J. T. Jarrett, E. P. Berger, and P. T. Lansbury, “The carboxy terminus of the β amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer's disease,” Biochemistry, vol. 32, no. 18, pp. 4693–4697, 1993.
[123]  M. Citron, T. Oltersdorf, C. Haass et al., “Mutation of the β-amyloid precursor protein in familial Alzheimer's disease increases β-protein production,” Nature, vol. 360, no. 6405, pp. 672–674, 1992.
[124]  M. Citron, D. Westaway, W. Xia et al., “Mutant presenilins of Alzheimer's disease increase production of 42-residue amyloid β-protein in both transfected cells and transgenic mice,” Nature Medicine, vol. 3, no. 1, pp. 67–72, 1997.
[125]  X. D. Cai, T. E. Golde, and S. G. Younkin, “Release of excess amyloid β protein from a mutant amyloid β protein precursor,” Science, vol. 259, no. 5094, pp. 514–516, 1993.
[126]  N. Suzuki, T. T. Cheung, X. D. Cai et al., “An increased percentage of long amyloid β protein secreted by familial amyloid β protein precursor (βAPP717) mutants,” Science, vol. 264, no. 5163, pp. 1336–1340, 1994.
[127]  H. Fukumoto, B. S. Cheung, B. T. Hyman, and M. C. Irizarry, “β-secretase protein and activity are increased in the neocortex in Alzheimer disease,” Archives of Neurology, vol. 59, no. 9, pp. 1381–1389, 2002.
[128]  L. B. Yang, K. Lindholm, R. Yan et al., “Elevated β-secretase expression and enzymatic activity detected in sporadic Alzheimer disease,” Nature Medicine, vol. 9, no. 1, pp. 3–4, 2003.
[129]  Q. Li and T. C. Südhof, “Cleavage of amyloid-beta precursor protein and amyloid-beta precursor-like protein by BACE 1,” The Journal of Biological Chemistry, vol. 279, no. 11, pp. 10542–10550, 2004.
[130]  B. Liang, B.-Y. Duan, X.-P. Zhou, J.-X. Gong, and Z.-G. Luo, “Calpain activation promotes BACE1 expression, amyloid precursor protein processing, and amyloid plaque formation in a transgenic mouse model of alzheimer disease,” The Journal of Biological Chemistry, vol. 285, no. 36, pp. 27737–27744, 2010.
[131]  M. Higuchi, N. Iwata, Y. Matsuba, et al., “Mechanistic involvement of the calpain-calpastatin system in Alzheimer neuropathology,” The FASEB Journal, vol. 26, pp. 1204–1217, 2012.
[132]  J. Biernat, N. Gustke, G. Drewes, E. M. Mandelkow, and E. Mandelkow, “Phosphorylation of Ser262 strongly reduces binding of tau to microtubules: distinction between PHF-like immunoreactivity and microtubule binding,” Neuron, vol. 11, no. 1, pp. 153–163, 1993.
[133]  J. Biernat, E.-M. Mandelkow, C. Schroter et al., “The switch of tau protein to an Alzheimer-like state includes the phosphorylation of two serine-proline motifs upstream of the microtubule binding region,” EMBO Journal, vol. 11, no. 4, pp. 1593–1597, 1992.
[134]  G. T. Bramblett, M. Goedert, R. Jakes, S. E. Merrick, J. Q. Trojanowski, and V. M.-Y. Lee, “Abnormal tau phosphorylation at Ser396 in Alzheimer's disease recapitulates development and contributes to reduced microtubule binding,” Neuron, vol. 10, no. 6, pp. 1089–1099, 1993.
[135]  J. P. Brion, C. Smith, A. M. Couck, J. M. Gallo, and B. H. Anderton, “Developmental changes in τ phosphorylation: fetal τ is transiently phosphorylated in a manner similar to paired helical filament-τ characteristic of Alzheimer's disease,” Journal of Neurochemistry, vol. 61, no. 6, pp. 2071–2080, 1993.
[136]  M. Rapoport and A. Ferreira, “PD98059 prevents neurite degeneration induced by fibrillar β-amyloid in mature hippocampal neurons,” Journal of Neurochemistry, vol. 74, no. 1, pp. 125–133, 2000.
[137]  J. Busciglio, A. Lorenzo, J. Yeh, and B. A. Yankner, “β-amyloid fibrils induce tau phosphorylation and loss of microtubule binding,” Neuron, vol. 14, no. 4, pp. 879–888, 1995.
[138]  A. Ferreira, Q. Lu, L. Orecchio, and K. S. Kosik, “Selective phosphorylation of adult tau isoforms in mature hippocampal neurons exposed to fibrillar Aβ,” Molecular and Cellular Neurosciences, vol. 9, no. 3, pp. 220–234, 1997.
[139]  T. B. Shea, A. N. Deargay, and F. J. Ekinci, “Beta-amyloid induced hyperphosphorylation of tau in human neuroblastoma cells involves MAP kinase,” Neuroscience Research Communications, vol. 22, pp. 45–49, 1998.
[140]  M. Pérez, F. Hernández, A. Gómez-Ramos, M. Smith, G. Perry, and J. Avila, “Formation of aberrant phosphotau fibrillar polymers in neural cultured cells,” European Journal of Biochemistry, vol. 269, no. 5, pp. 1484–1489, 2002.
[141]  M. Jin, N. Shepardson, T. Yang, G. Chen, D. Walsh, and D. J. Selkoe, “Soluble amyloid β-protein dimers isolated from Alzheimer cortex directly induce tau hyperphosphorylation and neuritic degeneration,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 14, pp. 5819–5824, 2011.
[142]  L. Martin, X. Latypova, and F. Terro, “Post-translational modifications of tau protein: implications for Alzheimer's disease,” Neurochemistry International, vol. 58, no. 4, pp. 458–471, 2011.
[143]  A. Takashima, K. Noguchi, K. Sato, T. Hoshino, and K. Imahori, “Tau protein kinase I is essential for amyloid β-protein-induced neurotoxicity,” Proceedings of the National Academy of Sciences of the United States of America, vol. 90, no. 16, pp. 7789–7793, 1993.
[144]  F. J. Ekinci, K. U. Malik, and T. B. Shea, “Activation of the L voltage-sensitive calcium channel by mitogen- activated protein (MAP) kinase following exposure of neuronal cells to β- amyloid. MAP kinase mediates β-amyloid-induced neurodegeneration,” The Journal of Biological Chemistry, vol. 274, no. 42, pp. 30322–30327, 1999.
[145]  A. Alvarez, R. Toro, A. Cáceres, and R. B. Maccioni, “Inhibition of tau phosphorylating protein kinase cdk5 prevents β-amyloid-induced neuronal death,” FEBS Letters, vol. 459, no. 3, pp. 421–426, 1999.
[146]  P. Go?i-Oliver, J. J. Lucas, J. Avila, and F. Hernández, “N-terminal cleavage of GSK-3 by calpain: a new form of GSK-3 regulation,” The Journal of Biological Chemistry, vol. 282, no. 31, pp. 22406–22413, 2007.
[147]  G. I. Kusakawa, T. Saito, R. Onuki, K. Ishiguro, T. Kishimoto, and S. I. Hisanaga, “Calpain-dependent proteolytic cleavage of the p35 cyclin-dependent kinase 5 activator to p25,” The Journal of Biological Chemistry, vol. 275, no. 22, pp. 17166–17172, 2000.
[148]  M.-S. Lee, Y. T. Kwon, M. Li, J. Peng, R. M. Friedlander, and L.-H. Tsai, “Neurotoxicity induces cleavage of p35 to p25 by calpain,” Nature, vol. 405, no. 6784, pp. 360–364, 2000.
[149]  R. Nath, M. Davis, A. W. Probert et al., “Processing of cdk5 activator p35 to its truncated form (p25) by Calpain in acutely injured neuronal cells,” Biochemical and Biophysical Research Communications, vol. 274, pp. 16–21, 2000.
[150]  Veeranna, T. Kaji, B. Boland et al., “Calpain mediates calcium-induced activation of the Erk 1,2 MARK pathway and cytoskeletal phosphorylation in neurons: relevance to alzheimer's disease,” American Journal of Pathology, vol. 165, no. 3, pp. 795–805, 2004.
[151]  A. L. Guillozet-Bongaarts, F. Garcia-Sierra, M. R. Reynolds et al., “Tau truncation during neurofibrillary tangle evolution in Alzheimer's disease,” Neurobiology of Aging, vol. 26, no. 7, pp. 1015–1022, 2005.
[152]  G. V. W. Johnson, R. S. Jope, and L. I. Binder, “Proteolysis of tau by calpain,” Biochemical and Biophysical Research Communications, vol. 163, no. 3, pp. 1505–1511, 1989.
[153]  M. Mercken, F. Grynspan, and R. A. Nixon, “Differential sensitivity to proteolysis by brain calpain of adult human tau, fetal human tau and PHF-tau,” FEBS Letters, vol. 368, no. 1, pp. 10–14, 1995.
[154]  L.-S. Yang and H. Ksiezak-Reding, “Calpain-induced proteolysis of normal human tau and tau associated with paired helical filaments,” European Journal of Biochemistry, vol. 233, no. 1, pp. 9–17, 1995.
[155]  L. S. Yang, W. Gordon-Krajcer, and H. Ksiezak-Reding, “Tau released from paired helical filaments with formic acid or guanidine is susceptible to calpain-mediated proteolysis,” Journal of Neurochemistry, vol. 69, no. 4, pp. 1548–1558, 1997.
[156]  M. C. Liu, F. Kobeissy, W. Zheng, Z. Zhang, R. L. Hayes, and K. K. Wang, “Dual vulnerability of tau to calpains and caspase-3 proteolysis under neurotoxic and neurodegenerative conditions,” ASN Neuro, vol. 3, no. 1, article e00051, 2011.
[157]  J. Reifert, D. Hartung-Cranston, and S. C. Feinstein, “Amyloid β-mediated cell death of cultured hippocampal neurons reveals extensive tau fragmentation without increased full-length tau phosphorylation,” The Journal of Biological Chemistry, vol. 286, no. 23, pp. 20797–20811, 2011.
[158]  G. Amadoro, V. Corsetti, A. Stringaro et al., “A NH2 tau fragment targets neuronal mitochondria at AD synapses: possible implications for neurodegeneration,” Journal of Alzheimer's Disease, vol. 21, no. 2, pp. 445–470, 2010.
[159]  A. Atlante, G. Amadoro, A. Bobba et al., “A peptide containing residues 26-44 of tau protein impairs mitochondrial oxidative phosphorylation acting at the level of the adenine nucleotide translocator,” Biochimica et Biophysica Acta, vol. 1777, no. 10, pp. 1289–1300, 2008.
[160]  S.-Y. Park and A. Ferreira, “The generation of a 17 kDa neurotoxic fragment: an alternative mechanism by which tau mediates β-amyloid-induced neurodegeneration,” Journal of Neuroscience, vol. 25, no. 22, pp. 5365–5375, 2005.
[161]  S.-Y. Park, C. E. Tournell, R. C. Sinjoanu, and A. Ferreira, “Caspase-3- and calpain-mediated tau cleavage are differentially prevented by estrogen and testosterone in beta-amyloid-treated hippocampal neurons,” Neuroscience, vol. 144, no. 1, pp. 119–127, 2007.
[162]  G. Amadoro, M. T. Ciotti, M. Costanzi, V. Cestari, P. Calissano, and N. Canu, “NMDA receptor mediates tau-induced neurotoxicity by calpain and ERK/MAPK activation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 8, pp. 2892–2897, 2006.
[163]  J. B. Reinecke, S. L. DeVos, J. P. McGrath, et al., “Implicating calpain in tau-mediated toxicity in vivo,” PLoS One, vol. 6, Article ID e23865, 2011.
[164]  S. Garg, T. Timm, E. M. Mandelkow, E. Mandelkow, and Y. Wang, “Cleavage of tau by calpain in Alzheimer's disease: the quest for the toxic 17 kD fragment,” Neurobiology of Aging, vol. 32, no. 1, pp. 1–14, 2011.
[165]  P. J. Yao, M. Zhu, E. I. Pyun et al., “Defects in expression of genes related to synaptic vesicle trafficking in frontal cortex of Alzheimer's disease,” Neurobiology of Disease, vol. 12, no. 2, pp. 97–109, 2003.
[166]  J. Liu, M. C. Liu, and K. K. Wang, “Calpain in the CNS: from synaptic function to neurotoxicity,” Science Signaling, vol. 1, no. 14, article re1, 2008.
[167]  S. G. Clark, D. L. Shurland, E. M. Meyerowitz, C. I. Bargmann, and A. M. van der Bliek, “A dynamin GTPase mutation causes a rapid and reversible temperature-inducible locomotion defect in C. elegans,” Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 19, pp. 10438–10443, 1997.
[168]  H. Damke, T. Baba, D. E. Warnock, and S. L. Schmid, “Induction of mutant dynamin specifically blocks endocytic coated vesicle formation,” Journal of Cell Biology, vol. 127, no. 4, pp. 915–934, 1994.
[169]  J. H. Koenig and K. Ikeda, “Disappearance and reformation of synaptic vesicle membrane upon transmitter release observed under reversible blockage of membrane retrieval,” Journal of Neuroscience, vol. 9, no. 11, pp. 3844–3860, 1989.
[170]  A. M. van der Bliek and E. M. Meyerowitz, “Dynamin-like protein encoded by the Drosophila shibire gene associated with vesicular traffic,” Nature, vol. 351, no. 6325, pp. 411–414, 1991.
[171]  R. C. Sinjoanu, S. Kleinschmidt, R. S. Bitner, J. D. Brioni, A. Moeller, and A. Ferreira, “The novel calpain inhibitor A-705253 potently inhibits oligomeric beta-amyloid-induced dynamin 1 and tau cleavage in hippocampal neurons,” Neurochemistry International, vol. 53, no. 3-4, pp. 79–88, 2008.
[172]  R. Bi, X. Bi, and M. Baudry, “Phosphorylation regulates calpain-mediated truncation of glutamate ionotropic receptors,” Brain Research, vol. 797, no. 1, pp. 154–158, 1998.
[173]  X. Bi, Y. Rong, J. Chen, S. Dang, Z. Wang, and M. Baudry, “Calpain-mediated regulation of NMDA receptor structure and function,” Brain Research, vol. 790, no. 1-2, pp. 245–253, 1998.
[174]  X. Lu, Y. Rong, and M. Baudry, “Calpain-mediated degradation of PSD-95 in developing and adult rat brain,” Neuroscience Letters, vol. 286, no. 2, pp. 149–153, 2000.
[175]  L. Vinade, J. D. Petersen, K. Do, A. Dosemeci, and T. S. Reese, “Activation of calpain may alter the postsynaptic density structure and modulate anchoring of NMDA receptors,” Synapse, vol. 40, no. 4, pp. 302–309, 2001.
[176]  H. Y. Wu, F. C. Hsu, A. J. Gleichman, I. Baconguis, D. A. Coulter, and D. R. Lynch, “Fyn-mediated phosphorylation of NR2B Tyr-1336 controls calpain-mediated NR2B cleavage in neurons and heterologous systems,” The Journal of Biological Chemistry, vol. 282, no. 28, pp. 20075–20087, 2007.
[177]  W. Xu, T. P. Wong, N. Chery, T. Gaertner, Y. T. Wang, and M. Baudry, “Calpain-mediated mGluR1α truncation: a key step in excitotoxicity,” Neuron, vol. 53, no. 3, pp. 399–412, 2007.
[178]  L. Calò, V. Bruno, P. Spinsanti et al., “Interactions between ephrin-B and metabotropic glutamate 1 receptors in brain tissue and cultured neurons,” Journal of Neuroscience, vol. 25, no. 9, pp. 2245–2254, 2005.
[179]  A. Barco, J. M. Alarcon, and E. R. Kandel, “Expression of constitutively active CREB protein facilitates the late phase of long-term potentiation by enhancing synaptic capture,” Cell, vol. 108, no. 5, pp. 689–703, 2002.
[180]  Z. Liang, F. Liu, I. Grundke-Iqbal, K. Iqbal, and C. X. Gong, “Down-regulation of cAMP-dependent protein kinase by over-activated calpain in Alzheimer disease brain,” Journal of Neurochemistry, vol. 103, no. 6, pp. 2462–2470, 2007.
[181]  J. S. C. Arthur, J. S. Elce, C. Hegadorn, K. Williams, and P. A. Greer, “Disruption of the murine calpain small subunit gene, Capn4: calpain is essential for embryonic development but not for cell growth and division,” Molecular and Cellular Biology, vol. 20, no. 12, pp. 4474–4481, 2000.
[182]  K. Taniguchi, K. Umeshita, M. Sakon et al., “Suppression of oxidative stress-induced hepatocyte injury by calpain antisense,” Journal of Surgical Research, vol. 111, no. 1, pp. 23–27, 2003.
[183]  G. Di Rosa, T. Odrljin, R. A. Nixon, and O. Arancio, “Calpain inhibitors: a treatment for alzheimer's disease,” Journal of Molecular Neuroscience, vol. 19, no. 1-2, pp. 135–141, 2002.
[184]  J. R. Brorson, C. J. Marcuccilli, and R. J. Miller, “Delayed antagonism of calpain reduces excitotoxicity in cultured neurons,” Stroke, vol. 26, no. 7, pp. 1259–1267, 1995.
[185]  A. T. McCollum, F. Jafarifar, B. C. Lynn et al., “Inhibition of calpain-mediated cell death by a novel peptide inhibitor,” Experimental Neurology, vol. 202, no. 2, pp. 506–513, 2006.
[186]  F. Battaglia, F. Trinchese, S. Liu, S. Walter, R. A. Nixon, and O. Arancio, “Calpain inhibitors, a treatment for Alzheimer's disease: position paper,” Journal of Molecular Neuroscience, vol. 20, no. 3, pp. 357–362, 2003.
[187]  S. Kunz, E. Niederberger, C. Ehnert et al., “The calpain inhibitor MDL 28170 prevents inflammation-induced neurofilament light chain breakdown in the spinal cord and reduces thermal hyperalgesia,” Pain, vol. 110, no. 1-2, pp. 409–418, 2004.
[188]  P. A. Li, W. Howlett, Q. P. He, H. Miyashita, M. Siddiqui, and A. Shuaib, “Postischemic treatment with calpain inhibitor MDL 28170 ameliorates brain damage in a gerbil model of global ischemia,” Neuroscience Letters, vol. 247, no. 1, pp. 17–20, 1998.
[189]  W. Lubisch, E. Beckenbach, S. Bopp et al., “Benzoylalanine-derived ketoamides carrying vinylbenzyl amino residues: discovery of potent water-soluble calpain inhibitors with oral bioavailability,” Journal of Medicinal Chemistry, vol. 46, no. 12, pp. 2404–2412, 2003.
[190]  I. Granic, C. Nyakas, P. G. M. Luiten et al., “Calpain inhibition prevents amyloid-β-induced neurodegeneration and associated behavioral dysfunction in rats,” Neuropharmacology, vol. 59, no. 4-5, pp. 334–342, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413