메뉴 건너뛰기




Volumn 6, Issue SEP, 2015, Pages

Cathepsin B is a new drug target for traumatic brain injury therapeutics: Evidence for E64d as a promising lead drug candidate

Author keywords

Cathepsin B; Drug; E64d; Protease; Therapeutics; Traumatic brain injury

Indexed keywords

ALOXISTATIN; ALOXISTATIN ACID; BENZYLOXYCARBONYLASPARTYLGLUTAMYLVALYLASPARTYL FLUOROMETHYL KETONE; BENZYLOXYCARBONYLPHENYLALANYLALANYL FLUOROMETHYL KETONE; CARBOBENOXYPHENYLSERINE(2 BENZYL) 2 [METHYL (4 PHENYLBENZOYL)AMINO]BENZOIC ACID; CATHEPSIN B; CATHEPSIN B INHIBITOR; CYSTATIN; CYSTEINE PROTEINASE; MORPHOLINUREA LEUCINEHOMOPHENYLALANINEVINYLSULFONE PHENYL; N (3 PROPYLCARBAMOYLOXIRANE 2 CARBONYL)ISOLEUCYLPROLINE; N (3 PROPYLCARBAMOYLOXIRANE 2 CARBONYL)ISOLEUCYLPROLINE METHYL ESTER; N METHYLPIPERAZINE PHENYLALANINE HOMOPHENYLALANINE VINYLSULFONE PHENYL; N [N (3 CARBOXYOXIRANE 2 CARBONYL)LEUCYL]AGMATINE; SODIUM 3 [(1 ISOBUTOXY 4 METHYLPENTAN 2 YL)CARBAMOYL]OXIRANE 2 CARBOXYLATE; UNCLASSIFIED DRUG;

EID: 84944725337     PISSN: None     EISSN: 16642295     Source Type: Journal    
DOI: 10.3389/fneur.2015.00178     Document Type: Review
Times cited : (74)

References (309)
  • 3
    • 33751379575 scopus 로고    scopus 로고
    • Incidence of traumatic brain injury in the United States, 2003
    • Rutland-Brown W, Langlois JA, Thomas KE, Xi YL. Incidence of traumatic brain injury in the United States, 2003. J Head Trauma Rehabil (2006) 21:544-8. doi:10.1097/00001199-200611000-00009
    • (2006) J Head Trauma Rehabil , vol.21 , pp. 544-548
    • Rutland-Brown, W.1    Langlois, J.A.2    Thomas, K.E.3    Xi, Y.L.4
  • 5
    • 33748775183 scopus 로고    scopus 로고
    • Military TBI during the Iraq and Afghanistan wars
    • Warden D. Military TBI during the Iraq and Afghanistan wars. J Head Trauma Rehabil (2006) 21:398-402. doi:10.1097/00001199-200609000-00004
    • (2006) J Head Trauma Rehabil , vol.21 , pp. 398-402
    • Warden, D.1
  • 7
    • 42649137566 scopus 로고    scopus 로고
    • Traumatic brain injury: can the consequences be stopped?
    • Park E, Bell JD, Baker AJ. Traumatic brain injury: can the consequences be stopped? CMAJ (2008) 178:1163-70. doi:10.1503/cmaj.080282
    • (2008) CMAJ , vol.178 , pp. 1163-1170
    • Park, E.1    Bell, J.D.2    Baker, A.J.3
  • 8
    • 82255182190 scopus 로고    scopus 로고
    • Neurobiological consequences of traumatic brain injury
    • McAllister TW. Neurobiological consequences of traumatic brain injury. Dialogues Clin Neurosci (2011) 13:287-300.
    • (2011) Dialogues Clin Neurosci , vol.13 , pp. 287-300
    • McAllister, T.W.1
  • 9
    • 34447527791 scopus 로고    scopus 로고
    • Pathophysiology of traumatic brain injury
    • Werner C, Engelhard K. Pathophysiology of traumatic brain injury. Br J Anaesth (2007) 99:4-9. doi:10.1093/bja/aem131
    • (2007) Br J Anaesth , vol.99 , pp. 4-9
    • Werner, C.1    Engelhard, K.2
  • 10
    • 34347397404 scopus 로고    scopus 로고
    • Cellular and subcellular change evoked by diffuse traumatic brain injury: a complex web of change extending far beyond focal damage
    • Farkas O, Povlishock JT. Cellular and subcellular change evoked by diffuse traumatic brain injury: a complex web of change extending far beyond focal damage. Prog Brain Res (2007) 161:43-59. doi:10.1016/S0079-6123(06)61004-2
    • (2007) Prog Brain Res , vol.161 , pp. 43-59
    • Farkas, O.1    Povlishock, J.T.2
  • 11
    • 84926319950 scopus 로고    scopus 로고
    • Inflammation and neuroprotection in traumatic brain injury
    • Corps KN, Roth TL, Mcgavern DB. Inflammation and neuroprotection in traumatic brain injury. JAMA Neurol (2015) 72:355-62. doi:10.1001/jamaneurol.2014.3558
    • (2015) JAMA Neurol , vol.72 , pp. 355-362
    • Corps, K.N.1    Roth, T.L.2    Mcgavern, D.B.3
  • 12
    • 0042022027 scopus 로고    scopus 로고
    • Microvascular basal lamina antigen loss after traumatic brain injury in the rat
    • Muellner A, Benz M, Kloss CU, Mautes A, Burggraf D, Hamann GF. Microvascular basal lamina antigen loss after traumatic brain injury in the rat. J Neurotrauma (2003) 20:745-54. doi:10.1089/089771503767869971
    • (2003) J Neurotrauma , vol.20 , pp. 745-754
    • Muellner, A.1    Benz, M.2    Kloss, C.U.3    Mautes, A.4    Burggraf, D.5    Hamann, G.F.6
  • 13
    • 1042278120 scopus 로고    scopus 로고
    • Defining ischemic burden after traumatic brain injury using 15O PET imaging of cerebral physiology
    • Coles JP, Fryer TD, Smielewski P, Rice K, Clark JC, Pickard JD, et al. Defining ischemic burden after traumatic brain injury using 15O PET imaging of cerebral physiology. J Cereb Blood Flow Metab (2004) 24:191-201. doi:10.1097/01.WCB.0000100045.07481.DE
    • (2004) J Cereb Blood Flow Metab , vol.24 , pp. 191-201
    • Coles, J.P.1    Fryer, T.D.2    Smielewski, P.3    Rice, K.4    Clark, J.C.5    Pickard, J.D.6
  • 14
    • 84890555691 scopus 로고    scopus 로고
    • Traumatic intracranial aneurysm formation following closed head injury
    • Miley JT, Rodriguez GJ, Qureshi AI. Traumatic intracranial aneurysm formation following closed head injury. J Vasc Interv Neurol (2008) 1:79-82.
    • (2008) J Vasc Interv Neurol , vol.1 , pp. 79-82
    • Miley, J.T.1    Rodriguez, G.J.2    Qureshi, A.I.3
  • 16
    • 49249127488 scopus 로고    scopus 로고
    • Prevalence of chronic pain after traumatic brain injury: a systematic review
    • Nampiaparampil DE. Prevalence of chronic pain after traumatic brain injury: a systematic review. JAMA (2008) 300:711-9. doi:10.1001/jama.300.6.711
    • (2008) JAMA , vol.300 , pp. 711-719
    • Nampiaparampil, D.E.1
  • 17
    • 0020640495 scopus 로고
    • Monitoring respirations for outpatient surgery
    • Grande DJ, Koranda FC, Guthrie D. Monitoring respirations for outpatient surgery. J Dermatol Surg Oncol (1983) 9:338-9. doi:10.1111/j.1524-4725.1983.tb00811.x
    • (1983) J Dermatol Surg Oncol , vol.9 , pp. 338-339
    • Grande, D.J.1    Koranda, F.C.2    Guthrie, D.3
  • 18
    • 84904644453 scopus 로고    scopus 로고
    • Incidence, prevalence, and occurrence rate of infection among adults hospitalized after traumatic brain injury: study protocol for a systematic review and meta-analysis
    • Scott BN, Roberts DJ, Robertson HL, Kramer AH, Laupland KB, Ousman SS, et al. Incidence, prevalence, and occurrence rate of infection among adults hospitalized after traumatic brain injury: study protocol for a systematic review and meta-analysis. Syst Rev (2013) 2:68. doi:10.1186/2046-4053-2-68
    • (2013) Syst Rev , vol.2 , pp. 68
    • Scott, B.N.1    Roberts, D.J.2    Robertson, H.L.3    Kramer, A.H.4    Laupland, K.B.5    Ousman, S.S.6
  • 19
    • 0027360954 scopus 로고
    • Traumatic brain injury-induced excitotoxicity assessed in a controlled cortical impact model
    • Palmer AM, Marion DW, Botscheller ML, Swedlow PE, Styren SD, Dekosky ST. Traumatic brain injury-induced excitotoxicity assessed in a controlled cortical impact model. J Neurochem (1993) 61:2015-24. doi:10.1111/j.1471-4159.1993.tb07437.x
    • (1993) J Neurochem , vol.61 , pp. 2015-2024
    • Palmer, A.M.1    Marion, D.W.2    Botscheller, M.L.3    Swedlow, P.E.4    Styren, S.D.5    Dekosky, S.T.6
  • 21
    • 84930029306 scopus 로고    scopus 로고
    • Traumatic brain injury in later life increases risk for Parkinson's disease
    • Gardner RC, Burke JF, Nettiksimmons J, Goldman S, Tanner CM, Yaffe K. Traumatic brain injury in later life increases risk for Parkinson's disease. Ann Neurol (2015) 77(6):987-95. doi:10.1002/ana.24396
    • (2015) Ann Neurol , vol.77 , Issue.6 , pp. 987-995
    • Gardner, R.C.1    Burke, J.F.2    Nettiksimmons, J.3    Goldman, S.4    Tanner, C.M.5    Yaffe, K.6
  • 22
    • 0037967245 scopus 로고    scopus 로고
    • Head injury as a risk factor for Alzheimer's disease: the evidence 10 years on; a partial replication
    • Fleminger S, Oliver DL, Lovestone S, Rabe-Hesketh S, Giora A. Head injury as a risk factor for Alzheimer's disease: the evidence 10 years on; a partial replication. J Neurol Neurosurg Psychiatry (2003) 74:857-62. doi:10.1136/jnnp.74.7.857
    • (2003) J Neurol Neurosurg Psychiatry , vol.74 , pp. 857-862
    • Fleminger, S.1    Oliver, D.L.2    Lovestone, S.3    Rabe-Hesketh, S.4    Giora, A.5
  • 23
    • 0031975401 scopus 로고    scopus 로고
    • A population-based study of seizures after traumatic brain injuries
    • Annegers JF, Hauser WA, Coan SP, Rocca WA. A population-based study of seizures after traumatic brain injuries. N Engl J Med (1998) 338:20-4. doi:10.1056/NEJM199801013380104
    • (1998) N Engl J Med , vol.338 , pp. 20-24
    • Annegers, J.F.1    Hauser, W.A.2    Coan, S.P.3    Rocca, W.A.4
  • 24
    • 84863081457 scopus 로고    scopus 로고
    • Increased risk of multiple sclerosis after traumatic brain injury: a nationwide population-based study
    • Kang JH, Lin HC. Increased risk of multiple sclerosis after traumatic brain injury: a nationwide population-based study. J Neurotrauma (2012) 29:90-5. doi:10.1089/neu.2011.1936
    • (2012) J Neurotrauma , vol.29 , pp. 90-95
    • Kang, J.H.1    Lin, H.C.2
  • 27
    • 55749111251 scopus 로고    scopus 로고
    • Pharmacology of traumatic brain injury: where is the "golden bullet"?
    • Beauchamp K, Mutlak H, Smith WR, Shohami E, Stahel PF. Pharmacology of traumatic brain injury: where is the "golden bullet"? Mol Med (2008) 14:731-40. doi:10.2119/2008-00050.Beauchamp
    • (2008) Mol Med , vol.14 , pp. 731-740
    • Beauchamp, K.1    Mutlak, H.2    Smith, W.R.3    Shohami, E.4    Stahel, P.F.5
  • 29
    • 84862675799 scopus 로고    scopus 로고
    • Updated report on comparative effectiveness of ACE inhibitors, ARBs, and direct renin inhibitors for patients with essential hypertension: much more data, little new information
    • Powers BJ, Coeytaux RR, Dolor RJ, Hasselblad V, Patel UD, Yancy WS Jr, et al. Updated report on comparative effectiveness of ACE inhibitors, ARBs, and direct renin inhibitors for patients with essential hypertension: much more data, little new information. J Gen Intern Med (2012) 27:716-29. doi:10.1007/s11606-011-1938-8
    • (2012) J Gen Intern Med , vol.27 , pp. 716-729
    • Powers, B.J.1    Coeytaux, R.R.2    Dolor, R.J.3    Hasselblad, V.4    Patel, U.D.5    Yancy, W.S.6
  • 30
    • 84922960278 scopus 로고    scopus 로고
    • Combination dolutegravir-abacavir-lamivudine in the management of HIV/AIDS: clinical utility and patient considerations
    • Cruciani M, Malena M. Combination dolutegravir-abacavir-lamivudine in the management of HIV/AIDS: clinical utility and patient considerations. Patient Prefer Adherence (2015) 9:299-310. doi:10.2147/PPA.S65199
    • (2015) Patient Prefer Adherence , vol.9 , pp. 299-310
    • Cruciani, M.1    Malena, M.2
  • 31
    • 79951702955 scopus 로고    scopus 로고
    • Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives
    • Chen D, Frezza M, Schmitt S, Kanwar J, Dou QP. Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives. Curr Cancer Drug Targets (2011) 11:239-53. doi:10.2174/156800911794519752
    • (2011) Curr Cancer Drug Targets , vol.11 , pp. 239-253
    • Chen, D.1    Frezza, M.2    Schmitt, S.3    Kanwar, J.4    Dou, Q.P.5
  • 32
    • 33846164404 scopus 로고    scopus 로고
    • Emerging roles of cysteine cathepsins in disease and their potential as drug targets
    • Vasiljeva O, Reinheckel T, Peters C, Turk D, Turk V, Turk B. Emerging roles of cysteine cathepsins in disease and their potential as drug targets. Curr Pharm Des (2007) 13:387-403. doi:10.2174/138161207779313542
    • (2007) Curr Pharm Des , vol.13 , pp. 387-403
    • Vasiljeva, O.1    Reinheckel, T.2    Peters, C.3    Turk, D.4    Turk, V.5    Turk, B.6
  • 33
    • 84873894410 scopus 로고    scopus 로고
    • Cathepsin B as a cancer target
    • Gondi CS, Rao JS. Cathepsin B as a cancer target. Expert Opin Ther Targets (2013) 17:281-91. doi:10.1517/14728222.2013.740461
    • (2013) Expert Opin Ther Targets , vol.17 , pp. 281-291
    • Gondi, C.S.1    Rao, J.S.2
  • 34
    • 37549070672 scopus 로고    scopus 로고
    • Cysteine cathepsin proteases as pharmacological targets in cancer
    • Palermo C, Joyce JA. Cysteine cathepsin proteases as pharmacological targets in cancer. Trends Pharmacol Sci (2008) 29:22-8. doi:10.1016/j.tips.2007.10.011
    • (2008) Trends Pharmacol Sci , vol.29 , pp. 22-28
    • Palermo, C.1    Joyce, J.A.2
  • 35
    • 0038486779 scopus 로고    scopus 로고
    • Molecular regulation of human cathepsin B: implication in pathologies
    • Yan S, Sloane BF. Molecular regulation of human cathepsin B: implication in pathologies. Biol Chem (2003) 384:845-54. doi:10.1515/BC.2003.095
    • (2003) Biol Chem , vol.384 , pp. 845-854
    • Yan, S.1    Sloane, B.F.2
  • 36
    • 84906771257 scopus 로고    scopus 로고
    • The current stage of cathepsin B inhibitors as potential anticancer agents
    • Kos J, Mitrovic A, Mirkovic B. The current stage of cathepsin B inhibitors as potential anticancer agents. Future Med Chem (2014) 6:1355-71. doi:10.4155/fmc.14.73
    • (2014) Future Med Chem , vol.6 , pp. 1355-1371
    • Kos, J.1    Mitrovic, A.2    Mirkovic, B.3
  • 38
  • 40
    • 27144524157 scopus 로고    scopus 로고
    • Amelioration of experimental arthritis by a calpain-inhibitory compound: regulation of cytokine production by E-64-d in vivo and in vitro
    • Yoshifuji H, Umehara H, Maruyama H, Itoh M, Tanaka M, Kawabata D, et al. Amelioration of experimental arthritis by a calpain-inhibitory compound: regulation of cytokine production by E-64-d in vivo and in vitro. Int Immunol (2005) 17:1327-36. doi:10.1093/intimm/dxh311
    • (2005) Int Immunol , vol.17 , pp. 1327-1336
    • Yoshifuji, H.1    Umehara, H.2    Maruyama, H.3    Itoh, M.4    Tanaka, M.5    Kawabata, D.6
  • 41
    • 19144365133 scopus 로고    scopus 로고
    • Endosomal proteolysis of the Ebola virus glycoprotein is necessary for infection
    • Chandran K, Sullivan NJ, Felbor U, Whelan SP, Cunningham JM. Endosomal proteolysis of the Ebola virus glycoprotein is necessary for infection. Science (2005) 308:1643-5. doi:10.1126/science.1110656
    • (2005) Science , vol.308 , pp. 1643-1645
    • Chandran, K.1    Sullivan, N.J.2    Felbor, U.3    Whelan, S.P.4    Cunningham, J.M.5
  • 42
    • 81455154929 scopus 로고    scopus 로고
    • The NLRP3 inflammasome contributes to brain injury in pneumococcal meningitis and is activated through ATP-dependent lysosomal cathepsin B release
    • Hoegen T, Tremel N, Klein M, Angele B, Wagner H, Kirschning C, et al. The NLRP3 inflammasome contributes to brain injury in pneumococcal meningitis and is activated through ATP-dependent lysosomal cathepsin B release. J Immunol (2011) 187:5440-51. doi:10.4049/jimmunol.1100790
    • (2011) J Immunol , vol.187 , pp. 5440-5451
    • Hoegen, T.1    Tremel, N.2    Klein, M.3    Angele, B.4    Wagner, H.5    Kirschning, C.6
  • 43
    • 57049113201 scopus 로고    scopus 로고
    • Development of protease inhibitors for protozoan infections
    • McKerrow JH, Rosenthal PJ, Swenerton R, Doyle P. Development of protease inhibitors for protozoan infections. Curr Opin Infect Dis (2008) 21:668-72. doi:10.1097/QCO.0b013e328315cca9
    • (2008) Curr Opin Infect Dis , vol.21 , pp. 668-672
    • McKerrow, J.H.1    Rosenthal, P.J.2    Swenerton, R.3    Doyle, P.4
  • 44
    • 79954514102 scopus 로고    scopus 로고
    • Apoptosis in nonalcoholic fatty liver disease: diagnostic and therapeutic implications
    • Alkhouri N, Carter-Kent C, Feldstein AE. Apoptosis in nonalcoholic fatty liver disease: diagnostic and therapeutic implications. Expert Rev Gastroenterol Hepatol (2011) 5:201-12. doi:10.1586/egh.11.6
    • (2011) Expert Rev Gastroenterol Hepatol , vol.5 , pp. 201-212
    • Alkhouri, N.1    Carter-Kent, C.2    Feldstein, A.E.3
  • 45
    • 79954478174 scopus 로고    scopus 로고
    • Efficacy of a reversible cathepsin B inhibitor in a rodent model of liver fibrosis and human pharmacokinetic profile
    • Holsinger LDC, Dener J, Green M, Booth R, Dalrymple S. Efficacy of a reversible cathepsin B inhibitor in a rodent model of liver fibrosis and human pharmacokinetic profile. Hepatology (2010) 52:1128A.
    • (2010) Hepatology , vol.52 , pp. 1128A
    • Holsinger, L.D.C.1    Dener, J.2    Green, M.3    Booth, R.4    Dalrymple, S.5
  • 46
    • 35848971033 scopus 로고    scopus 로고
    • A cysteine protease inhibitor cures Chagas' disease in an immunodeficient-mouse model of infection
    • Doyle PS, Zhou YM, Engel JC, Mckerrow JH. A cysteine protease inhibitor cures Chagas' disease in an immunodeficient-mouse model of infection. Antimicrob Agents Chemother (2007) 51:3932-9. doi:10.1128/AAC.00436-07
    • (2007) Antimicrob Agents Chemother , vol.51 , pp. 3932-3939
    • Doyle, P.S.1    Zhou, Y.M.2    Engel, J.C.3    Mckerrow, J.H.4
  • 47
    • 84930706588 scopus 로고    scopus 로고
    • Proteases in traumatic brain injury
    • Lendeckel U, Hooper NM, editors. Springer
    • Knoblach SM, Faden AI. Proteases in traumatic brain injury. In: Lendeckel U, Hooper NM, editors. Proteases in the Brain. Springer (2005).
    • (2005) Proteases in the Brain
    • Knoblach, S.M.1    Faden, A.I.2
  • 48
    • 84896829756 scopus 로고    scopus 로고
    • The cysteine protease cathepsin B is a key drug target and cysteine protease inhibitors are potential therapeutics for traumatic brain injury
    • Hook GR, Yu J, Sipes N, Pierschbacher MD, Hook V, Kindy MS. The cysteine protease cathepsin B is a key drug target and cysteine protease inhibitors are potential therapeutics for traumatic brain injury. J Neurotrauma (2014) 31:515-29. doi:10.1089/neu.2013.2944
    • (2014) J Neurotrauma , vol.31 , pp. 515-529
    • Hook, G.R.1    Yu, J.2    Sipes, N.3    Pierschbacher, M.D.4    Hook, V.5    Kindy, M.S.6
  • 49
    • 84892697645 scopus 로고    scopus 로고
    • Pharmacotherapy of traumatic brain injury: state of the science and the road forward report of the Department of Defense Neurotrauma Pharmacology Workgroup
    • Diaz-Arrastia R, Kochanek PM, Bergold P, Kenney K, Marx C, Grimes J, et al. Pharmacotherapy of traumatic brain injury: state of the science and the road forward report of the Department of Defense Neurotrauma Pharmacology Workgroup. J Neurotrauma (2013) 31(2):135-58. doi:10.1089/neu.2013.3019
    • (2013) J Neurotrauma , vol.31 , Issue.2 , pp. 135-158
    • Diaz-Arrastia, R.1    Kochanek, P.M.2    Bergold, P.3    Kenney, K.4    Marx, C.5    Grimes, J.6
  • 50
    • 84892603033 scopus 로고    scopus 로고
    • Neuroprotective strategies for traumatic brain injury: improving clinical translation
    • Kabadi SV, Faden AI. Neuroprotective strategies for traumatic brain injury: improving clinical translation. Int J Mol Sci (2014) 15:1216-36. doi:10.3390/ijms15011216
    • (2014) Int J Mol Sci , vol.15 , pp. 1216-1236
    • Kabadi, S.V.1    Faden, A.I.2
  • 52
    • 0003316192 scopus 로고
    • On the proteolytic enzymes of animal tissues: beef spleen
    • Fruton JS, Bergmann M. On the proteolytic enzymes of animal tissues: beef spleen. J Biol Chem (1939) 130:19-27.
    • (1939) J Biol Chem , vol.130 , pp. 19-27
    • Fruton, J.S.1    Bergmann, M.2
  • 53
    • 2642612296 scopus 로고
    • One the proteolytic enzymes of beef spleen, beef kidney, and swine kidney. Classification of the cathepsins
    • Fruton JS, Irving GW, Bergmann M. One the proteolytic enzymes of beef spleen, beef kidney, and swine kidney. Classification of the cathepsins. J Biol Chem (1941) 138:249-62.
    • (1941) J Biol Chem , vol.138 , pp. 249-262
    • Fruton, J.S.1    Irving, G.W.2    Bergmann, M.3
  • 54
    • 0000428279 scopus 로고
    • On the proteolytic enzymes of animal tissues. X. Beef spleen cathepsin C
    • Tallan HH, Jones ME, Fruton JS. On the proteolytic enzymes of animal tissues. X. Beef spleen cathepsin C. J Biol Chem (1952) 194:793-805.
    • (1952) J Biol Chem , vol.194 , pp. 793-805
    • Tallan, H.H.1    Jones, M.E.2    Fruton, J.S.3
  • 55
    • 0344268937 scopus 로고
    • Purification and properties of beef spleen cathepsin B
    • Greenbaum LM, Fruton JS. Purification and properties of beef spleen cathepsin B. J Biol Chem (1957) 226:173-80.
    • (1957) J Biol Chem , vol.226 , pp. 173-180
    • Greenbaum, L.M.1    Fruton, J.S.2
  • 56
    • 0020770394 scopus 로고
    • Homology of amino acid sequences of rat liver cathepsins B and H with that of papain
    • Takio K, Towatari T, Katunuma N, Teller DC, Titani K. Homology of amino acid sequences of rat liver cathepsins B and H with that of papain. Proc Natl Acad Sci U S A (1983) 80:3666-70. doi:10.1073/pnas.80.12.3666
    • (1983) Proc Natl Acad Sci U S A , vol.80 , pp. 3666-3670
    • Takio, K.1    Towatari, T.2    Katunuma, N.3    Teller, D.C.4    Titani, K.5
  • 57
    • 0000249898 scopus 로고
    • Nucleotide and predicted amino acid sequences of cloned human and mouse preprocathepsin B cDNAs
    • Chan SJ, San Segundo B, Mccormick MB, Steiner DF. Nucleotide and predicted amino acid sequences of cloned human and mouse preprocathepsin B cDNAs. Proc Natl Acad Sci U S A (1986) 83:7721-5. doi:10.1073/pnas.83.20.7721
    • (1986) Proc Natl Acad Sci U S A , vol.83 , pp. 7721-7725
    • Chan, S.J.1    San Segundo, B.2    Mccormick, M.B.3    Steiner, D.F.4
  • 58
    • 12044253640 scopus 로고
    • The refined 2.15 A X-ray crystal structure of human liver cathepsin B: the structural basis for its specificity
    • Musil D, Zucic D, Turk D, Engh RA, Mayr I, Huber R, et al. The refined 2.15 A X-ray crystal structure of human liver cathepsin B: the structural basis for its specificity. EMBO J (1991) 10:2321-30.
    • (1991) EMBO J , vol.10 , pp. 2321-2330
    • Musil, D.1    Zucic, D.2    Turk, D.3    Engh, R.A.4    Mayr, I.5    Huber, R.6
  • 59
    • 0032516003 scopus 로고    scopus 로고
    • Cathepsins B and D are dispensable for major histocompatibility complex class II-mediated antigen presentation
    • Deussing J, Roth W, Saftig P, Peters C, Ploegh HL, Villadangos JA. Cathepsins B and D are dispensable for major histocompatibility complex class II-mediated antigen presentation. Proc Natl Acad Sci U S A (1998) 95:4516-21. doi:10.1073/pnas.95.8.4516
    • (1998) Proc Natl Acad Sci U S A , vol.95 , pp. 4516-4521
    • Deussing, J.1    Roth, W.2    Saftig, P.3    Peters, C.4    Ploegh, H.L.5    Villadangos, J.A.6
  • 60
    • 33746893630 scopus 로고    scopus 로고
    • 333. Cathepsin B
    • Second ed. Barrett AJ, Rawlings ND, Woessner JF, editors. Amsterdam: Elsevier Academic Press
    • Mort JS. 333. Cathepsin B. Second ed. In: Barrett AJ, Rawlings ND, Woessner JF, editors. Handbook of Proteolytic Enzymes. Amsterdam: Elsevier Academic Press (2004) 1079-86.
    • (2004) Handbook of Proteolytic Enzymes , pp. 1079-1086
    • Mort, J.S.1
  • 61
    • 0017886726 scopus 로고
    • The specificity of cathepsin B. Hydrolysis of glucagon at the C-terminus by a peptidyldipeptidase mechanism
    • Aronson NN Jr, Barrett AJ. The specificity of cathepsin B. Hydrolysis of glucagon at the C-terminus by a peptidyldipeptidase mechanism. Biochem J (1978) 171:759-65.
    • (1978) Biochem J , vol.171 , pp. 759-765
    • Aronson, N.N.1    Barrett, A.J.2
  • 64
    • 0014211618 scopus 로고
    • On the size of the active site in proteases. I. Papain
    • Schechter I, Berger A. On the size of the active site in proteases. I. Papain. Biochem Biophys Res Commun (1967) 27:157-62. doi:10.1016/S0006-291X(67)80055-X
    • (1967) Biochem Biophys Res Commun , vol.27 , pp. 157-162
    • Schechter, I.1    Berger, A.2
  • 65
    • 20944437268 scopus 로고    scopus 로고
    • High throughput substrate specificity profiling of serine and cysteine proteases using solution-phase fluorogenic peptide microarrays
    • Gosalia DN, Salisbury CM, Ellman JA, Diamond SL. High throughput substrate specificity profiling of serine and cysteine proteases using solution-phase fluorogenic peptide microarrays. Mol Cell Proteomics (2005) 4:626-36. doi:10.1074/mcp.M500004-MCP200
    • (2005) Mol Cell Proteomics , vol.4 , pp. 626-636
    • Gosalia, D.N.1    Salisbury, C.M.2    Ellman, J.A.3    Diamond, S.L.4
  • 66
    • 33744961634 scopus 로고    scopus 로고
    • Substrate profiling of cysteine proteases using a combinatorial peptide library identifies functionally unique specificities
    • Choe Y, Leonetti F, Greenbaum DC, Lecaille F, Bogyo M, Bromme D, et al. Substrate profiling of cysteine proteases using a combinatorial peptide library identifies functionally unique specificities. J Biol Chem (2006) 281:12824-32. doi:10.1074/jbc.M513331200
    • (2006) J Biol Chem , vol.281 , pp. 12824-12832
    • Choe, Y.1    Leonetti, F.2    Greenbaum, D.C.3    Lecaille, F.4    Bogyo, M.5    Bromme, D.6
  • 68
    • 0014405092 scopus 로고
    • On the active site of proteases. 3. Mapping the active site of papain; specific peptide inhibitors of papain
    • Schechter I, Berger A. On the active site of proteases. 3. Mapping the active site of papain; specific peptide inhibitors of papain. Biochem Biophys Res Commun (1968) 32:898-902. doi:10.1016/0006-291X(68)90326-4
    • (1968) Biochem Biophys Res Commun , vol.32 , pp. 898-902
    • Schechter, I.1    Berger, A.2
  • 69
    • 50849088334 scopus 로고    scopus 로고
    • Alternative pathways for production of beta-amyloid peptides of Alzheimer's disease
    • Hook V, Schechter I, Demuth HU, Hook G. Alternative pathways for production of beta-amyloid peptides of Alzheimer's disease. Biol Chem (2008) 389:993-1006. doi:10.1515/BC.2008.124
    • (2008) Biol Chem , vol.389 , pp. 993-1006
    • Hook, V.1    Schechter, I.2    Demuth, H.U.3    Hook, G.4
  • 70
    • 82755161948 scopus 로고    scopus 로고
    • Cysteine cathepsins: from structure, function and regulation to new frontiers
    • Turk V, Stoka V, Vasiljeva O, Renko M, Sun T, Turk B, et al. Cysteine cathepsins: from structure, function and regulation to new frontiers. Biochim Biophys Acta (2012) 1824:68-88. doi:10.1016/j.bbapap.2011.10.002
    • (2012) Biochim Biophys Acta , vol.1824 , pp. 68-88
    • Turk, V.1    Stoka, V.2    Vasiljeva, O.3    Renko, M.4    Sun, T.5    Turk, B.6
  • 71
    • 2942718876 scopus 로고    scopus 로고
    • Comprehensive search for cysteine cathepsins in the human genome
    • Rossi A, Deveraux Q, Turk B, Sali A. Comprehensive search for cysteine cathepsins in the human genome. Biol Chem (2004) 385:363-72. doi:10.1515/BC.2004.040
    • (2004) Biol Chem , vol.385 , pp. 363-372
    • Rossi, A.1    Deveraux, Q.2    Turk, B.3    Sali, A.4
  • 72
    • 0026518253 scopus 로고
    • Confirmation of the human cathepsin B gene (CTSB) assignment to chromosome 8
    • Fong D, Chan MM, Hsieh WT, Menninger JC, Ward DC. Confirmation of the human cathepsin B gene (CTSB) assignment to chromosome 8. Hum Genet (1992) 89:10-2. doi:10.1007/BF00207033
    • (1992) Hum Genet , vol.89 , pp. 10-12
    • Fong, D.1    Chan, M.M.2    Hsieh, W.T.3    Menninger, J.C.4    Ward, D.C.5
  • 73
    • 0029048724 scopus 로고
    • Identification of two new exons and multiple transcription start points in the 5'-untranslated region of the human cathepsin-B-encoding gene
    • Berquin IM, Cao L, Fong D, Sloane BF. Identification of two new exons and multiple transcription start points in the 5'-untranslated region of the human cathepsin-B-encoding gene. Gene (1995) 159:143-9. doi:10.1016/0378-1119(95)00072-E
    • (1995) Gene , vol.159 , pp. 143-149
    • Berquin, I.M.1    Cao, L.2    Fong, D.3    Sloane, B.F.4
  • 74
    • 0027319177 scopus 로고
    • Characterization of the cathepsin B gene and multiple mRNAs in human tissues: evidence for alternative splicing of cathepsin B pre-mRNA
    • Gong Q, Chan SJ, Bajkowski AS, Steiner DF, Frankfater A. Characterization of the cathepsin B gene and multiple mRNAs in human tissues: evidence for alternative splicing of cathepsin B pre-mRNA. DNA Cell Biol (1993) 12:299-309. doi:10.1089/dna.1993.12.299
    • (1993) DNA Cell Biol , vol.12 , pp. 299-309
    • Gong, Q.1    Chan, S.J.2    Bajkowski, A.S.3    Steiner, D.F.4    Frankfater, A.5
  • 75
    • 67349208210 scopus 로고    scopus 로고
    • Chromosome 8p as a potential hub for developmental neuropsychiatric disorders: implications for schizophrenia, autism and cancer
    • Tabares-Seisdedos R, Rubenstein JL. Chromosome 8p as a potential hub for developmental neuropsychiatric disorders: implications for schizophrenia, autism and cancer. Mol Psychiatry (2009) 14:563-89. doi:10.1038/mp.2009.2
    • (2009) Mol Psychiatry , vol.14 , pp. 563-589
    • Tabares-Seisdedos, R.1    Rubenstein, J.L.2
  • 76
    • 33747773624 scopus 로고    scopus 로고
    • Association of cathepsin B gene polymorphisms with tropical calcific pancreatitis
    • Mahurkar S, Idris MM, Reddy DN, Bhaskar S, Rao GV, Thomas V, et al. Association of cathepsin B gene polymorphisms with tropical calcific pancreatitis. Gut (2006) 55:1270-5. doi:10.1136/gut.2005.087403
    • (2006) Gut , vol.55 , pp. 1270-1275
    • Mahurkar, S.1    Idris, M.M.2    Reddy, D.N.3    Bhaskar, S.4    Rao, G.V.5    Thomas, V.6
  • 77
    • 0025728140 scopus 로고
    • The structure of the mouse cathepsin B gene and its putative promoter
    • Qian F, Frankfater A, Chan SJ, Steiner DF. The structure of the mouse cathepsin B gene and its putative promoter. DNA Cell Biol (1991) 10:159-68. doi:10.1089/dna.1991.10.159
    • (1991) DNA Cell Biol , vol.10 , pp. 159-168
    • Qian, F.1    Frankfater, A.2    Chan, S.J.3    Steiner, D.F.4
  • 78
    • 0031109750 scopus 로고    scopus 로고
    • The genes of the lysosomal cysteine proteinases cathepsin B, H, L, and S map to different mouse chromosomes
    • Deussing J, Roth W, Rommerskirch W, Wiederanders B, Von Figura K, Peters C. The genes of the lysosomal cysteine proteinases cathepsin B, H, L, and S map to different mouse chromosomes. Mamm Genome (1997) 8:241-5. doi:10.1007/s003359900401
    • (1997) Mamm Genome , vol.8 , pp. 241-245
    • Deussing, J.1    Roth, W.2    Rommerskirch, W.3    Wiederanders, B.4    Von Figura, K.5    Peters, C.6
  • 80
    • 60149088848 scopus 로고    scopus 로고
    • Origins and mechanisms of miRNAs and siRNAs
    • Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs. Cell (2009) 136:642-55. doi:10.1016/j.cell.2009.01.035
    • (2009) Cell , vol.136 , pp. 642-655
    • Carthew, R.W.1    Sontheimer, E.J.2
  • 81
    • 1942494040 scopus 로고    scopus 로고
    • Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation
    • Sempere LF, Freemantle S, Pitha-Rowe I, Moss E, Dmitrovsky E, Ambros V. Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation. Genome Biol (2004) 5:R13. doi:10.1186/gb-2004-5-3-r13
    • (2004) Genome Biol , vol.5 , pp. R13
    • Sempere, L.F.1    Freemantle, S.2    Pitha-Rowe, I.3    Moss, E.4    Dmitrovsky, E.5    Ambros, V.6
  • 82
    • 84878945439 scopus 로고    scopus 로고
    • The emerging roles of microRNAs in CNS injuries
    • Bhalala OG, Srikanth M, Kessler JA. The emerging roles of microRNAs in CNS injuries. Nat Rev Neurol (2013) 9:328-39. doi:10.1038/nrneurol.2013.67
    • (2013) Nat Rev Neurol , vol.9 , pp. 328-339
    • Bhalala, O.G.1    Srikanth, M.2    Kessler, J.A.3
  • 83
    • 65549158252 scopus 로고    scopus 로고
    • Traumatic brain injury alters expression of hippocampal microRNAs: potential regulators of multiple pathophysiological processes
    • Redell JB, Liu Y, Dash PK. Traumatic brain injury alters expression of hippocampal microRNAs: potential regulators of multiple pathophysiological processes. J Neurosci Res (2009) 87:1435-48. doi:10.1002/jnr.21945
    • (2009) J Neurosci Res , vol.87 , pp. 1435-1448
    • Redell, J.B.1    Liu, Y.2    Dash, P.K.3
  • 84
    • 84872726735 scopus 로고    scopus 로고
    • MicroRNA 218 acts as a tumor suppressor by targeting multiple cancer phenotype-associated genes in medulloblastoma
    • Venkataraman S, Birks DK, Balakrishnan I, Alimova I, Harris PS, Patel PR, et al. MicroRNA 218 acts as a tumor suppressor by targeting multiple cancer phenotype-associated genes in medulloblastoma. J Biol Chem (2013) 288:1918-28. doi:10.1074/jbc.M112.396762
    • (2013) J Biol Chem , vol.288 , pp. 1918-1928
    • Venkataraman, S.1    Birks, D.K.2    Balakrishnan, I.3    Alimova, I.4    Harris, P.S.5    Patel, P.R.6
  • 85
    • 84887243168 scopus 로고    scopus 로고
    • miR128 up-regulation correlates with impaired amyloid beta(1-42) degradation in monocytes from patients with sporadic Alzheimer's disease
    • Tiribuzi R, Crispoltoni L, Porcellati S, Di Lullo M, Florenzano F, Pirro M, et al. miR128 up-regulation correlates with impaired amyloid beta(1-42) degradation in monocytes from patients with sporadic Alzheimer's disease. Neurobiol Aging (2014) 35:345-56. doi:10.1016/j.neurobiolaging.2013.08.003
    • (2014) Neurobiol Aging , vol.35 , pp. 345-356
    • Tiribuzi, R.1    Crispoltoni, L.2    Porcellati, S.3    Di Lullo, M.4    Florenzano, F.5    Pirro, M.6
  • 86
    • 33846833594 scopus 로고    scopus 로고
    • Post-transcriptional regulation of the let-7 microRNA during neural cell specification
    • Wulczyn FG, Smirnova L, Rybak A, Brandt C, Kwidzinski E, Ninnemann O, et al. Post-transcriptional regulation of the let-7 microRNA during neural cell specification. FASEB J (2007) 21:415-26. doi:10.1096/fj.06-6130com
    • (2007) FASEB J , vol.21 , pp. 415-426
    • Wulczyn, F.G.1    Smirnova, L.2    Rybak, A.3    Brandt, C.4    Kwidzinski, E.5    Ninnemann, O.6
  • 87
    • 84871917589 scopus 로고    scopus 로고
    • Posttranslational processing and modification of cathepsins and cystatins
    • Katunuma N. Posttranslational processing and modification of cathepsins and cystatins. J Signal Transduct (2010) 2010:375345. doi:10.1155/2010/375345
    • (2010) J Signal Transduct , vol.2010
    • Katunuma, N.1
  • 88
    • 0034615570 scopus 로고    scopus 로고
    • Lysosomal cysteine proteases: more than scavengers
    • Turk B, Turk D, Turk V. Lysosomal cysteine proteases: more than scavengers. Biochim Biophys Acta (2000) 1477:98-111. doi:10.1016/S0167-4838(99)00263-0
    • (2000) Biochim Biophys Acta , vol.1477 , pp. 98-111
    • Turk, B.1    Turk, D.2    Turk, V.3
  • 89
    • 37049178293 scopus 로고
    • Evolution of proteolytic enzymes
    • Neurath H. Evolution of proteolytic enzymes. Science (1984) 224:350-7. doi:10.1126/science.6369538
    • (1984) Science , vol.224 , pp. 350-357
    • Neurath, H.1
  • 90
    • 58449134643 scopus 로고    scopus 로고
    • Autocatalytic processing of procathepsin B is triggered by proenzyme activity
    • Pungercar JR, Caglic D, Sajid M, Dolinar M, Vasiljeva O, Pozgan U, et al. Autocatalytic processing of procathepsin B is triggered by proenzyme activity. FEBS J (2009) 276:660-8. doi:10.1111/j.1742-4658.2008.06815.x
    • (2009) FEBS J , vol.276 , pp. 660-668
    • Pungercar, J.R.1    Caglic, D.2    Sajid, M.3    Dolinar, M.4    Vasiljeva, O.5    Pozgan, U.6
  • 91
    • 0028237920 scopus 로고
    • Maturation of human procathepsin B. Proenzyme activation and proteolytic processing of the precursor to the mature proteinase, in vitro, are primarily unimolecular processes
    • Mach L, Mort JS, Glossl J. Maturation of human procathepsin B. Proenzyme activation and proteolytic processing of the precursor to the mature proteinase, in vitro, are primarily unimolecular processes. J Biol Chem (1994) 269:13030-5.
    • (1994) J Biol Chem , vol.269 , pp. 13030-13035
    • Mach, L.1    Mort, J.S.2    Glossl, J.3
  • 93
    • 0142057464 scopus 로고    scopus 로고
    • Gene expression profile changes are commonly modulated across models and species after traumatic brain injury
    • Natale JE, Ahmed F, Cernak I, Stoica B, Faden AI. Gene expression profile changes are commonly modulated across models and species after traumatic brain injury. J Neurotrauma (2003) 20:907-27. doi:10.1089/089771503770195777
    • (2003) J Neurotrauma , vol.20 , pp. 907-927
    • Natale, J.E.1    Ahmed, F.2    Cernak, I.3    Stoica, B.4    Faden, A.I.5
  • 94
    • 77957239085 scopus 로고    scopus 로고
    • Cathepsin B contributes to traumatic brain injury-induced cell death through a mitochondria-mediated apoptotic pathway
    • Luo CL, Chen XP, Yang R, Sun YX, Li QQ, Bao HJ, et al. Cathepsin B contributes to traumatic brain injury-induced cell death through a mitochondria-mediated apoptotic pathway. J Neurosci Res (2010) 88:2847-58. doi:10.1002/jnr.22453
    • (2010) J Neurosci Res , vol.88 , pp. 2847-2858
    • Luo, C.L.1    Chen, X.P.2    Yang, R.3    Sun, Y.X.4    Li, Q.Q.5    Bao, H.J.6
  • 95
    • 84899148921 scopus 로고    scopus 로고
    • Hydrogen sulfide offers neuroprotection on traumatic brain injury in parallel with reduced apoptosis and autophagy in mice
    • Zhang M, Shan H, Chang P, Wang T, Dong W, Chen X, et al. Hydrogen sulfide offers neuroprotection on traumatic brain injury in parallel with reduced apoptosis and autophagy in mice. PLoS One (2014) 9:e87241. doi:10.1371/journal.pone.0087241
    • (2014) PLoS One , vol.9
    • Zhang, M.1    Shan, H.2    Chang, P.3    Wang, T.4    Dong, W.5    Chen, X.6
  • 96
    • 47649100288 scopus 로고    scopus 로고
    • Expression of cathepsin-B and-D in rat's brain after traumatic brain injury
    • Zhang YB, Chen XP, Tao LY, Qin ZH, Li SX, Yang L, et al. Expression of cathepsin-B and-D in rat's brain after traumatic brain injury. Fa Yi Xue Za Zhi (2006) 22(404-6):410.
    • (2006) Fa Yi Xue Za Zhi , vol.22 , pp. 404-406+410
    • Zhang, Y.B.1    Chen, X.P.2    Tao, L.Y.3    Qin, Z.H.4    Li, S.X.5    Yang, L.6
  • 98
    • 84879693531 scopus 로고    scopus 로고
    • Therapeutic effect of SN50, an inhibitor of nuclear factor-kappaB, in treatment of TBI in mice
    • Sun YX, Dai DK, Liu R, Wang T, Luo CL, Bao HJ, et al. Therapeutic effect of SN50, an inhibitor of nuclear factor-kappaB, in treatment of TBI in mice. Neurol Sci (2013) 34:345-55. doi:10.1007/s10072-012-1007-z
    • (2013) Neurol Sci , vol.34 , pp. 345-355
    • Sun, Y.X.1    Dai, D.K.2    Liu, R.3    Wang, T.4    Luo, C.L.5    Bao, H.J.6
  • 99
    • 0942276781 scopus 로고    scopus 로고
    • Cathepsin B mRNA and protein expression following contusion spinal cord injury in rats
    • Ellis RC, Earnhardt JN, Hayes RL, Wang KK, Anderson DK. Cathepsin B mRNA and protein expression following contusion spinal cord injury in rats. J Neurochem (2004) 88:689-97. doi:10.1046/j.1471-4159.2003.02197.x
    • (2004) J Neurochem , vol.88 , pp. 689-697
    • Ellis, R.C.1    Earnhardt, J.N.2    Hayes, R.L.3    Wang, K.K.4    Anderson, D.K.5
  • 100
    • 16244416504 scopus 로고    scopus 로고
    • Cellular localization and enzymatic activity of cathepsin B after spinal cord injury in the rat
    • Ellis RC, O'steen WA, Hayes RL, Nick HS, Wang KK, Anderson DK. Cellular localization and enzymatic activity of cathepsin B after spinal cord injury in the rat. Exp Neurol (2005) 193:19-28. doi:10.1016/j.expneurol.2004.11.034
    • (2005) Exp Neurol , vol.193 , pp. 19-28
    • Ellis, R.C.1    O'steen, W.A.2    Hayes, R.L.3    Nick, H.S.4    Wang, K.K.5    Anderson, D.K.6
  • 101
    • 67649589450 scopus 로고    scopus 로고
    • Cathepsin B release from rodent intestine mucosa due to mechanical injury results in extracellular matrix damage in early post-traumatic phases
    • Vreemann A, Qu H, Mayer K, Andersen LB, Stefana MI, Wehner S, et al. Cathepsin B release from rodent intestine mucosa due to mechanical injury results in extracellular matrix damage in early post-traumatic phases. Biol Chem (2009) 390:481-92. doi:10.1515/BC.2009.055
    • (2009) Biol Chem , vol.390 , pp. 481-492
    • Vreemann, A.1    Qu, H.2    Mayer, K.3    Andersen, L.B.4    Stefana, M.I.5    Wehner, S.6
  • 102
    • 84896035602 scopus 로고    scopus 로고
    • Possible involvement of cathepsin B/D and caspase-3 in deferoxamine-related neuroprotection of early brain injury after subarachnoid haemorrhage in rats
    • Yu ZQ, Jia Y, Chen G. Possible involvement of cathepsin B/D and caspase-3 in deferoxamine-related neuroprotection of early brain injury after subarachnoid haemorrhage in rats. Neuropathol Appl Neurobiol (2014) 40:270-83. doi:10.1111/nan.12091
    • (2014) Neuropathol Appl Neurobiol , vol.40 , pp. 270-283
    • Yu, Z.Q.1    Jia, Y.2    Chen, G.3
  • 103
    • 84937977758 scopus 로고    scopus 로고
    • The neuroprotection of lysosomotropic agents in experimental subarachnoid hemorrhage probably involving the apoptosis pathway triggering by cathepsins via chelating intralysosomal iron
    • Wang Y, Gao A, Xu X, Dang B, You W, Li H, et al. The neuroprotection of lysosomotropic agents in experimental subarachnoid hemorrhage probably involving the apoptosis pathway triggering by cathepsins via chelating intralysosomal iron. Mol Neurobiol (2014) 52(1):64-77. doi:10.1007/s12035-014-8846-y
    • (2014) Mol Neurobiol , vol.52 , Issue.1 , pp. 64-77
    • Wang, Y.1    Gao, A.2    Xu, X.3    Dang, B.4    You, W.5    Li, H.6
  • 104
    • 52449101897 scopus 로고    scopus 로고
    • Cathepsin B, K, and S are expressed in cerebral aneurysms and promote the progression of cerebral aneurysms
    • Aoki T, Kataoka H, Ishibashi R, Nozaki K, Hashimoto N. Cathepsin B, K, and S are expressed in cerebral aneurysms and promote the progression of cerebral aneurysms. Stroke (2008) 39:2603-10. doi:10.1161/STROKEAHA.107.513648
    • (2008) Stroke , vol.39 , pp. 2603-2610
    • Aoki, T.1    Kataoka, H.2    Ishibashi, R.3    Nozaki, K.4    Hashimoto, N.5
  • 105
    • 0028250042 scopus 로고
    • Causative role of lysosomal enzymes in the pathogenesis of cerebral lesions due to brain edema under chronic hypertension
    • Yamada E, Chue CH, Yukioka N, Hazama F. Causative role of lysosomal enzymes in the pathogenesis of cerebral lesions due to brain edema under chronic hypertension. Acta Neurochir Suppl (Wien) (1994) 60:83-5.
    • (1994) Acta Neurochir Suppl (Wien) , vol.60 , pp. 83-85
    • Yamada, E.1    Chue, C.H.2    Yukioka, N.3    Hazama, F.4
  • 107
    • 0030668593 scopus 로고    scopus 로고
    • Cathepsin B and middle cerebral artery occlusion in the rat
    • Seyfried D, Han Y, Zheng Z, Day N, Moin K, Rempel S, et al. Cathepsin B and middle cerebral artery occlusion in the rat. J Neurosurg (1997) 87:716-23. doi:10.3171/jns.1997.87.5.0716
    • (1997) J Neurosurg , vol.87 , pp. 716-723
    • Seyfried, D.1    Han, Y.2    Zheng, Z.3    Day, N.4    Moin, K.5    Rempel, S.6
  • 108
    • 0031817004 scopus 로고    scopus 로고
    • Inhibition of ischaemic hippocampal neuronal death in primates with cathepsin B inhibitor CA-074: a novel strategy for neuroprotection based on 'calpain-cathepsin hypothesis'
    • Yamashima T, Kohda Y, Tsuchiya K, Ueno T, Yamashita J, Yoshioka T, et al. Inhibition of ischaemic hippocampal neuronal death in primates with cathepsin B inhibitor CA-074: a novel strategy for neuroprotection based on 'calpain-cathepsin hypothesis'. Eur J Neurosci (1998) 10:1723-33. doi:10.1046/j.1460-9568.1998.00184.x
    • (1998) Eur J Neurosci , vol.10 , pp. 1723-1733
    • Yamashima, T.1    Kohda, Y.2    Tsuchiya, K.3    Ueno, T.4    Yamashita, J.5    Yoshioka, T.6
  • 109
    • 0032976738 scopus 로고    scopus 로고
    • Postictal blockade of ischemic hippocampal neuronal death in primates using selective cathepsin inhibitors
    • Tsuchiya K, Kohda Y, Yoshida M, Zhao L, Ueno T, Yamashita J, et al. Postictal blockade of ischemic hippocampal neuronal death in primates using selective cathepsin inhibitors. Exp Neurol (1999) 155:187-94. doi:10.1006/exnr.1998.6988
    • (1999) Exp Neurol , vol.155 , pp. 187-194
    • Tsuchiya, K.1    Kohda, Y.2    Yoshida, M.3    Zhao, L.4    Ueno, T.5    Yamashita, J.6
  • 110
    • 37549031387 scopus 로고    scopus 로고
    • Effects of cathepsins B and L inhibition on postischemic protein alterations in the brain
    • Anagli J, Abounit K, Stemmer P, Han Y, Allred L, Weinsheimer S, et al. Effects of cathepsins B and L inhibition on postischemic protein alterations in the brain. Biochem Biophys Res Commun (2008) 366:86-91. doi:10.1016/j.bbrc.2007.11.104
    • (2008) Biochem Biophys Res Commun , vol.366 , pp. 86-91
    • Anagli, J.1    Abounit, K.2    Stemmer, P.3    Han, Y.4    Allred, L.5    Weinsheimer, S.6
  • 111
    • 84856251253 scopus 로고    scopus 로고
    • Long-term effects of recurrent neonatal seizures on neurobehavioral function and related gene expression and its intervention by inhibitor of cathepsin B
    • Ni H, Yan JZ, Zhang LL, Feng X, Wu XR. Long-term effects of recurrent neonatal seizures on neurobehavioral function and related gene expression and its intervention by inhibitor of cathepsin B. Neurochem Res (2012) 37:31-9. doi:10.1007/s11064-011-0578-z
    • (2012) Neurochem Res , vol.37 , pp. 31-39
    • Ni, H.1    Yan, J.Z.2    Zhang, L.L.3    Feng, X.4    Wu, X.R.5
  • 112
    • 32844464580 scopus 로고    scopus 로고
    • Lysosomal enzyme cathepsin B is involved in kainic acid-induced excitotoxicity in rat striatum
    • Wang Y, Gu ZL, Cao Y, Liang ZQ, Han R, Bennett MC, et al. Lysosomal enzyme cathepsin B is involved in kainic acid-induced excitotoxicity in rat striatum. Brain Res (2006) 1071:245-9. doi:10.1016/j.brainres.2005.10.074
    • (2006) Brain Res , vol.1071 , pp. 245-249
    • Wang, Y.1    Gu, Z.L.2    Cao, Y.3    Liang, Z.Q.4    Han, R.5    Bennett, M.C.6
  • 113
    • 0021319829 scopus 로고
    • Inhibitors of prostaglandin synthesis or cathepsin B prevent muscle wasting due to sepsis in the rat
    • Ruff RL, Secrist D. Inhibitors of prostaglandin synthesis or cathepsin B prevent muscle wasting due to sepsis in the rat. J Clin Invest (1984) 73:1483-6. doi:10.1172/JCI111352
    • (1984) J Clin Invest , vol.73 , pp. 1483-1486
    • Ruff, R.L.1    Secrist, D.2
  • 114
    • 0023835222 scopus 로고
    • Evidence that cathepsin B contributes to skeletal muscle protein breakdown during sepsis
    • Hummel RP III, James JH, Warner BW, Hasselgren PO, Fischer JE. Evidence that cathepsin B contributes to skeletal muscle protein breakdown during sepsis. Arch Surg (1988) 123:221-4. doi:10.1001/archsurg.1988.01400260105013
    • (1988) Arch Surg , vol.123 , pp. 221-224
    • Hummel, R.P.1    James, J.H.2    Warner, B.W.3    Hasselgren, P.O.4    Fischer, J.E.5
  • 115
    • 84865013141 scopus 로고    scopus 로고
    • Microglial cathepsin B contributes to the initiation of peripheral inflammation-induced chronic pain
    • Sun L, Wu Z, Hayashi Y, Peters C, Tsuda M, Inoue K, et al. Microglial cathepsin B contributes to the initiation of peripheral inflammation-induced chronic pain. J Neurosci (2012) 32:11330-42. doi:10.1523/JNEUROSCI.0677-12.2012
    • (2012) J Neurosci , vol.32 , pp. 11330-11342
    • Sun, L.1    Wu, Z.2    Hayashi, Y.3    Peters, C.4    Tsuda, M.5    Inoue, K.6
  • 116
    • 73349135899 scopus 로고    scopus 로고
    • Involvement of cathepsin B in the processing and secretion of interleukin-1beta in chromogranin A-stimulated microglia
    • Terada K, Yamada J, Hayashi Y, Wu Z, Uchiyama Y, Peters C, et al. Involvement of cathepsin B in the processing and secretion of interleukin-1beta in chromogranin A-stimulated microglia. Glia (2010) 58:114-24. doi:10.1002/glia.20906
    • (2010) Glia , vol.58 , pp. 114-124
    • Terada, K.1    Yamada, J.2    Hayashi, Y.3    Wu, Z.4    Uchiyama, Y.5    Peters, C.6
  • 117
    • 69249220446 scopus 로고    scopus 로고
    • Spinal cord mRNA profile in patients with ALS: comparison with transgenic mice expressing the human SOD-1 mutant
    • Offen D, Barhum Y, Melamed E, Embacher N, Schindler C, Ransmayr G. Spinal cord mRNA profile in patients with ALS: comparison with transgenic mice expressing the human SOD-1 mutant. J Mol Neurosci (2009) 38:85-93. doi:10.1007/s12031-007-9004-z
    • (2009) J Mol Neurosci , vol.38 , pp. 85-93
    • Offen, D.1    Barhum, Y.2    Melamed, E.3    Embacher, N.4    Schindler, C.5    Ransmayr, G.6
  • 118
    • 34548157024 scopus 로고    scopus 로고
    • Microarray analysis of the cellular pathways involved in the adaptation to and progression of motor neuron injury in the SOD1 G93A mouse model of familial ALS
    • Ferraiuolo L, Heath PR, Holden H, Kasher P, Kirby J, Shaw PJ. Microarray analysis of the cellular pathways involved in the adaptation to and progression of motor neuron injury in the SOD1 G93A mouse model of familial ALS. J Neurosci (2007) 27:9201-19. doi:10.1523/JNEUROSCI.1470-07.2007
    • (2007) J Neurosci , vol.27 , pp. 9201-9219
    • Ferraiuolo, L.1    Heath, P.R.2    Holden, H.3    Kasher, P.4    Kirby, J.5    Shaw, P.J.6
  • 119
    • 84901378800 scopus 로고    scopus 로고
    • Deciphering the molecular profile of plaques, memory decline and neuron loss in two mouse models for Alzheimer's disease by deep sequencing
    • Bouter Y, Kacprowski T, Weissmann R, Dietrich K, Borgers H, Brauss A, et al. Deciphering the molecular profile of plaques, memory decline and neuron loss in two mouse models for Alzheimer's disease by deep sequencing. Front Aging Neurosci (2014) 6:75. doi:10.3389/fnagi.2014.00075
    • (2014) Front Aging Neurosci , vol.6 , pp. 75
    • Bouter, Y.1    Kacprowski, T.2    Weissmann, R.3    Dietrich, K.4    Borgers, H.5    Brauss, A.6
  • 120
    • 84924200328 scopus 로고    scopus 로고
    • Translational study of Alzheimer's disease (AD) biomarkers from brain tissues in AbetaPP/PS1 mice and serum of AD patients
    • Sun Y, Rong X, Lu W, Peng Y, Li J, Xu S, et al. Translational study of Alzheimer's disease (AD) biomarkers from brain tissues in AbetaPP/PS1 mice and serum of AD patients. J Alzheimers Dis (2015) 45:269-82. doi:10.3233/JAD-142805
    • (2015) J Alzheimers Dis , vol.45 , pp. 269-282
    • Sun, Y.1    Rong, X.2    Lu, W.3    Peng, Y.4    Li, J.5    Xu, S.6
  • 121
    • 84875740460 scopus 로고    scopus 로고
    • Meta-analysis of gene expression profiling in amyotrophic lateral sclerosis: a comparison between transgenic mouse models and human patients
    • Saris CG, Groen EJ, Koekkoek JA, Veldink JH, Van Den Berg LH. Meta-analysis of gene expression profiling in amyotrophic lateral sclerosis: a comparison between transgenic mouse models and human patients. Amyotroph Lateral Scler Frontotemporal Degener (2013) 14:177-89. doi:10.3109/21678421.2012.729842
    • (2013) Amyotroph Lateral Scler Frontotemporal Degener , vol.14 , pp. 177-189
    • Saris, C.G.1    Groen, E.J.2    Koekkoek, J.A.3    Veldink, J.H.4    Van Den Berg, L.H.5
  • 122
    • 84872658755 scopus 로고    scopus 로고
    • Expression profiles of hippocampal regenerative sprouting-related genes and their regulation by E-64d in a developmental rat model of penicillin-induced recurrent epilepticus
    • Ni H, Ren SY, Zhang LL, Sun Q, Tian T, Feng X. Expression profiles of hippocampal regenerative sprouting-related genes and their regulation by E-64d in a developmental rat model of penicillin-induced recurrent epilepticus. Toxicol Lett (2013) 217:162-9. doi:10.1016/j.toxlet.2012.12.010
    • (2013) Toxicol Lett , vol.217 , pp. 162-169
    • Ni, H.1    Ren, S.Y.2    Zhang, L.L.3    Sun, Q.4    Tian, T.5    Feng, X.6
  • 123
    • 0025129673 scopus 로고
    • Cathepsin B-indicator for the release of lysosomal cysteine proteinases in severe trauma and inflammation
    • Assfalg-Machleidt I, Jochum M, Nast-Kolb D, Siebeck M, Billing A, Joka T, et al. Cathepsin B-indicator for the release of lysosomal cysteine proteinases in severe trauma and inflammation. Biol Chem Hoppe Seyler (1990) 371(Suppl):211-22.
    • (1990) Biol Chem Hoppe Seyler , vol.371 , pp. 211-222
    • Assfalg-Machleidt, I.1    Jochum, M.2    Nast-Kolb, D.3    Siebeck, M.4    Billing, A.5    Joka, T.6
  • 124
    • 0347171553 scopus 로고
    • Phagocyte proteinases in multiple trauma and sepsi: pathomechanims and related therapeutic approaches
    • Neugebauer EA, Holaday JW, editors. CRC Press
    • Jochum M, Machleidt W, Fritz H. Phagocyte proteinases in multiple trauma and sepsi: pathomechanims and related therapeutic approaches. In: Neugebauer EA, Holaday JW, editors. Handbook of Mediators in Septic Shock. CRC Press (1993). p. 335-61.
    • (1993) Handbook of Mediators in Septic Shock , pp. 335-361
    • Jochum, M.1    Machleidt, W.2    Fritz, H.3
  • 125
    • 0034719070 scopus 로고    scopus 로고
    • Cystatin C and cathepsin B in CSF from patients with inflammatory neurologic diseases
    • Nagai A, Murakawa Y, Terashima M, Shimode K, Umegae N, Takeuchi H, et al. Cystatin C and cathepsin B in CSF from patients with inflammatory neurologic diseases. Neurology (2000) 55:1828-32. doi:10.1212/WNL.55.12.1828
    • (2000) Neurology , vol.55 , pp. 1828-1832
    • Nagai, A.1    Murakawa, Y.2    Terashima, M.3    Shimode, K.4    Umegae, N.5    Takeuchi, H.6
  • 126
    • 0029004145 scopus 로고
    • Increased cathepsin B activity in multiple sclerosis brain
    • Bever CT Jr, Garver DW. Increased cathepsin B activity in multiple sclerosis brain. J Neurol Sci (1995) 131:71-3. doi:10.1016/0022-510X(95)00039-5
    • (1995) J Neurol Sci , vol.131 , pp. 71-73
    • Bever, C.T.1    Garver, D.W.2
  • 127
    • 1442282943 scopus 로고    scopus 로고
    • Molecular signature of late-stage human ALS revealed by expression profiling of postmortem spinal cord gray matter
    • Dangond F, Hwang D, Camelo S, Pasinelli P, Frosch MP, Stephanopoulos G, et al. Molecular signature of late-stage human ALS revealed by expression profiling of postmortem spinal cord gray matter. Physiol Genomics (2004) 16:229-39. doi:10.1152/physiolgenomics.00087.2001
    • (2004) Physiol Genomics , vol.16 , pp. 229-239
    • Dangond, F.1    Hwang, D.2    Camelo, S.3    Pasinelli, P.4    Frosch, M.P.5    Stephanopoulos, G.6
  • 128
    • 0038388993 scopus 로고    scopus 로고
    • Involvement of cathepsin B in the motor neuron degeneration of amyotrophic lateral sclerosis
    • Kikuchi H, Yamada T, Furuya H, Doh-Ura K, Ohyagi Y, Iwaki T, et al. Involvement of cathepsin B in the motor neuron degeneration of amyotrophic lateral sclerosis. Acta Neuropathol (2003) 105:462-8. doi:10.1007/s00401-002-0667-9
    • (2003) Acta Neuropathol , vol.105 , pp. 462-468
    • Kikuchi, H.1    Yamada, T.2    Furuya, H.3    Doh-Ura, K.4    Ohyagi, Y.5    Iwaki, T.6
  • 129
    • 0025195944 scopus 로고
    • Enzymatically active lysosomal proteases are associated with amyloid deposits in Alzheimer brain
    • Cataldo AM, Nixon RA. Enzymatically active lysosomal proteases are associated with amyloid deposits in Alzheimer brain. Proc Natl Acad Sci U S A (1990) 87:3861-5. doi:10.1073/pnas.87.10.3861
    • (1990) Proc Natl Acad Sci U S A , vol.87 , pp. 3861-3865
    • Cataldo, A.M.1    Nixon, R.A.2
  • 131
    • 20944443414 scopus 로고    scopus 로고
    • Quantitative proteomics of cerebrospinal fluid from patients with Alzheimer disease
    • Zhang J, Goodlett DR, Quinn JF, Peskind E, Kaye JA, Zhou Y, et al. Quantitative proteomics of cerebrospinal fluid from patients with Alzheimer disease. J Alzheimers Dis (2005) 7:125-33.
    • (2005) J Alzheimers Dis , vol.7 , pp. 125-133
    • Zhang, J.1    Goodlett, D.R.2    Quinn, J.F.3    Peskind, E.4    Kaye, J.A.5    Zhou, Y.6
  • 132
    • 84894283814 scopus 로고    scopus 로고
    • Lysosomal network proteins as potential novel CSF biomarkers for Alzheimer's disease
    • Armstrong A, Mattsson N, Appelqvist H, Janefjord C, Sandin L, Agholme L, et al. Lysosomal network proteins as potential novel CSF biomarkers for Alzheimer's disease. Neuromolecular Med (2014) 16:150-60. doi:10.1007/s12017-013-8269-3
    • (2014) Neuromolecular Med , vol.16 , pp. 150-160
    • Armstrong, A.1    Mattsson, N.2    Appelqvist, H.3    Janefjord, C.4    Sandin, L.5    Agholme, L.6
  • 133
    • 0025987727 scopus 로고
    • Cathepsin B in synovial cells at the site of joint destruction in rheumatoid arthritis
    • Trabandt A, Gay RE, Fassbender HG, Gay S. Cathepsin B in synovial cells at the site of joint destruction in rheumatoid arthritis. Arthritis Rheum (1991) 34:1444-51. doi:10.1002/art.1780341116
    • (1991) Arthritis Rheum , vol.34 , pp. 1444-1451
    • Trabandt, A.1    Gay, R.E.2    Fassbender, H.G.3    Gay, S.4
  • 134
    • 0028901908 scopus 로고
    • Cathepsin B in osteoarthritis: cytochemical and histochemical analysis of human femoral head cartilage
    • Baici A, Lang A, Horler D, Kissling R, Merlin C. Cathepsin B in osteoarthritis: cytochemical and histochemical analysis of human femoral head cartilage. Ann Rheum Dis (1995) 54:289-97. doi:10.1136/ard.54.4.281
    • (1995) Ann Rheum Dis , vol.54 , pp. 289-297
    • Baici, A.1    Lang, A.2    Horler, D.3    Kissling, R.4    Merlin, C.5
  • 135
    • 33748710916 scopus 로고    scopus 로고
    • Cathepsins B, L and D in inflammatory bowel disease macrophages and potential therapeutic effects of cathepsin inhibition in vivo
    • Menzel K, Hausmann M, Obermeier F, Schreiter K, Dunger N, Bataille F, et al. Cathepsins B, L and D in inflammatory bowel disease macrophages and potential therapeutic effects of cathepsin inhibition in vivo. Clin Exp Immunol (2006) 146:169-80. doi:10.1111/j.1365-2249.2006.03188.x
    • (2006) Clin Exp Immunol , vol.146 , pp. 169-180
    • Menzel, K.1    Hausmann, M.2    Obermeier, F.3    Schreiter, K.4    Dunger, N.5    Bataille, F.6
  • 136
    • 0034930528 scopus 로고    scopus 로고
    • Towards specific functions of lysosomal cysteine peptidases: phenotypes of mice deficient for cathepsin B or cathepsin L
    • Reinheckel T, Deussing J, Roth W, Peters C. Towards specific functions of lysosomal cysteine peptidases: phenotypes of mice deficient for cathepsin B or cathepsin L. Biol Chem (2001) 382:735-41. doi:10.1515/BC.2001.089
    • (2001) Biol Chem , vol.382 , pp. 735-741
    • Reinheckel, T.1    Deussing, J.2    Roth, W.3    Peters, C.4
  • 138
    • 0043011297 scopus 로고    scopus 로고
    • Cathepsin B but not cathepsins L or S contributes to the pathogenesis of Unverricht-Lundborg progressive myoclonus epilepsy (EPM1)
    • Houseweart MK, Pennacchio LA, Vilaythong A, Peters C, Noebels JL, Myers RM. Cathepsin B but not cathepsins L or S contributes to the pathogenesis of Unverricht-Lundborg progressive myoclonus epilepsy (EPM1). J Neurobiol (2003) 56:315-27. doi:10.1002/neu.10253
    • (2003) J Neurobiol , vol.56 , pp. 315-327
    • Houseweart, M.K.1    Pennacchio, L.A.2    Vilaythong, A.3    Peters, C.4    Noebels, J.L.5    Myers, R.M.6
  • 139
    • 67649656100 scopus 로고    scopus 로고
    • Genetic cathepsin B deficiency reduces beta-amyloid in transgenic mice expressing human wild-type amyloid precursor protein
    • Hook VY, Kindy M, Reinheckel T, Peters C, Hook G. Genetic cathepsin B deficiency reduces beta-amyloid in transgenic mice expressing human wild-type amyloid precursor protein. Biochem Biophys Res Commun (2009) 386:284-8. doi:10.1016/j.bbrc.2009.05.131
    • (2009) Biochem Biophys Res Commun , vol.386 , pp. 284-288
    • Hook, V.Y.1    Kindy, M.2    Reinheckel, T.3    Peters, C.4    Hook, G.5
  • 140
    • 84860156823 scopus 로고    scopus 로고
    • Deletion of the cathepsin B gene improves memory deficits in a transgenic alzheimer's disease mouse model expressing AbetaPP containing the wild-type beta-secretase site sequence
    • Kindy MS, Yu J, Zhu H, El-Amouri SS, Hook V, Hook GR. Deletion of the cathepsin B gene improves memory deficits in a transgenic alzheimer's disease mouse model expressing AbetaPP containing the wild-type beta-secretase site sequence. J Alzheimers Dis (2012) 29:827-40. doi:10.3233/JAD-2012-111604
    • (2012) J Alzheimers Dis , vol.29 , pp. 827-840
    • Kindy, M.S.1    Yu, J.2    Zhu, H.3    El-Amouri, S.S.4    Hook, V.5    Hook, G.R.6
  • 141
    • 84902261433 scopus 로고    scopus 로고
    • Brain pyroglutamate amyloid-beta is produced by cathepsin B and is reduced by the cysteine protease inhibitor E64d, representing a potential Alzheimer's disease therapeutic
    • Hook G, Yu J, Toneff T, Kindy M, Hook V. Brain pyroglutamate amyloid-beta is produced by cathepsin B and is reduced by the cysteine protease inhibitor E64d, representing a potential Alzheimer's disease therapeutic. J Alzheimers Dis (2014) 41:129-49. doi:10.3233/JAD-131370
    • (2014) J Alzheimers Dis , vol.41 , pp. 129-149
    • Hook, G.1    Yu, J.2    Toneff, T.3    Kindy, M.4    Hook, V.5
  • 142
    • 33748524564 scopus 로고    scopus 로고
    • Antiamyloidogenic and neuroprotective functions of cathepsin B: implications for Alzheimer's disease
    • Mueller-Steiner S, Zhou Y, Arai H, Roberson ED, Sun B, Chen J, et al. Antiamyloidogenic and neuroprotective functions of cathepsin B: implications for Alzheimer's disease. Neuron (2006) 51:703-14. doi:10.1016/j.neuron.2006.07.027
    • (2006) Neuron , vol.51 , pp. 703-714
    • Mueller-Steiner, S.1    Zhou, Y.2    Arai, H.3    Roberson, E.D.4    Sun, B.5    Chen, J.6
  • 143
    • 84884163207 scopus 로고    scopus 로고
    • Differential pathways for interleukin-1beta production activated by chromogranin A and amyloid beta in microglia
    • Wu Z, Sun L, Hashioka S, Yu S, Schwab C, Okada R, et al. Differential pathways for interleukin-1beta production activated by chromogranin A and amyloid beta in microglia. Neurobiol Aging (2013) 34:2715-25. doi:10.1016/j.neurobiolaging.2013.05.018
    • (2013) Neurobiol Aging , vol.34 , pp. 2715-2725
    • Wu, Z.1    Sun, L.2    Hashioka, S.3    Yu, S.4    Schwab, C.5    Okada, R.6
  • 144
    • 47949098831 scopus 로고    scopus 로고
    • Cathepsin B is involved in the trafficking of TNF-alpha-containing vesicles to the plasma membrane in macrophages
    • Ha SD, Martins A, Khazaie K, Han J, Chan BM, Kim SO. Cathepsin B is involved in the trafficking of TNF-alpha-containing vesicles to the plasma membrane in macrophages. J Immunol (2008) 181:690-7. doi:10.4049/jimmunol.181.1.690
    • (2008) J Immunol , vol.181 , pp. 690-697
    • Ha, S.D.1    Martins, A.2    Khazaie, K.3    Han, J.4    Chan, B.M.5    Kim, S.O.6
  • 145
    • 22344435528 scopus 로고    scopus 로고
    • Bid is upstream of lysosome-mediated caspase 2 activation in tumor necrosis factor alpha-induced hepatocyte apoptosis
    • Guicciardi ME, Bronk SF, Werneburg NW, Yin XM, Gores GJ. Bid is upstream of lysosome-mediated caspase 2 activation in tumor necrosis factor alpha-induced hepatocyte apoptosis. Gastroenterology (2005) 129:269-84. doi:10.1053/j.gastro.2005.05.022
    • (2005) Gastroenterology , vol.129 , pp. 269-284
    • Guicciardi, M.E.1    Bronk, S.F.2    Werneburg, N.W.3    Yin, X.M.4    Gores, G.J.5
  • 146
    • 84936991055 scopus 로고    scopus 로고
    • Redundancy between cysteine cathepsins in murine experimental autoimmune encephalomyelitis
    • Allan ER, Yates RM. Redundancy between cysteine cathepsins in murine experimental autoimmune encephalomyelitis. PLoS One (2015) 10:e0128945. doi:10.1371/journal.pone.0128945
    • (2015) PLoS One , vol.10
    • Allan, E.R.1    Yates, R.M.2
  • 147
    • 80655144756 scopus 로고    scopus 로고
    • Pyroglutamate amyloid-beta (Abeta): a hatchet man in Alzheimer disease
    • Jawhar S, Wirths O, Bayer TA. Pyroglutamate amyloid-beta (Abeta): a hatchet man in Alzheimer disease. J Biol Chem (2011) 286:38825-32. doi:10.1074/jbc.R111.288308
    • (2011) J Biol Chem , vol.286 , pp. 38825-38832
    • Jawhar, S.1    Wirths, O.2    Bayer, T.A.3
  • 148
    • 26844559355 scopus 로고    scopus 로고
    • Inhibition of cathepsin B reduces β-amyloid production in regulated secretory vesicles of neuronal chromaffin cells: evidence for cathepsin B as a candidate β-secretase of Alzheimer's disease
    • Hook V, Toneff T, Bogyo M, Medzihradszky KF, Nevenu J, Lane W, et al. Inhibition of cathepsin B reduces β-amyloid production in regulated secretory vesicles of neuronal chromaffin cells: evidence for cathepsin B as a candidate β-secretase of Alzheimer's disease. Biol Chem (2005) 386:931-40. doi:10.1515/BC.2005.108
    • (2005) Biol Chem , vol.386 , pp. 931-940
    • Hook, V.1    Toneff, T.2    Bogyo, M.3    Medzihradszky, K.F.4    Nevenu, J.5    Lane, W.6
  • 149
    • 33846621573 scopus 로고    scopus 로고
    • Cysteine protease inhibitors effectively reduce in vivo levels of brain beta-amyloid related to Alzheimer's disease
    • Hook V, Kindy M, Hook G. Cysteine protease inhibitors effectively reduce in vivo levels of brain beta-amyloid related to Alzheimer's disease. Biol Chem (2007) 388:247-52. doi:10.1515/BC.2007.027
    • (2007) Biol Chem , vol.388 , pp. 247-252
    • Hook, V.1    Kindy, M.2    Hook, G.3
  • 150
    • 34547901415 scopus 로고    scopus 로고
    • Cysteine protease inhibitors reduce brain beta-amyloid and beta-secretase activity in vivo and are potential Alzheimer's disease therapeutics
    • Hook G, Hook VY, Kindy M. Cysteine protease inhibitors reduce brain beta-amyloid and beta-secretase activity in vivo and are potential Alzheimer's disease therapeutics. Biol Chem (2007) 388:979-83. doi:10.1515/BC.2007.027
    • (2007) Biol Chem , vol.388 , pp. 979-983
    • Hook, G.1    Hook, V.Y.2    Kindy, M.3
  • 151
    • 43149100823 scopus 로고    scopus 로고
    • Inhibitors of cathepsin B improve memory and reduce Abeta in transgenic Alzheimer's Disease mice expressing the wild-type, but not the Swedish mutant, beta-secretase APP site
    • Hook VY, Kindy M, Hook G. Inhibitors of cathepsin B improve memory and reduce Abeta in transgenic Alzheimer's Disease mice expressing the wild-type, but not the Swedish mutant, beta-secretase APP site. J Biol Chem (2008) 283:7745-53. doi:10.1074/jbc.M708362200
    • (2008) J Biol Chem , vol.283 , pp. 7745-7753
    • Hook, V.Y.1    Kindy, M.2    Hook, G.3
  • 152
    • 0028174629 scopus 로고
    • Beta amyloid protein deposition in the brain after severe head injury: implications for the pathogenesis of Alzheimer's disease
    • Roberts GW, Gentleman SM, Lynch A, Murray L, Landon M, Graham DI. Beta amyloid protein deposition in the brain after severe head injury: implications for the pathogenesis of Alzheimer's disease. J Neurol Neurosurg Psychiatry (1994) 57:419-25. doi:10.1136/jnnp.57.4.419
    • (1994) J Neurol Neurosurg Psychiatry , vol.57 , pp. 419-425
    • Roberts, G.W.1    Gentleman, S.M.2    Lynch, A.3    Murray, L.4    Landon, M.5    Graham, D.I.6
  • 153
    • 61649120633 scopus 로고    scopus 로고
    • A lack of amyloid beta plaques despite persistent accumulation of amyloid beta in axons of long-term survivors of traumatic brain injury
    • Chen XH, Johnson VE, Uryu K, Trojanowski JQ, Smith DH. A lack of amyloid beta plaques despite persistent accumulation of amyloid beta in axons of long-term survivors of traumatic brain injury. Brain Pathol (2009) 19:214-23. doi:10.1111/j.1750-3639.2008.00176.x
    • (2009) Brain Pathol , vol.19 , pp. 214-223
    • Chen, X.H.1    Johnson, V.E.2    Uryu, K.3    Trojanowski, J.Q.4    Smith, D.H.5
  • 155
    • 0036326050 scopus 로고    scopus 로고
    • Elevated intracranial IL-18 in humans and mice after traumatic brain injury and evidence of neuroprotective effects of IL-18-binding protein after experimental closed head injury
    • Yatsiv I, Morganti-Kossmann MC, Perez D, Dinarello CA, Novick D, Rubinstein M, et al. Elevated intracranial IL-18 in humans and mice after traumatic brain injury and evidence of neuroprotective effects of IL-18-binding protein after experimental closed head injury. J Cereb Blood Flow Metab (2002) 22:971-8. doi:10.1097/00004647-200208000-00008
    • (2002) J Cereb Blood Flow Metab , vol.22 , pp. 971-978
    • Yatsiv, I.1    Morganti-Kossmann, M.C.2    Perez, D.3    Dinarello, C.A.4    Novick, D.5    Rubinstein, M.6
  • 156
    • 33646083015 scopus 로고    scopus 로고
    • The effect of traumatic brain injury upon the concentration and expression of interleukin-1beta and interleukin-10 in the rat
    • Kamm K, Vanderkolk W, Lawrence C, Jonker M, Davis AT. The effect of traumatic brain injury upon the concentration and expression of interleukin-1beta and interleukin-10 in the rat. J Trauma (2006) 60:152-7. doi:10.1097/01.ta.0000196345.81169.a1
    • (2006) J Trauma , vol.60 , pp. 152-157
    • Kamm, K.1    Vanderkolk, W.2    Lawrence, C.3    Jonker, M.4    Davis, A.T.5
  • 157
    • 84896702066 scopus 로고    scopus 로고
    • Activation and regulation of cellular inflammasomes: gaps in our knowledge for central nervous system injury
    • de Rivero Vaccari JP, Dietrich WD, Keane RW. Activation and regulation of cellular inflammasomes: gaps in our knowledge for central nervous system injury. J Cereb Blood Flow Metab (2014) 34:369-75. doi:10.1038/jcbfm.2013.227
    • (2014) J Cereb Blood Flow Metab , vol.34 , pp. 369-375
    • de Rivero Vaccari, J.P.1    Dietrich, W.D.2    Keane, R.W.3
  • 158
    • 47849085872 scopus 로고    scopus 로고
    • The NALP3 inflammasome is involved in the innate immune response to amyloid-beta
    • Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, et al. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat Immunol (2008) 9:857-65. doi:10.1038/ni.1636
    • (2008) Nat Immunol , vol.9 , pp. 857-865
    • Halle, A.1    Hornung, V.2    Petzold, G.C.3    Stewart, C.R.4    Monks, B.G.5    Reinheckel, T.6
  • 159
    • 0035164416 scopus 로고    scopus 로고
    • A lysosomal protease enters the death scene
    • Salvesen GS. A lysosomal protease enters the death scene. J Clin Invest (2001) 107:21-2. doi:10.1172/JCI11829
    • (2001) J Clin Invest , vol.107 , pp. 21-22
    • Salvesen, G.S.1
  • 161
    • 0033756474 scopus 로고    scopus 로고
    • Cathepsin B contributes to TNF-alpha-mediated hepatocyte apoptosis by promoting mitochondrial release of cytochrome c
    • Guicciardi ME, Deussing J, Miyoshi H, Bronk SF, Svingen PA, Peters C, et al. Cathepsin B contributes to TNF-alpha-mediated hepatocyte apoptosis by promoting mitochondrial release of cytochrome c. J Clin Invest (2000) 106:1127-37. doi:10.1172/JCI9914
    • (2000) J Clin Invest , vol.106 , pp. 1127-1137
    • Guicciardi, M.E.1    Deussing, J.2    Miyoshi, H.3    Bronk, S.F.4    Svingen, P.A.5    Peters, C.6
  • 163
    • 33746255613 scopus 로고    scopus 로고
    • Human cathepsin L rescues the neurodegeneration and lethality in cathepsin B/L double-deficient mice
    • Sevenich L, Pennacchio LA, Peters C, Reinheckel T. Human cathepsin L rescues the neurodegeneration and lethality in cathepsin B/L double-deficient mice. Biol Chem (2006) 387:885-91. doi:10.1515/BC.2006.112
    • (2006) Biol Chem , vol.387 , pp. 885-891
    • Sevenich, L.1    Pennacchio, L.A.2    Peters, C.3    Reinheckel, T.4
  • 164
    • 0021882035 scopus 로고
    • The distribution of cathepsin B in human tissues
    • Howie AJ, Burnett D, Crocker J. The distribution of cathepsin B in human tissues. J Pathol (1985) 145:307-14. doi:10.1002/path.1711450404
    • (1985) J Pathol , vol.145 , pp. 307-314
    • Howie, A.J.1    Burnett, D.2    Crocker, J.3
  • 165
    • 0021875579 scopus 로고
    • Distribution of cathepsins B and H in rat tissues and peripheral blood cells
    • Kominami E, Tsukahara T, Bando Y, Katunuma N. Distribution of cathepsins B and H in rat tissues and peripheral blood cells. J Biochem (1985) 98:87-93.
    • (1985) J Biochem , vol.98 , pp. 87-93
    • Kominami, E.1    Tsukahara, T.2    Bando, Y.3    Katunuma, N.4
  • 166
    • 0022532266 scopus 로고
    • Differences in cathepsin B mRNA levels in rat tissues suggest specialized functions
    • San Segundo B, Chan SJ, Steiner DF. Differences in cathepsin B mRNA levels in rat tissues suggest specialized functions. FEBS Lett (1986) 201:251-6. doi:10.1016/0014-5793(86)80618-4
    • (1986) FEBS Lett , vol.201 , pp. 251-256
    • San Segundo, B.1    Chan, S.J.2    Steiner, D.F.3
  • 170
    • 0025775712 scopus 로고
    • Abnormal distribution of cathepsins in the brain of patients with Alzheimer's disease
    • Nakamura Y, Takeda M, Suzuki H, Hattori H, Tada K, Hariguchi S, et al. Abnormal distribution of cathepsins in the brain of patients with Alzheimer's disease. Neurosci Lett (1991) 130:195-8. doi:10.1016/0304-3940(91)90395-A
    • (1991) Neurosci Lett , vol.130 , pp. 195-198
    • Nakamura, Y.1    Takeda, M.2    Suzuki, H.3    Hattori, H.4    Tada, K.5    Hariguchi, S.6
  • 171
    • 0027378622 scopus 로고
    • Abnormal distribution of cathepsin proteinases and endogenous inhibitors (cystatins) in the hippocampus of patients with Alzheimer's disease, parkinsonism-dementia complex on Guam, and senile dementia and in the aged
    • Ii K, Ito H, Kominami E, Hirano A. Abnormal distribution of cathepsin proteinases and endogenous inhibitors (cystatins) in the hippocampus of patients with Alzheimer's disease, parkinsonism-dementia complex on Guam, and senile dementia and in the aged. Virchows Arch A Pathol Anat Histopathol (1993) 423:185-94. doi:10.1007/BF01614769
    • (1993) Virchows Arch A Pathol Anat Histopathol , vol.423 , pp. 185-194
    • Ii, K.1    Ito, H.2    Kominami, E.3    Hirano, A.4
  • 172
    • 0024412132 scopus 로고
    • Cathepsin B immunoreactive neurons in rat brain. A combined light and electron microscopic study
    • Bernstein HG, Sormunen R, Jarvinen M, Kloss P, Kirschke H, Rinne A. Cathepsin B immunoreactive neurons in rat brain. A combined light and electron microscopic study. J Hirnforsch (1989) 30:313-7.
    • (1989) J Hirnforsch , vol.30 , pp. 313-317
    • Bernstein, H.G.1    Sormunen, R.2    Jarvinen, M.3    Kloss, P.4    Kirschke, H.5    Rinne, A.6
  • 173
    • 0032852272 scopus 로고    scopus 로고
    • Age-related changes in ultrastructural features of cathepsin B-and D-containing neurons in rat cerebral cortex
    • Jung H, Lee EY, Lee SI. Age-related changes in ultrastructural features of cathepsin B-and D-containing neurons in rat cerebral cortex. Brain Res (1999) 844:43-54. doi:10.1016/S0006-8993(99)01888-0
    • (1999) Brain Res , vol.844 , pp. 43-54
    • Jung, H.1    Lee, E.Y.2    Lee, S.I.3
  • 174
    • 0028206592 scopus 로고
    • Differential distribution of messenger RNAs for cathepsins B, L and S in adult rat brain: an in situ hybridization study
    • Petanceska S, Burke S, Watson SJ, Devi L. Differential distribution of messenger RNAs for cathepsins B, L and S in adult rat brain: an in situ hybridization study. Neuroscience (1994) 59:729-38. doi:10.1016/0306-4522(94)90190-2
    • (1994) Neuroscience , vol.59 , pp. 729-738
    • Petanceska, S.1    Burke, S.2    Watson, S.J.3    Devi, L.4
  • 175
    • 84866738362 scopus 로고    scopus 로고
    • Cerebrospinal fluid cathepsin B and S
    • Nilsson E, Bodolea C, Gordh T, Larsson A. Cerebrospinal fluid cathepsin B and S. Neurol Sci (2013) 34:445-8. doi:10.1007/s10072-012-1022-0
    • (2013) Neurol Sci , vol.34 , pp. 445-448
    • Nilsson, E.1    Bodolea, C.2    Gordh, T.3    Larsson, A.4
  • 176
    • 0028323067 scopus 로고
    • Age-related changes in activities and localizations of cathepsins D, E, B, and L in the rat brain tissues
    • Nakanishi H, Tominaga K, Amano T, Hirotsu I, Inoue T, Yamamoto K. Age-related changes in activities and localizations of cathepsins D, E, B, and L in the rat brain tissues. Exp Neurol (1994) 126:119-28. doi:10.1006/exnr.1994.1048
    • (1994) Exp Neurol , vol.126 , pp. 119-128
    • Nakanishi, H.1    Tominaga, K.2    Amano, T.3    Hirotsu, I.4    Inoue, T.5    Yamamoto, K.6
  • 177
    • 0021040946 scopus 로고
    • Lysosomes revisited
    • de Duve C. Lysosomes revisited. Eur J Biochem (1983) 137:391-7. doi:10.1111/j.1432-1033.1983.tb07841.x
    • (1983) Eur J Biochem , vol.137 , pp. 391-397
    • de Duve, C.1
  • 178
    • 0023858711 scopus 로고
    • Regulation of microautophagy and basal protein turnover in rat liver. Effects of short-term starvation
    • Mortimore GE, Lardeux BR, Adams CE. Regulation of microautophagy and basal protein turnover in rat liver. Effects of short-term starvation. J Biol Chem (1988) 263:2506-12.
    • (1988) J Biol Chem , vol.263 , pp. 2506-2512
    • Mortimore, G.E.1    Lardeux, B.R.2    Adams, C.E.3
  • 179
    • 0026555037 scopus 로고
    • Proteases and proteolysis in the lysosome
    • Bohley P, Seglen PO. Proteases and proteolysis in the lysosome. Experientia (1992) 48:151-7. doi:10.1007/BF01923508
    • (1992) Experientia , vol.48 , pp. 151-157
    • Bohley, P.1    Seglen, P.O.2
  • 181
    • 0028171897 scopus 로고
    • Engineering the S2 subsite specificity of human cathepsin S to a cathepsin L-and cathepsin B-like specificity
    • Bromme D, Bonneau PR, Lachance P, Storer AC. Engineering the S2 subsite specificity of human cathepsin S to a cathepsin L-and cathepsin B-like specificity. J Biol Chem (1994) 269:30238-42.
    • (1994) J Biol Chem , vol.269 , pp. 30238-30242
    • Bromme, D.1    Bonneau, P.R.2    Lachance, P.3    Storer, A.C.4
  • 183
    • 34547562467 scopus 로고    scopus 로고
    • Regulated ATP release from astrocytes through lysosome exocytosis
    • Zhang Z, Chen G, Zhou W, Song A, Xu T, Luo Q, et al. Regulated ATP release from astrocytes through lysosome exocytosis. Nat Cell Biol (2007) 9:945-53. doi:10.1038/ncb1620
    • (2007) Nat Cell Biol , vol.9 , pp. 945-953
    • Zhang, Z.1    Chen, G.2    Zhou, W.3    Song, A.4    Xu, T.5    Luo, Q.6
  • 184
    • 84861354544 scopus 로고    scopus 로고
    • TI-VAMP/VAMP7 is the SNARE of secretory lysosomes contributing to ATP secretion from astrocytes
    • Verderio C, Cagnoli C, Bergami M, Francolini M, Schenk U, Colombo A, et al. TI-VAMP/VAMP7 is the SNARE of secretory lysosomes contributing to ATP secretion from astrocytes. Biol Cell (2012) 104:213-28. doi:10.1111/boc.201100070
    • (2012) Biol Cell , vol.104 , pp. 213-228
    • Verderio, C.1    Cagnoli, C.2    Bergami, M.3    Francolini, M.4    Schenk, U.5    Colombo, A.6
  • 185
    • 0030764160 scopus 로고    scopus 로고
    • Identification and characterization of cathepsin B as the cellular MARCKS cleaving enzyme
    • Spizz G, Blackshear PJ. Identification and characterization of cathepsin B as the cellular MARCKS cleaving enzyme. J Biol Chem (1997) 272:23833-42. doi:10.1074/jbc.272.38.23833
    • (1997) J Biol Chem , vol.272 , pp. 23833-23842
    • Spizz, G.1    Blackshear, P.J.2
  • 186
    • 5344224076 scopus 로고    scopus 로고
    • Cathepsin B-like proteolysis and MARCKS degradation in sub-lethal NMDA-induced collapse of dendritic spines
    • Graber S, Maiti S, Halpain S. Cathepsin B-like proteolysis and MARCKS degradation in sub-lethal NMDA-induced collapse of dendritic spines. Neuropharmacology (2004) 47:706-13. doi:10.1016/j.neuropharm.2004.08.004
    • (2004) Neuropharmacology , vol.47 , pp. 706-713
    • Graber, S.1    Maiti, S.2    Halpain, S.3
  • 187
    • 34547497862 scopus 로고    scopus 로고
    • Cleavage of the myristoylated alanine-rich C kinase substrate (MARCKS) by cysteine cathepsins in cells and tissues of stefin B-deficient mice
    • Kopitar-Jerala N, Turk B. Cleavage of the myristoylated alanine-rich C kinase substrate (MARCKS) by cysteine cathepsins in cells and tissues of stefin B-deficient mice. Biol Chem (2007) 388:847-52. doi:10.1515/BC.2007.092
    • (2007) Biol Chem , vol.388 , pp. 847-852
    • Kopitar-Jerala, N.1    Turk, B.2
  • 188
    • 0037115483 scopus 로고    scopus 로고
    • Trafficking of lysosomal cathepsin B-green fluorescent protein to the surface of thyroid epithelial cells involves the endosomal/lysosomal compartment
    • Linke M, Herzog V, Brix K. Trafficking of lysosomal cathepsin B-green fluorescent protein to the surface of thyroid epithelial cells involves the endosomal/lysosomal compartment. J Cell Sci (2002) 115:4877-89. doi:10.1242/jcs.00184
    • (2002) J Cell Sci , vol.115 , pp. 4877-4889
    • Linke, M.1    Herzog, V.2    Brix, K.3
  • 189
    • 79551550809 scopus 로고    scopus 로고
    • 14-3-3zeta binds class III phosphatidylinositol-3-kinase and inhibits autophagy
    • Pozuelo-Rubio M. 14-3-3zeta binds class III phosphatidylinositol-3-kinase and inhibits autophagy. Autophagy (2011) 7:240-2. doi:10.4161/auto.7.2.14286
    • (2011) Autophagy , vol.7 , pp. 240-242
    • Pozuelo-Rubio, M.1
  • 190
    • 84936776635 scopus 로고    scopus 로고
    • Proteolysis mediated by cysteine cathepsins and legumain-recent advances and cell biological challenges
    • Brix K, Mcinnes J, Al-Hashimi A, Rehders M, Tamhane T, Haugen MH. Proteolysis mediated by cysteine cathepsins and legumain-recent advances and cell biological challenges. Protoplasma (2015) 252:755-74. doi:10.1007/s00709-014-0730-0
    • (2015) Protoplasma , vol.252 , pp. 755-774
    • Brix, K.1    Mcinnes, J.2    Al-Hashimi, A.3    Rehders, M.4    Tamhane, T.5    Haugen, M.H.6
  • 191
    • 0022402201 scopus 로고
    • Pathways of protein secretion in eukaryotes
    • Kelly RB. Pathways of protein secretion in eukaryotes. Science (1985) 230:25-32. doi:10.1126/science.2994224
    • (1985) Science , vol.230 , pp. 25-32
    • Kelly, R.B.1
  • 192
    • 0037077262 scopus 로고    scopus 로고
    • Presence of cathepsin B in the human pancreatic secretory pathway and its role in trypsinogen activation during hereditary pancreatitis
    • Kukor Z, Mayerle J, Kruger B, Toth M, Steed PM, Halangk W, et al. Presence of cathepsin B in the human pancreatic secretory pathway and its role in trypsinogen activation during hereditary pancreatitis. J Biol Chem (2002) 277:21389-96. doi:10.1074/jbc.M200878200
    • (2002) J Biol Chem , vol.277 , pp. 21389-21396
    • Kukor, Z.1    Mayerle, J.2    Kruger, B.3    Toth, M.4    Steed, P.M.5    Halangk, W.6
  • 193
    • 0033795691 scopus 로고    scopus 로고
    • Role of cathepsin B in intracellular trypsinogen activation and the onset of acute pancreatitis
    • Halangk W, Lerch MM, Brandt-Nedelev B, Roth W, Ruthenbuerger M, Reinheckel T, et al. Role of cathepsin B in intracellular trypsinogen activation and the onset of acute pancreatitis. J Clin Invest (2000) 106:773-81. doi:10.1172/JCI9411
    • (2000) J Clin Invest , vol.106 , pp. 773-781
    • Halangk, W.1    Lerch, M.M.2    Brandt-Nedelev, B.3    Roth, W.4    Ruthenbuerger, M.5    Reinheckel, T.6
  • 194
    • 0030943904 scopus 로고    scopus 로고
    • Differential sorting of lysosomal enzymes out of the regulated secretory pathway in pancreatic beta-cells
    • Kuliawat R, Klumperman J, Ludwig T, Arvan P. Differential sorting of lysosomal enzymes out of the regulated secretory pathway in pancreatic beta-cells. J Cell Biol (1997) 137:595-608. doi:10.1083/jcb.137.3.595
    • (1997) J Cell Biol , vol.137 , pp. 595-608
    • Kuliawat, R.1    Klumperman, J.2    Ludwig, T.3    Arvan, P.4
  • 196
    • 0028566942 scopus 로고
    • Human cathepsin B is a metastable enzyme stabilized by specific ionic interactions associated with the active site
    • Turk B, Dolenc I, Zerovnik E, Turk D, Gubensek F, Turk V. Human cathepsin B is a metastable enzyme stabilized by specific ionic interactions associated with the active site. Biochemistry (1994) 33:14800-6. doi:10.1021/bi00253a019
    • (1994) Biochemistry , vol.33 , pp. 14800-14806
    • Turk, B.1    Dolenc, I.2    Zerovnik, E.3    Turk, D.4    Gubensek, F.5    Turk, V.6
  • 197
    • 70349163894 scopus 로고    scopus 로고
    • Direct measurement of cathepsin B activity in the cytosol of apoptotic cells by an activity-based probe
    • Pratt MR, Sekedat MD, Chiang KP, Muir TW. Direct measurement of cathepsin B activity in the cytosol of apoptotic cells by an activity-based probe. Chem Biol (2009) 16:1001-12. doi:10.1016/j.chembiol.2009.07.011
    • (2009) Chem Biol , vol.16 , pp. 1001-1012
    • Pratt, M.R.1    Sekedat, M.D.2    Chiang, K.P.3    Muir, T.W.4
  • 198
    • 0023665211 scopus 로고
    • Dissociation of ionizing groups in the binding cleft inversely controls the endo-and exopeptidase activities of cathepsin B
    • Polgar L, Csoma C. Dissociation of ionizing groups in the binding cleft inversely controls the endo-and exopeptidase activities of cathepsin B. J Biol Chem (1987) 262:14448-53.
    • (1987) J Biol Chem , vol.262 , pp. 14448-14453
    • Polgar, L.1    Csoma, C.2
  • 199
    • 0035847011 scopus 로고    scopus 로고
    • Cathepsin B activity regulation. Heparin-like glycosaminogylcans protect human cathepsin B from alkaline pH-induced inactivation
    • Almeida PC, Nantes IL, Chagas JR, Rizzi CC, Faljoni-Alario A, Carmona E, et al. Cathepsin B activity regulation. Heparin-like glycosaminogylcans protect human cathepsin B from alkaline pH-induced inactivation. J Biol Chem (2001) 276:944-51. doi:10.1074/jbc.M003820200
    • (2001) J Biol Chem , vol.276 , pp. 944-951
    • Almeida, P.C.1    Nantes, I.L.2    Chagas, J.R.3    Rizzi, C.C.4    Faljoni-Alario, A.5    Carmona, E.6
  • 200
    • 0027459566 scopus 로고
    • Kinetics of the pH-induced inactivation of human cathepsin L
    • Turk B, Dolenc I, Turk V, Bieth JG. Kinetics of the pH-induced inactivation of human cathepsin L. Biochemistry (1993) 32:375-80. doi:10.1021/bi00052a046
    • (1993) Biochemistry , vol.32 , pp. 375-380
    • Turk, B.1    Dolenc, I.2    Turk, V.3    Bieth, J.G.4
  • 201
    • 0035986219 scopus 로고    scopus 로고
    • Regulating cysteine protease activity: essential role of protease inhibitors as guardians and regulators
    • Turk B, Turk D, Salvesen GS. Regulating cysteine protease activity: essential role of protease inhibitors as guardians and regulators. Curr Pharm Des (2002) 8:1623-37. doi:10.2174/1381612023394124
    • (2002) Curr Pharm Des , vol.8 , pp. 1623-1637
    • Turk, B.1    Turk, D.2    Salvesen, G.S.3
  • 202
    • 0023024774 scopus 로고
    • Isolation of six cysteine proteinase inhibitors from human urine. Their physicochemical and enzyme kinetic properties and concentrations in biological fluids
    • Abrahamson M, Barrett AJ, Salvesen G, Grubb A. Isolation of six cysteine proteinase inhibitors from human urine. Their physicochemical and enzyme kinetic properties and concentrations in biological fluids. J Biol Chem (1986) 261:11282-9.
    • (1986) J Biol Chem , vol.261 , pp. 11282-11289
    • Abrahamson, M.1    Barrett, A.J.2    Salvesen, G.3    Grubb, A.4
  • 203
    • 77957352489 scopus 로고    scopus 로고
    • Cystatin C levels are positively correlated with both Abeta42 and tau levels in cerebrospinal fluid in persons with Alzheimer's disease, mild cognitive impairment, and healthy controls
    • Sundelof J, Sundstrom J, Hansson O, Eriksdotter-Jonhagen M, Giedraitis V, Larsson A, et al. Cystatin C levels are positively correlated with both Abeta42 and tau levels in cerebrospinal fluid in persons with Alzheimer's disease, mild cognitive impairment, and healthy controls. J Alzheimers Dis (2010) 21:471-8. doi:10.3233/JAD-2010-091594
    • (2010) J Alzheimers Dis , vol.21 , pp. 471-478
    • Sundelof, J.1    Sundstrom, J.2    Hansson, O.3    Eriksdotter-Jonhagen, M.4    Giedraitis, V.5    Larsson, A.6
  • 205
    • 84867063815 scopus 로고    scopus 로고
    • Increased intracranial pressure after diffuse traumatic brain injury exacerbates neuronal somatic membrane poration but not axonal injury: evidence for primary intracranial pressure-induced neuronal perturbation
    • Lafrenaye AD, Mcginn MJ, Povlishock JT. Increased intracranial pressure after diffuse traumatic brain injury exacerbates neuronal somatic membrane poration but not axonal injury: evidence for primary intracranial pressure-induced neuronal perturbation. J Cereb Blood Flow Metab (2012) 32:1919-32. doi:10.1038/jcbfm.2012.95
    • (2012) J Cereb Blood Flow Metab , vol.32 , pp. 1919-1932
    • Lafrenaye, A.D.1    Mcginn, M.J.2    Povlishock, J.T.3
  • 206
    • 84876903470 scopus 로고    scopus 로고
    • Poloxamer 188 attenuates in vitro traumatic brain injury-induced mitochondrial and lysosomal membrane permeabilization damage in cultured primary neurons
    • Luo CL, Chen XP, Li LL, Li QQ, Li BX, Xue AM, et al. Poloxamer 188 attenuates in vitro traumatic brain injury-induced mitochondrial and lysosomal membrane permeabilization damage in cultured primary neurons. J Neurotrauma (2013) 30:597-607. doi:10.1089/neu.2012.2425
    • (2013) J Neurotrauma , vol.30 , pp. 597-607
    • Luo, C.L.1    Chen, X.P.2    Li, L.L.3    Li, Q.Q.4    Li, B.X.5    Xue, A.M.6
  • 207
    • 84871401547 scopus 로고    scopus 로고
    • Poloxamer-188 attenuates TBI-induced blood-brain barrier damage leading to decreased brain edema and reduced cellular death
    • Bao HJ, Wang T, Zhang MY, Liu R, Dai DK, Wang YQ, et al. Poloxamer-188 attenuates TBI-induced blood-brain barrier damage leading to decreased brain edema and reduced cellular death. Neurochem Res (2012) 37:2856-67. doi:10.1007/s11064-012-0880-4
    • (2012) Neurochem Res , vol.37 , pp. 2856-2867
    • Bao, H.J.1    Wang, T.2    Zhang, M.Y.3    Liu, R.4    Dai, D.K.5    Wang, Y.Q.6
  • 208
    • 84876177830 scopus 로고    scopus 로고
    • Poloxamer 188 protects neurons against ischemia/reperfusion injury through preserving integrity of cell membranes and blood brain barrier
    • Gu JH, Ge JB, Li M, Xu HD, Wu F, Qin ZH. Poloxamer 188 protects neurons against ischemia/reperfusion injury through preserving integrity of cell membranes and blood brain barrier. PLoS One (2013) 8:e61641. doi:10.1371/journal.pone.0061641
    • (2013) PLoS One , vol.8
    • Gu, J.H.1    Ge, J.B.2    Li, M.3    Xu, H.D.4    Wu, F.5    Qin, Z.H.6
  • 209
    • 73549105909 scopus 로고    scopus 로고
    • Increased activity and altered subcellular distribution of lysosomal enzymes determine neuronal vulnerability in Niemann-Pick type C1-deficient mice
    • Amritraj A, Peake K, Kodam A, Salio C, Merighi A, Vance JE, et al. Increased activity and altered subcellular distribution of lysosomal enzymes determine neuronal vulnerability in Niemann-Pick type C1-deficient mice. Am J Pathol (2009) 175:2540-56. doi:10.2353/ajpath.2009.081096
    • (2009) Am J Pathol , vol.175 , pp. 2540-2556
    • Amritraj, A.1    Peake, K.2    Kodam, A.3    Salio, C.4    Merighi, A.5    Vance, J.E.6
  • 210
    • 77952396366 scopus 로고    scopus 로고
    • Nuclear translocation of mitochondrial cytochrome c, lysosomal cathepsins B and D, and three other death-promoting proteins within the first 60 minutes of generalized seizures
    • Zhao S, Aviles ER Jr, Fujikawa DG. Nuclear translocation of mitochondrial cytochrome c, lysosomal cathepsins B and D, and three other death-promoting proteins within the first 60 minutes of generalized seizures. J Neurosci Res (2010) 88:1727-37. doi:10.1002/jnr.22338
    • (2010) J Neurosci Res , vol.88 , pp. 1727-1737
    • Zhao, S.1    Aviles, E.R.2    Fujikawa, D.G.3
  • 211
    • 84873123076 scopus 로고    scopus 로고
    • Triggering of inflammasome by aggregated alpha-synuclein, an inflammatory response in synucleinopathies
    • Codolo G, Plotegher N, Pozzobon T, Brucale M, Tessari I, Bubacco L, et al. Triggering of inflammasome by aggregated alpha-synuclein, an inflammatory response in synucleinopathies. PLoS One (2013) 8:e55375. doi:10.1371/journal.pone.0055375
    • (2013) PLoS One , vol.8
    • Codolo, G.1    Plotegher, N.2    Pozzobon, T.3    Brucale, M.4    Tessari, I.5    Bubacco, L.6
  • 212
    • 84876722731 scopus 로고    scopus 로고
    • Alpha-synuclein induces lysosomal rupture and cathepsin dependent reactive oxygen species following endocytosis
    • Freeman D, Cedillos R, Choyke S, Lukic Z, Mcguire K, Marvin S, et al. Alpha-synuclein induces lysosomal rupture and cathepsin dependent reactive oxygen species following endocytosis. PLoS One (2013) 8:e62143. doi:10.1371/journal.pone.0062143
    • (2013) PLoS One , vol.8
    • Freeman, D.1    Cedillos, R.2    Choyke, S.3    Lukic, Z.4    Mcguire, K.5    Marvin, S.6
  • 213
    • 0035110055 scopus 로고    scopus 로고
    • Lysosomal membrane damage in soluble Abeta-mediated cell death in Alzheimer's disease
    • Ditaranto K, Tekirian TL, Yang AJ. Lysosomal membrane damage in soluble Abeta-mediated cell death in Alzheimer's disease. Neurobiol Dis (2001) 8:19-31. doi:10.1006/nbdi.2000.0364
    • (2001) Neurobiol Dis , vol.8 , pp. 19-31
    • Ditaranto, K.1    Tekirian, T.L.2    Yang, A.J.3
  • 214
    • 84878643872 scopus 로고    scopus 로고
    • Lysosomal cell death at a glance
    • Aits S, Jaattela M. Lysosomal cell death at a glance. J Cell Sci (2013) 126:1905-12. doi:10.1242/jcs.091181
    • (2013) J Cell Sci , vol.126 , pp. 1905-1912
    • Aits, S.1    Jaattela, M.2
  • 215
    • 70450222302 scopus 로고    scopus 로고
    • The role of lysosomal rupture in neuronal death
    • Yamashima T, Oikawa S. The role of lysosomal rupture in neuronal death. Prog Neurobiol (2009) 89:343-58. doi:10.1016/j.pneurobio.2009.09.003
    • (2009) Prog Neurobiol , vol.89 , pp. 343-358
    • Yamashima, T.1    Oikawa, S.2
  • 216
    • 84878111480 scopus 로고    scopus 로고
    • Reconsider Alzheimer's disease by the 'calpain-cathepsin hypothesis'-a perspective review
    • Yamashima T. Reconsider Alzheimer's disease by the 'calpain-cathepsin hypothesis'-a perspective review. Prog Neurobiol (2013) 105:1-23. doi:10.1016/j.pneurobio.2013.02.004
    • (2013) Prog Neurobiol , vol.105 , pp. 1-23
    • Yamashima, T.1
  • 217
    • 84907504498 scopus 로고    scopus 로고
    • Heat shock protein 70.1 (Hsp70.1) affects neuronal cell fate by regulating lysosomal acid sphingomyelinase
    • Zhu H, Yoshimoto T, Yamashima T. Heat shock protein 70.1 (Hsp70.1) affects neuronal cell fate by regulating lysosomal acid sphingomyelinase. J Biol Chem (2014) 289:27432-43. doi:10.1074/jbc.M114.560334
    • (2014) J Biol Chem , vol.289 , pp. 27432-27443
    • Zhu, H.1    Yoshimoto, T.2    Yamashima, T.3
  • 218
  • 220
    • 18944407809 scopus 로고    scopus 로고
    • Enhanced oxidative stress in iNOS-deficient mice after traumatic brain injury: support for a neuroprotective role of iNOS
    • Bayir H, Kagan VE, Borisenko GG, Tyurina YY, Janesko KL, Vagni VA, et al. Enhanced oxidative stress in iNOS-deficient mice after traumatic brain injury: support for a neuroprotective role of iNOS. J Cereb Blood Flow Metab (2005) 25:673-84. doi:10.1038/sj.jcbfm.9600068
    • (2005) J Cereb Blood Flow Metab , vol.25 , pp. 673-684
    • Bayir, H.1    Kagan, V.E.2    Borisenko, G.G.3    Tyurina, Y.Y.4    Janesko, K.L.5    Vagni, V.A.6
  • 221
    • 41049114693 scopus 로고    scopus 로고
    • Lysosomes in iron metabolism, ageing and apoptosis
    • Kurz T, Terman A, Gustafsson B, Brunk UT. Lysosomes in iron metabolism, ageing and apoptosis. Histochem Cell Biol (2008) 129:389-406. doi:10.1007/s00418-008-0394-y
    • (2008) Histochem Cell Biol , vol.129 , pp. 389-406
    • Kurz, T.1    Terman, A.2    Gustafsson, B.3    Brunk, U.T.4
  • 222
    • 54949137644 scopus 로고    scopus 로고
    • Lysosomal membrane permeabilization in cell death
    • Boya P, Kroemer G. Lysosomal membrane permeabilization in cell death. Oncogene (2008) 27:6434-51. doi:10.1038/onc.2008.310
    • (2008) Oncogene , vol.27 , pp. 6434-6451
    • Boya, P.1    Kroemer, G.2
  • 223
    • 0033398499 scopus 로고    scopus 로고
    • Ischemic delayed neuronal death: role of the cysteine proteases calpain and cathepsins
    • Tontchev AB, Yamashima T. Ischemic delayed neuronal death: role of the cysteine proteases calpain and cathepsins. Neuropathology (1999) 19:356-65. doi:10.1046/j.1440-1789.1999.00259.x
    • (1999) Neuropathology , vol.19 , pp. 356-365
    • Tontchev, A.B.1    Yamashima, T.2
  • 224
    • 2442476459 scopus 로고    scopus 로고
    • Lysosomes in cell death
    • Guicciardi ME, Leist M, Gores GJ. Lysosomes in cell death. Oncogene (2004) 23:2881-90. doi:10.1038/sj.onc.1207512
    • (2004) Oncogene , vol.23 , pp. 2881-2890
    • Guicciardi, M.E.1    Leist, M.2    Gores, G.J.3
  • 226
    • 0034663829 scopus 로고    scopus 로고
    • tBID, a membrane-targeted death ligand, oligomerizes BAK to release cytochrome c
    • Wei MC, Lindsten T, Mootha VK, Weiler S, Gross A, Ashiya M, et al. tBID, a membrane-targeted death ligand, oligomerizes BAK to release cytochrome c. Genes Dev (2000) 14:2060-71.
    • (2000) Genes Dev , vol.14 , pp. 2060-2071
    • Wei, M.C.1    Lindsten, T.2    Mootha, V.K.3    Weiler, S.4    Gross, A.5    Ashiya, M.6
  • 227
    • 0242579153 scopus 로고    scopus 로고
    • Cathepsin-B-dependent apoptosis triggered by antithymocyte globulins: a novel mechanism of T-cell depletion
    • Michallet MC, Saltel F, Preville X, Flacher M, Revillard JP, Genestier L. Cathepsin-B-dependent apoptosis triggered by antithymocyte globulins: a novel mechanism of T-cell depletion. Blood (2003) 102:3719-26. doi:10.1182/blood-2003-04-1075
    • (2003) Blood , vol.102 , pp. 3719-3726
    • Michallet, M.C.1    Saltel, F.2    Preville, X.3    Flacher, M.4    Revillard, J.P.5    Genestier, L.6
  • 228
    • 0035096992 scopus 로고    scopus 로고
    • Microglial secreted cathepsin B induces neuronal apoptosis
    • Kingham PJ, Pocock JM. Microglial secreted cathepsin B induces neuronal apoptosis. J Neurochem (2001) 76:1475-84. doi:10.1046/j.1471-4159.2001.00146.x
    • (2001) J Neurochem , vol.76 , pp. 1475-1484
    • Kingham, P.J.1    Pocock, J.M.2
  • 229
    • 47849097202 scopus 로고    scopus 로고
    • Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization
    • Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol (2008) 9:847-56. doi:10.1038/ni.1631
    • (2008) Nat Immunol , vol.9 , pp. 847-856
    • Hornung, V.1    Bauernfeind, F.2    Halle, A.3    Samstad, E.O.4    Kono, H.5    Rock, K.L.6
  • 231
    • 0034192466 scopus 로고    scopus 로고
    • Dual role of caspase-11 in mediating activation of caspase-1 and caspase-3 under pathological conditions
    • Kang SJ, Wang S, Hara H, Peterson EP, Namura S, Amin-Hanjani S, et al. Dual role of caspase-11 in mediating activation of caspase-1 and caspase-3 under pathological conditions. J Cell Biol (2000) 149:613-22. doi:10.1083/jcb.149.3.613
    • (2000) J Cell Biol , vol.149 , pp. 613-622
    • Kang, S.J.1    Wang, S.2    Hara, H.3    Peterson, E.P.4    Namura, S.5    Amin-Hanjani, S.6
  • 232
    • 43949146665 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptors: "key" regulators of neuroinflammation after traumatic brain injury
    • Stahel PF, Smith WR, Bruchis J, Rabb CH. Peroxisome proliferator-activated receptors: "key" regulators of neuroinflammation after traumatic brain injury. PPAR Res (2008) 2008:538141. doi:10.1155/2008/538141
    • (2008) PPAR Res , vol.2008
    • Stahel, P.F.1    Smith, W.R.2    Bruchis, J.3    Rabb, C.H.4
  • 233
    • 27244452751 scopus 로고    scopus 로고
    • Fenofibrate, a peroxisome proliferator-activated receptor alpha agonist, exerts neuroprotective effects in traumatic brain injury
    • Besson VC, Chen XR, Plotkine M, Marchand-Verrecchia C. Fenofibrate, a peroxisome proliferator-activated receptor alpha agonist, exerts neuroprotective effects in traumatic brain injury. Neurosci Lett (2005) 388:7-12. doi:10.1016/j.neulet.2005.06.019
    • (2005) Neurosci Lett , vol.388 , pp. 7-12
    • Besson, V.C.1    Chen, X.R.2    Plotkine, M.3    Marchand-Verrecchia, C.4
  • 234
    • 34547118091 scopus 로고    scopus 로고
    • Neurological recovery-promoting, anti-inflammatory, and anti-oxidative effects afforded by fenofibrate, a PPAR alpha agonist, in traumatic brain injury
    • Chen XR, Besson VC, Palmier B, Garcia Y, Plotkine M, Marchand-Leroux C. Neurological recovery-promoting, anti-inflammatory, and anti-oxidative effects afforded by fenofibrate, a PPAR alpha agonist, in traumatic brain injury. J Neurotrauma (2007) 24:1119-31. doi:10.1089/neu.2006.0216
    • (2007) J Neurotrauma , vol.24 , pp. 1119-1131
    • Chen, X.R.1    Besson, V.C.2    Palmier, B.3    Garcia, Y.4    Plotkine, M.5    Marchand-Leroux, C.6
  • 235
    • 84871772296 scopus 로고    scopus 로고
    • Ligand activation of peroxisome proliferator-activated receptor delta suppresses cathepsin B expression in human endothelial cells in a posttranslational manner
    • Reichenbach G, Starzinski-Powitz A, Doll M, Hrgovic I, Valesky EM, Kippenberger S, et al. Ligand activation of peroxisome proliferator-activated receptor delta suppresses cathepsin B expression in human endothelial cells in a posttranslational manner. Exp Dermatol (2012) 21:751-7. doi:10.1111/exd.12002
    • (2012) Exp Dermatol , vol.21 , pp. 751-757
    • Reichenbach, G.1    Starzinski-Powitz, A.2    Doll, M.3    Hrgovic, I.4    Valesky, E.M.5    Kippenberger, S.6
  • 236
    • 0037370914 scopus 로고    scopus 로고
    • Axonal damage: a key predictor of outcome in human CNS diseases
    • Medana IM, Esiri MM. Axonal damage: a key predictor of outcome in human CNS diseases. Brain (2003) 126:515-30. doi:10.1093/brain/awg061
    • (2003) Brain , vol.126 , pp. 515-530
    • Medana, I.M.1    Esiri, M.M.2
  • 237
    • 25644436461 scopus 로고    scopus 로고
    • Essential role for the PKC target MARCKS in maintaining dendritic spine morphology
    • Calabrese B, Halpain S. Essential role for the PKC target MARCKS in maintaining dendritic spine morphology. Neuron (2005) 48:77-90. doi:10.1016/j.neuron.2005.08.027
    • (2005) Neuron , vol.48 , pp. 77-90
    • Calabrese, B.1    Halpain, S.2
  • 238
    • 39749103417 scopus 로고    scopus 로고
    • Autophagy is increased after traumatic brain injury in mice and is partially inhibited by the antioxidant gamma-glutamylcysteinyl ethyl ester
    • Lai Y, Hickey RW, Chen Y, Bayir H, Sullivan ML, Chu CT, et al. Autophagy is increased after traumatic brain injury in mice and is partially inhibited by the antioxidant gamma-glutamylcysteinyl ethyl ester. J Cereb Blood Flow Metab (2008) 28:540-50. doi:10.1038/sj.jcbfm.9600551
    • (2008) J Cereb Blood Flow Metab , vol.28 , pp. 540-550
    • Lai, Y.1    Hickey, R.W.2    Chen, Y.3    Bayir, H.4    Sullivan, M.L.5    Chu, C.T.6
  • 239
    • 47649126513 scopus 로고    scopus 로고
    • Autophagy is activated and might protect neurons from degeneration after traumatic brain injury
    • Zhang YB, Li SX, Chen XP, Yang L, Zhang YG, Liu R, et al. Autophagy is activated and might protect neurons from degeneration after traumatic brain injury. Neurosci Bull (2008) 24:143-9. doi:10.1007/s12264-008-1108-0
    • (2008) Neurosci Bull , vol.24 , pp. 143-149
    • Zhang, Y.B.1    Li, S.X.2    Chen, X.P.3    Yang, L.4    Zhang, Y.G.5    Liu, R.6
  • 240
    • 84866742588 scopus 로고    scopus 로고
    • Necrostatin-1 suppresses autophagy and apoptosis in mice traumatic brain injury model
    • Wang YQ, Wang L, Zhang MY, Wang T, Bao HJ, Liu WL, et al. Necrostatin-1 suppresses autophagy and apoptosis in mice traumatic brain injury model. Neurochem Res (2012) 37:1849-58. doi:10.1007/s11064-012-0791-4
    • (2012) Neurochem Res , vol.37 , pp. 1849-1858
    • Wang, Y.Q.1    Wang, L.2    Zhang, M.Y.3    Wang, T.4    Bao, H.J.5    Liu, W.L.6
  • 241
    • 79956011038 scopus 로고    scopus 로고
    • Autophagy is involved in traumatic brain injury-induced cell death and contributes to functional outcome deficits in mice
    • Luo CL, Li BX, Li QQ, Chen XP, Sun YX, Bao HJ, et al. Autophagy is involved in traumatic brain injury-induced cell death and contributes to functional outcome deficits in mice. Neuroscience (2011) 184:54-63. doi:10.1016/j.neuroscience.2011.03.021
    • (2011) Neuroscience , vol.184 , pp. 54-63
    • Luo, C.L.1    Li, B.X.2    Li, Q.Q.3    Chen, X.P.4    Sun, Y.X.5    Bao, H.J.6
  • 242
    • 59249102240 scopus 로고    scopus 로고
    • Cytosolic LC3 ratio as a quantitative index of macroautophagy
    • Kadowaki M, Karim MR. Cytosolic LC3 ratio as a quantitative index of macroautophagy. Methods Enzymol (2009) 452:199-213. doi:10.1016/S0076-6879(08)03613-6
    • (2009) Methods Enzymol , vol.452 , pp. 199-213
    • Kadowaki, M.1    Karim, M.R.2
  • 243
    • 79952628267 scopus 로고    scopus 로고
    • The beclin 1 network regulates autophagy and apoptosis
    • Kang R, Zeh HJ, Lotze MT, Tang D. The beclin 1 network regulates autophagy and apoptosis. Cell Death Differ (2011) 18:571-80. doi:10.1038/cdd.2010.191
    • (2011) Cell Death Differ , vol.18 , pp. 571-580
    • Kang, R.1    Zeh, H.J.2    Lotze, M.T.3    Tang, D.4
  • 245
    • 77951217000 scopus 로고    scopus 로고
    • Dual role of 3-methyladenine in modulation of autophagy via different temporal patterns of inhibition on class I and III phosphoinositide 3-kinase
    • Wu YT, Tan HL, Shui G, Bauvy C, Huang Q, Wenk MR, et al. Dual role of 3-methyladenine in modulation of autophagy via different temporal patterns of inhibition on class I and III phosphoinositide 3-kinase. J Biol Chem (2010) 285:10850-61. doi:10.1074/jbc.M109.080796
    • (2010) J Biol Chem , vol.285 , pp. 10850-10861
    • Wu, Y.T.1    Tan, H.L.2    Shui, G.3    Bauvy, C.4    Huang, Q.5    Wenk, M.R.6
  • 246
    • 38949108670 scopus 로고    scopus 로고
    • Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes
    • Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, Askew DS, et al. Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy (2008) 4:151-75. doi:10.4161/auto.5338
    • (2008) Autophagy , vol.4 , pp. 151-175
    • Klionsky, D.J.1    Abeliovich, H.2    Agostinis, P.3    Agrawal, D.K.4    Aliev, G.5    Askew, D.S.6
  • 247
    • 4944256085 scopus 로고    scopus 로고
    • Caspase inhibitor z-DEVD-fmk attenuates calpain and necrotic cell death in vitro and after traumatic brain injury
    • Knoblach SM, Alroy DA, Nikolaeva M, Cernak I, Stoica BA, Faden AI. Caspase inhibitor z-DEVD-fmk attenuates calpain and necrotic cell death in vitro and after traumatic brain injury. J Cereb Blood Flow Metab (2004) 24:1119-32. doi:10.1097/01.WCB.0000138664.17682.32
    • (2004) J Cereb Blood Flow Metab , vol.24 , pp. 1119-1132
    • Knoblach, S.M.1    Alroy, D.A.2    Nikolaeva, M.3    Cernak, I.4    Stoica, B.A.5    Faden, A.I.6
  • 248
    • 84887389490 scopus 로고    scopus 로고
    • Inhibition of cathepsin S produces neuroprotective effects after traumatic brain injury in mice
    • Xu J, Wang H, Ding K, Lu X, Li T, Wang J, et al. Inhibition of cathepsin S produces neuroprotective effects after traumatic brain injury in mice. Mediators Inflamm (2013) 2013:187873. doi:10.1155/2013/187873
    • (2013) Mediators Inflamm , vol.2013
    • Xu, J.1    Wang, H.2    Ding, K.3    Lu, X.4    Li, T.5    Wang, J.6
  • 249
    • 0034625693 scopus 로고    scopus 로고
    • E-64-d prevents both calpain upregulation and apoptosis in the lesion and penumbra following spinal cord injury in rats
    • Ray SK, Matzelle DC, Wilford GG, Hogan EL, Banik NL. E-64-d prevents both calpain upregulation and apoptosis in the lesion and penumbra following spinal cord injury in rats. Brain Res (2000) 867:80-9. doi:10.1016/S0006-8993(00)02260-5
    • (2000) Brain Res , vol.867 , pp. 80-89
    • Ray, S.K.1    Matzelle, D.C.2    Wilford, G.G.3    Hogan, E.L.4    Banik, N.L.5
  • 250
    • 0025184647 scopus 로고
    • Suppressive effect of E-64c on ischemic degradation of cerebral proteins following occlusion of the middle cerebral artery in rats
    • Inuzuka T, Tamura A, Sato S, Kirino T, Toyoshima I, Miyatake T. Suppressive effect of E-64c on ischemic degradation of cerebral proteins following occlusion of the middle cerebral artery in rats. Brain Res (1990) 526:177-9. doi:10.1016/0006-8993(90)90269-H
    • (1990) Brain Res , vol.526 , pp. 177-179
    • Inuzuka, T.1    Tamura, A.2    Sato, S.3    Kirino, T.4    Toyoshima, I.5    Miyatake, T.6
  • 251
    • 0035906534 scopus 로고    scopus 로고
    • A selective cysteine protease inhibitor is non-toxic and cerebroprotective in rats undergoing transient middle cerebral artery ischemia
    • Seyfried DM, Veyna R, Han Y, Li K, Tang N, Betts RL, et al. A selective cysteine protease inhibitor is non-toxic and cerebroprotective in rats undergoing transient middle cerebral artery ischemia. Brain Res (2001) 901:94-101. doi:10.1016/S0006-8993(01)02289-2
    • (2001) Brain Res , vol.901 , pp. 94-101
    • Seyfried, D.M.1    Veyna, R.2    Han, Y.3    Li, K.4    Tang, N.5    Betts, R.L.6
  • 252
    • 0036938197 scopus 로고    scopus 로고
    • Primate neurons show different vulnerability to transient ischemia and response to cathepsin inhibition
    • Yoshida M, Yamashima T, Zhao L, Tsuchiya K, Kohda Y, Tonchev AB, et al. Primate neurons show different vulnerability to transient ischemia and response to cathepsin inhibition. Acta Neuropathol (2002) 104:267-72.
    • (2002) Acta Neuropathol , vol.104 , pp. 267-272
    • Yoshida, M.1    Yamashima, T.2    Zhao, L.3    Tsuchiya, K.4    Kohda, Y.5    Tonchev, A.B.6
  • 253
    • 79953692131 scopus 로고    scopus 로고
    • Improvement in recovery after experimental intracerebral hemorrhage using a selective cathepsin B and L inhibitor
    • Yang D, Han Y, Zhang J, Ding C, Anagli J, Seyfried DM. Improvement in recovery after experimental intracerebral hemorrhage using a selective cathepsin B and L inhibitor. J Neurosurg (2011) 114:1110-6. doi:10.3171/2010.6.JNS091856
    • (2011) J Neurosurg , vol.114 , pp. 1110-1116
    • Yang, D.1    Han, Y.2    Zhang, J.3    Ding, C.4    Anagli, J.5    Seyfried, D.M.6
  • 255
    • 48749089723 scopus 로고    scopus 로고
    • Inhibition of calpains improves memory and synaptic transmission in a mouse model of Alzheimer disease
    • Trinchese F, Fa M, Liu S, Zhang H, Hidalgo A, Schmidt SD, et al. Inhibition of calpains improves memory and synaptic transmission in a mouse model of Alzheimer disease. J Clin Invest (2008) 118:2796-807. doi:10.1172/JCI34254
    • (2008) J Clin Invest , vol.118 , pp. 2796-2807
    • Trinchese, F.1    Fa, M.2    Liu, S.3    Zhang, H.4    Hidalgo, A.5    Schmidt, S.D.6
  • 256
    • 80052821752 scopus 로고    scopus 로고
    • The cysteine protease inhibitor, E64d, reduces brain amyloid-beta and improves memory deficits in Alzheimer's disease animal models by inhibiting cathepsin B, but not BACE1, beta-secretase activity
    • Hook G, Hook V, Kindy M. The cysteine protease inhibitor, E64d, reduces brain amyloid-beta and improves memory deficits in Alzheimer's disease animal models by inhibiting cathepsin B, but not BACE1, beta-secretase activity. J Alzheimers Dis (2011) 26:387-408. doi:10.3233/JAD-2011-110101
    • (2011) J Alzheimers Dis , vol.26 , pp. 387-408
    • Hook, G.1    Hook, V.2    Kindy, M.3
  • 257
    • 0017841848 scopus 로고
    • Isolation and characterization of E-64, a new thiol protease inhibitor
    • Hanada K, Tamai M, Yamagishi S, Ohmura J, Sawada I, Tanaka H. Isolation and characterization of E-64, a new thiol protease inhibitor. Agric Biol Chem (1978) 42:523-8. doi:10.1271/bbb1961.42.523
    • (1978) Agric Biol Chem , vol.42 , pp. 523-528
    • Hanada, K.1    Tamai, M.2    Yamagishi, S.3    Ohmura, J.4    Sawada, I.5    Tanaka, H.6
  • 258
  • 259
    • 0022632034 scopus 로고
    • Inactivation of calpain I and calpain II by specificity-oriented tripeptidyl chloromethyl ketones
    • Sasaki T, Kikuchi T, Fukui I, Murachi T. Inactivation of calpain I and calpain II by specificity-oriented tripeptidyl chloromethyl ketones. J Biochem (1986) 99:173-9.
    • (1986) J Biochem , vol.99 , pp. 173-179
    • Sasaki, T.1    Kikuchi, T.2    Fukui, I.3    Murachi, T.4
  • 260
    • 4344652798 scopus 로고    scopus 로고
    • Human primary osteoclasts: in vitro generation and applications as pharmacological and clinical assay
    • Susa M, Luong-Nguyen NH, Cappellen D, Zamurovic N, Gamse R. Human primary osteoclasts: in vitro generation and applications as pharmacological and clinical assay. J Transl Med (2004) 2:6. doi:10.1186/1479-5876-2-6
    • (2004) J Transl Med , vol.2 , pp. 6
    • Susa, M.1    Luong-Nguyen, N.H.2    Cappellen, D.3    Zamurovic, N.4    Gamse, R.5
  • 261
    • 0019274745 scopus 로고
    • Inhibitions by E-64 derivatives of rat liver cathepsin B and cathepsin L in vitro and in vivo
    • Hashida S, Towatari T, Kominami E, Katunuma N. Inhibitions by E-64 derivatives of rat liver cathepsin B and cathepsin L in vitro and in vivo. J Biochem (1980) 88:1805-11.
    • (1980) J Biochem , vol.88 , pp. 1805-1811
    • Hashida, S.1    Towatari, T.2    Kominami, E.3    Katunuma, N.4
  • 262
    • 0022997931 scopus 로고
    • In vitro and in vivo inhibition of cysteine proteinases by EST, a new analog of E-64
    • Tamai M, Matsumoto K, Omura S, Koyama I, Ozawa Y, Hanada K. In vitro and in vivo inhibition of cysteine proteinases by EST, a new analog of E-64. J Pharmacobiodyn (1986) 9:672-7. doi:10.1248/bpb1978.9.672
    • (1986) J Pharmacobiodyn , vol.9 , pp. 672-677
    • Tamai, M.1    Matsumoto, K.2    Omura, S.3    Koyama, I.4    Ozawa, Y.5    Hanada, K.6
  • 264
    • 0026101682 scopus 로고
    • Novel epoxysuccinyl peptides. Selective inhibitors of cathepsin B, in vitro
    • Murata M, Miyashita S, Yokoo C, Tamai M, Hanada K, Hatayama K, et al. Novel epoxysuccinyl peptides. Selective inhibitors of cathepsin B, in vitro. FEBS Lett (1991) 280:307-10. doi:10.1016/0014-5793(91)80318-W
    • (1991) FEBS Lett , vol.280 , pp. 307-310
    • Murata, M.1    Miyashita, S.2    Yokoo, C.3    Tamai, M.4    Hanada, K.5    Hatayama, K.6
  • 265
    • 0026029556 scopus 로고
    • Novel epoxysuccinyl peptides. A selective inhibitor of cathepsin B, in vivo
    • Towatari T, Nikawa T, Murata M, Yokoo C, Tamai M, Hanada K, et al. Novel epoxysuccinyl peptides. A selective inhibitor of cathepsin B, in vivo. FEBS Lett (1991) 280:311-5. doi:10.1016/0014-5793(91)80319-X
    • (1991) FEBS Lett , vol.280 , pp. 311-315
    • Towatari, T.1    Nikawa, T.2    Murata, M.3    Yokoo, C.4    Tamai, M.5    Hanada, K.6
  • 266
    • 79951613664 scopus 로고    scopus 로고
    • Structure-based development of specific inhibitors for individual cathepsins and their medical applications
    • Katunuma N. Structure-based development of specific inhibitors for individual cathepsins and their medical applications. Proc Jpn Acad Ser B Phys Biol Sci (2011) 87:29-39. doi:10.2183/pjab.87.29
    • (2011) Proc Jpn Acad Ser B Phys Biol Sci , vol.87 , pp. 29-39
    • Katunuma, N.1
  • 267
    • 0036661078 scopus 로고    scopus 로고
    • CA-074, but not its methyl ester CA-074Me, is a selective inhibitor of cathepsin B within living cells
    • Montaser M, Lalmanach G, Mach L. CA-074, but not its methyl ester CA-074Me, is a selective inhibitor of cathepsin B within living cells. Biol Chem (2002) 383:1305-8. doi:10.1515/BC.2002.147
    • (2002) Biol Chem , vol.383 , pp. 1305-1308
    • Montaser, M.1    Lalmanach, G.2    Mach, L.3
  • 268
    • 0034054576 scopus 로고    scopus 로고
    • Selective targeting of lysosomal cysteine proteases with radiolabeled electrophilic substrate analogs
    • Bogyo M, Verhelst S, Bellingard-Dubouchaud V, Toba S, Greenbaum D. Selective targeting of lysosomal cysteine proteases with radiolabeled electrophilic substrate analogs. Chem Biol (2000) 7:27-38. doi:10.1016/S1074-5521(00)00061-2
    • (2000) Chem Biol , vol.7 , pp. 27-38
    • Bogyo, M.1    Verhelst, S.2    Bellingard-Dubouchaud, V.3    Toba, S.4    Greenbaum, D.5
  • 269
    • 0027053210 scopus 로고
    • CA074 methyl ester: a proinhibitor for intracellular cathepsin B
    • Buttle DJ, Murata M, Knight CG, Barrett AJ. CA074 methyl ester: a proinhibitor for intracellular cathepsin B. Arch Biochem Biophys (1992) 299:377-80. doi:10.1016/0003-9861(92)90290-D
    • (1992) Arch Biochem Biophys , vol.299 , pp. 377-380
    • Buttle, D.J.1    Murata, M.2    Knight, C.G.3    Barrett, A.J.4
  • 270
    • 12444315984 scopus 로고    scopus 로고
    • Inhibition of papain-like cysteine proteases and legumain by caspase-specific inhibitors: when reaction mechanism is more important than specificity
    • Rozman-Pungercar J, Kopitar-Jerala N, Bogyo M, Turk D, Vasiljeva O, Stefe I, et al. Inhibition of papain-like cysteine proteases and legumain by caspase-specific inhibitors: when reaction mechanism is more important than specificity. Cell Death Differ (2003) 10:881-8. doi:10.1038/sj.cdd.4401247
    • (2003) Cell Death Differ , vol.10 , pp. 881-888
    • Rozman-Pungercar, J.1    Kopitar-Jerala, N.2    Bogyo, M.3    Turk, D.4    Vasiljeva, O.5    Stefe, I.6
  • 271
    • 0032898735 scopus 로고    scopus 로고
    • Non-specific effects of methyl ketone peptide inhibitors of caspases
    • Schotte P, Declercq W, Van Huffel S, Vandenabeele P, Beyaert R. Non-specific effects of methyl ketone peptide inhibitors of caspases. FEBS Lett (1999) 442:117-21. doi:10.1016/S0014-5793(98)01640-8
    • (1999) FEBS Lett , vol.442 , pp. 117-121
    • Schotte, P.1    Declercq, W.2    Van Huffel, S.3    Vandenabeele, P.4    Beyaert, R.5
  • 272
    • 0031793017 scopus 로고    scopus 로고
    • Atractyloside-induced release of cathepsin B, a protease with caspase-processing activity
    • Vancompernolle K, Van Herreweghe F, Pynaert G, Van De Craen M, De Vos K, Totty N, et al. Atractyloside-induced release of cathepsin B, a protease with caspase-processing activity. FEBS Lett (1998) 438:150-8. doi:10.1016/S0014-5793(98)01275-7
    • (1998) FEBS Lett , vol.438 , pp. 150-158
    • Vancompernolle, K.1    Van Herreweghe, F.2    Pynaert, G.3    Van De Craen, M.4    De Vos, K.5    Totty, N.6
  • 273
    • 0033791657 scopus 로고    scopus 로고
    • In vitro evaluation of the disposition of a novel cysteine protease inhibitor
    • Jacobsen W, Christians U, Benet LZ. In vitro evaluation of the disposition of a novel cysteine protease inhibitor. Drug Metab Dispos (2000) 28:1343-51.
    • (2000) Drug Metab Dispos , vol.28 , pp. 1343-1351
    • Jacobsen, W.1    Christians, U.2    Benet, L.Z.3
  • 274
    • 84952937311 scopus 로고    scopus 로고
    • Serum GFAP predicts tissue GFAP break down products and therapeutic efficacy after penetrating ballistic-like brain injury
    • Boutte A, Deng-Bryant Y, Johnson D, Tortella FC, Dave J, Shear DA, et al. Serum GFAP predicts tissue GFAP break down products and therapeutic efficacy after penetrating ballistic-like brain injury. J Neurotrauma (2015). doi:10.1089/neu.2014.3672
    • (2015) J Neurotrauma
    • Boutte, A.1    Deng-Bryant, Y.2    Johnson, D.3    Tortella, F.C.4    Dave, J.5    Shear, D.A.6
  • 275
    • 79960391913 scopus 로고    scopus 로고
    • A novel multicenter preclinical drug screening and biomarker consortium for experimental traumatic brain injury: operation brain trauma therapy
    • Kochanek PM, Bramlett H, Dietrich WD, Dixon CE, Hayes RL, Povlishock J, et al. A novel multicenter preclinical drug screening and biomarker consortium for experimental traumatic brain injury: operation brain trauma therapy. J Trauma (2011) 71:S15-24. doi:10.1097/TA.0b013e31822117fe
    • (2011) J Trauma , vol.71 , pp. S15-S24
    • Kochanek, P.M.1    Bramlett, H.2    Dietrich, W.D.3    Dixon, C.E.4    Hayes, R.L.5    Povlishock, J.6
  • 276
    • 34247203094 scopus 로고    scopus 로고
    • Temporal relationship of peroxynitrite-induced oxidative damage, calpain-mediated cytoskeletal degradation and neurodegeneration after traumatic brain injury
    • Deng Y, Thompson BM, Gao X, Hall ED. Temporal relationship of peroxynitrite-induced oxidative damage, calpain-mediated cytoskeletal degradation and neurodegeneration after traumatic brain injury. Exp Neurol (2007) 205:154-65. doi:10.1016/j.expneurol.2007.01.023
    • (2007) Exp Neurol , vol.205 , pp. 154-165
    • Deng, Y.1    Thompson, B.M.2    Gao, X.3    Hall, E.D.4
  • 277
    • 84859748670 scopus 로고    scopus 로고
    • Targeted gene inactivation of calpain-1 suppresses cortical degeneration due to traumatic brain injury and neuronal apoptosis induced by oxidative stress
    • Yamada KH, Kozlowski DA, Seidl SE, Lance S, Wieschhaus AJ, Sundivakkam P, et al. Targeted gene inactivation of calpain-1 suppresses cortical degeneration due to traumatic brain injury and neuronal apoptosis induced by oxidative stress. J Biol Chem (2012) 287:13182-93. doi:10.1074/jbc.M111.302612
    • (2012) J Biol Chem , vol.287 , pp. 13182-13193
    • Yamada, K.H.1    Kozlowski, D.A.2    Seidl, S.E.3    Lance, S.4    Wieschhaus, A.J.5    Sundivakkam, P.6
  • 278
    • 0034666112 scopus 로고    scopus 로고
    • Effects of matrix metalloproteinase-9 gene knock-out on morphological and motor outcomes after traumatic brain injury
    • Wang X, Jung J, Asahi M, Chwang W, Russo L, Moskowitz MA, et al. Effects of matrix metalloproteinase-9 gene knock-out on morphological and motor outcomes after traumatic brain injury. J Neurosci (2000) 20:7037-42.
    • (2000) J Neurosci , vol.20 , pp. 7037-7042
    • Wang, X.1    Jung, J.2    Asahi, M.3    Chwang, W.4    Russo, L.5    Moskowitz, M.A.6
  • 279
    • 1842538729 scopus 로고    scopus 로고
    • Blockade of cathepsin B expression in human glioblastoma cells is associated with suppression of angiogenesis
    • Yanamandra N, Gumidyala KV, Waldron KG, Gujrati M, Olivero WC, Dinh DH, et al. Blockade of cathepsin B expression in human glioblastoma cells is associated with suppression of angiogenesis. Oncogene (2004) 23:2224-30. doi:10.1038/sj.onc.1207338
    • (2004) Oncogene , vol.23 , pp. 2224-2230
    • Yanamandra, N.1    Gumidyala, K.V.2    Waldron, K.G.3    Gujrati, M.4    Olivero, W.C.5    Dinh, D.H.6
  • 280
    • 33745968813 scopus 로고    scopus 로고
    • Cathepsin and calpain inhibitor E64d attenuates matrix metalloproteinase-9 activity after focal cerebral ischemia in rats
    • Tsubokawa T, Solaroglu I, Yatsushige H, Cahill J, Yata K, Zhang JH. Cathepsin and calpain inhibitor E64d attenuates matrix metalloproteinase-9 activity after focal cerebral ischemia in rats. Stroke (2006) 37:1888-94. doi:10.1161/01.STR.0000227259.15506.24
    • (2006) Stroke , vol.37 , pp. 1888-1894
    • Tsubokawa, T.1    Solaroglu, I.2    Yatsushige, H.3    Cahill, J.4    Yata, K.5    Zhang, J.H.6
  • 281
    • 84881466563 scopus 로고    scopus 로고
    • Design, synthesis, and optimization of novel epoxide incorporating peptidomimetics as selective calpain inhibitors
    • Schiefer IT, Tapadar S, Litosh V, Siklos M, Scism R, Wijewickrama GT, et al. Design, synthesis, and optimization of novel epoxide incorporating peptidomimetics as selective calpain inhibitors. J Med Chem (2013) 56:6054-68. doi:10.1021/jm4006719
    • (2013) J Med Chem , vol.56 , pp. 6054-6068
    • Schiefer, I.T.1    Tapadar, S.2    Litosh, V.3    Siklos, M.4    Scism, R.5    Wijewickrama, G.T.6
  • 282
    • 0023607871 scopus 로고
    • Prolongation of life span of dystrophic hamster by cysteine proteinase inhibitor, loxistation (EST)
    • Tamai M, Omura S, Kimura M, Hanada K, Sugita H. Prolongation of life span of dystrophic hamster by cysteine proteinase inhibitor, loxistation (EST). J Pharmacobiodyn (1987) 10:678-81. doi:10.1248/bpb1978.10.678
    • (1987) J Pharmacobiodyn , vol.10 , pp. 678-681
    • Tamai, M.1    Omura, S.2    Kimura, M.3    Hanada, K.4    Sugita, H.5
  • 283
    • 0026656202 scopus 로고
    • Therapeutic trials on progressive muscular dystrophy
    • Satoyoshi E. Therapeutic trials on progressive muscular dystrophy. Intern Med (1992) 31:841-6. doi:10.2169/internalmedicine.31.841
    • (1992) Intern Med , vol.31 , pp. 841-846
    • Satoyoshi, E.1
  • 284
    • 80052817648 scopus 로고
    • Pharmacokinetics of EST (report 5): pharmacokinetics of EST ih humans
    • Watanabe T, Fukushima K, Ushiyama Y, Noda K, Suwa T. Pharmacokinetics of EST (report 5): pharmacokinetics of EST ih humans. Kiso Rinsho (1986) 20:362-6.
    • (1986) Kiso Rinsho , vol.20 , pp. 362-366
    • Watanabe, T.1    Fukushima, K.2    Ushiyama, Y.3    Noda, K.4    Suwa, T.5
  • 285
    • 0019948262 scopus 로고
    • L-trans-Epoxysuccinyl-leucylamido(4-guanidino)butane (E-64) and its analogues as inhibitors of cysteine proteinases including cathepsins B, H and L
    • Barrett AJ, Kembhavi AA, Brown MA, Kirschke H, Knight CG, Tamai M, et al. L-trans-Epoxysuccinyl-leucylamido(4-guanidino)butane (E-64) and its analogues as inhibitors of cysteine proteinases including cathepsins B, H and L. Biochem J (1982) 201:189-98.
    • (1982) Biochem J , vol.201 , pp. 189-198
    • Barrett, A.J.1    Kembhavi, A.A.2    Brown, M.A.3    Kirschke, H.4    Knight, C.G.5    Tamai, M.6
  • 286
    • 0020755321 scopus 로고
    • Reaction of calcium-activated neutral protease (CANP) with an epoxysuccinyl derivative (E64c) and iodoacetic acid
    • Suzuki K. Reaction of calcium-activated neutral protease (CANP) with an epoxysuccinyl derivative (E64c) and iodoacetic acid. J Biochem (1983) 93:1305-12.
    • (1983) J Biochem , vol.93 , pp. 1305-1312
    • Suzuki, K.1
  • 287
    • 50649112213 scopus 로고    scopus 로고
    • Activity-based protein profiling: from enzyme chemistry to proteomic chemistry
    • Cravatt BF, Wright AT, Kozarich JW. Activity-based protein profiling: from enzyme chemistry to proteomic chemistry. Annu Rev Biochem (2008) 77:383-414. doi:10.1146/annurev.biochem.75.101304.124125
    • (2008) Annu Rev Biochem , vol.77 , pp. 383-414
    • Cravatt, B.F.1    Wright, A.T.2    Kozarich, J.W.3
  • 288
    • 84902142324 scopus 로고    scopus 로고
    • Enzyme inhibitor discovery by activity-based protein profiling
    • Niphakis MJ, Cravatt BF. Enzyme inhibitor discovery by activity-based protein profiling. Annu Rev Biochem (2014) 83:341-77. doi:10.1146/annurev-biochem-060713-035708
    • (2014) Annu Rev Biochem , vol.83 , pp. 341-377
    • Niphakis, M.J.1    Cravatt, B.F.2
  • 289
    • 84902181290 scopus 로고    scopus 로고
    • Activity-based profiling of proteases
    • Sanman LE, Bogyo M. Activity-based profiling of proteases. Annu Rev Biochem (2014) 83:249-73. doi:10.1146/annurev-biochem-060713-035352
    • (2014) Annu Rev Biochem , vol.83 , pp. 249-273
    • Sanman, L.E.1    Bogyo, M.2
  • 290
    • 34548666006 scopus 로고    scopus 로고
    • Noninvasive optical imaging of cysteine protease activity using fluorescently quenched activity-based probes
    • Blum G, Von Degenfeld G, Merchant MJ, Blau HM, Bogyo M. Noninvasive optical imaging of cysteine protease activity using fluorescently quenched activity-based probes. Nat Chem Biol (2007) 3:668-77. doi:10.1038/nchembio.2007.26
    • (2007) Nat Chem Biol , vol.3 , pp. 668-677
    • Blum, G.1    Von Degenfeld, G.2    Merchant, M.J.3    Blau, H.M.4    Bogyo, M.5
  • 291
    • 80051504987 scopus 로고    scopus 로고
    • Identification of a myeloid-derived suppressor cell cystatin-like protein that inhibits metastasis
    • Boutte AM, Friedman DB, Bogyo M, Min Y, Yang L, Lin PC. Identification of a myeloid-derived suppressor cell cystatin-like protein that inhibits metastasis. FASEB J (2011) 25:2626-37. doi:10.1096/fj.10-180604
    • (2011) FASEB J , vol.25 , pp. 2626-2637
    • Boutte, A.M.1    Friedman, D.B.2    Bogyo, M.3    Min, Y.4    Yang, L.5    Lin, P.C.6
  • 292
  • 293
  • 294
    • 0019800744 scopus 로고
    • The effect of an in vivo-injected thiol protease inhibitor, E-64-c, on the calcium-induced degeneration of myofilaments
    • Ishiura S, Hanada K, Tamai M, Kashiwagi K, Sugita H. The effect of an in vivo-injected thiol protease inhibitor, E-64-c, on the calcium-induced degeneration of myofilaments. J Biochem (1981) 90:1557-60.
    • (1981) J Biochem , vol.90 , pp. 1557-1560
    • Ishiura, S.1    Hanada, K.2    Tamai, M.3    Kashiwagi, K.4    Sugita, H.5
  • 295
    • 80052818257 scopus 로고
    • Clinical phase I trial of thiol protease inhibitor (Report 2): safety and pharmacokinetics in continuous administration
    • Miyahara T, Shimojo S, Toyohara K, Imai T, Miyajima M, Honda H, et al. Clinical phase I trial of thiol protease inhibitor (Report 2): safety and pharmacokinetics in continuous administration. Rinsho Yakuri (1985) 16:537-46. doi:10.3999/jscpt.16.357
    • (1985) Rinsho Yakuri , vol.16 , pp. 537-546
    • Miyahara, T.1    Shimojo, S.2    Toyohara, K.3    Imai, T.4    Miyajima, M.5    Honda, H.6
  • 296
    • 80052811526 scopus 로고
    • Phase I clinical trial of thiol protease inhibitor EST (report 1): safety and pharmacokinetics with single administration
    • Miyahara T, Shimojo S, Toyohara K, Imai T, Miyajima M, Honda H, et al. Phase I clinical trial of thiol protease inhibitor EST (report 1): safety and pharmacokinetics with single administration. Rinsho Yakuri (1985) 16:357-65. doi:10.3999/jscpt.16.357
    • (1985) Rinsho Yakuri , vol.16 , pp. 357-365
    • Miyahara, T.1    Shimojo, S.2    Toyohara, K.3    Imai, T.4    Miyajima, M.5    Honda, H.6
  • 298
    • 84896842296 scopus 로고
    • Toxicological studies on ethyl( + )-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST)(report IX) mutagenicity study
    • Yasui H, Goto H, Suzuki H, Sakai S, Takamura T, Nakane S. Toxicological studies on ethyl( + )-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST)(report IX) mutagenicity study. Iyakuhin Kenkyu (1986) 17:815-25.
    • (1986) Iyakuhin Kenkyu , vol.17 , pp. 815-825
    • Yasui, H.1    Goto, H.2    Suzuki, H.3    Sakai, S.4    Takamura, T.5    Nakane, S.6
  • 299
    • 80052805278 scopus 로고
    • Toxicological studies of ethyl (+)-(2S,3S)-3-[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report 1): acute toxicity studies of EST and metabolite and by-product of EST
    • Setoyama K, Koike M, Abe S, Tsutsui Y, Tarumoto Y, Nakane S. Toxicological studies of ethyl (+)-(2S,3S)-3-[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report 1): acute toxicity studies of EST and metabolite and by-product of EST. Iyakuhin Kenkyu (1986) 17:736-43.
    • (1986) Iyakuhin Kenkyu , vol.17 , pp. 736-743
    • Setoyama, K.1    Koike, M.2    Abe, S.3    Tsutsui, Y.4    Tarumoto, Y.5    Nakane, S.6
  • 300
    • 84896899541 scopus 로고
    • Toxicological studies on ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST)(report V) subacute toxicity in dogs
    • Tarumoto Y, Sakagawa T, Tsutsui Y, Kawanishi M, Kimura M, Nakane S. Toxicological studies on ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST)(report V) subacute toxicity in dogs. Iyakuhin Kenkyu (1986) 17:768-80.
    • (1986) Iyakuhin Kenkyu , vol.17 , pp. 768-780
    • Tarumoto, Y.1    Sakagawa, T.2    Tsutsui, Y.3    Kawanishi, M.4    Kimura, M.5    Nakane, S.6
  • 301
    • 84896820649 scopus 로고
    • Toxicological studies on ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report III) subacute toxicity in rats
    • Kimura M, Yagi K, Fujinuma S, Tsuchida T, Tarumoto Y, Noda K, et al. Toxicological studies on ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report III) subacute toxicity in rats. Iyakuhin Kenkyu (1986) 17:744-67.
    • (1986) Iyakuhin Kenkyu , vol.17 , pp. 744-767
    • Kimura, M.1    Yagi, K.2    Fujinuma, S.3    Tsuchida, T.4    Tarumoto, Y.5    Noda, K.6
  • 302
    • 84896899541 scopus 로고
    • Toxicological studies on ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST)(report VII) chronic toxicity study in dogs
    • Tarumoto Y, Sakagawa T, Tsutsui Y, Kawanishi M, Watanabe T, Nakane S. Toxicological studies on ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST)(report VII) chronic toxicity study in dogs. Iyakuhin Kenkyu (1986) 17:802-14.
    • (1986) Iyakuhin Kenkyu , vol.17 , pp. 802-814
    • Tarumoto, Y.1    Sakagawa, T.2    Tsutsui, Y.3    Kawanishi, M.4    Watanabe, T.5    Nakane, S.6
  • 303
    • 84896811168 scopus 로고
    • Toxicological studies on ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST)(report VI) Chronic toxicity study in rats
    • Ohshima T, Watanabe T, Nagato C, Kimura M, Tsuchida T, Nakane S. Toxicological studies on ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST)(report VI) Chronic toxicity study in rats. Iyakuhin Kenkyu (1986) 17:781-801.
    • (1986) Iyakuhin Kenkyu , vol.17 , pp. 781-801
    • Ohshima, T.1    Watanabe, T.2    Nagato, C.3    Kimura, M.4    Tsuchida, T.5    Nakane, S.6
  • 304
    • 0025123216 scopus 로고
    • An epoxysuccinic acid derivative(loxistatin)-induced hepatic injury in rats and hamsters
    • Fukushima K, Arai M, Kohno Y, Suwa T, Satoh T. An epoxysuccinic acid derivative(loxistatin)-induced hepatic injury in rats and hamsters. Toxicol Appl Pharmacol (1990) 105:1-12. doi:10.1016/0041-008X(90)90353-V
    • (1990) Toxicol Appl Pharmacol , vol.105 , pp. 1-12
    • Fukushima, K.1    Arai, M.2    Kohno, Y.3    Suwa, T.4    Satoh, T.5
  • 305
    • 84896805010 scopus 로고
    • Reproduction studies of ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report III): study of administration to rabbits during organogenesis
    • Yamada T, Nishiyama T, Ohno H, Nakane S. Reproduction studies of ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report III): study of administration to rabbits during organogenesis. Iyakuhin Kenkyu (1986) 17:632-8.
    • (1986) Iyakuhin Kenkyu , vol.17 , pp. 632-638
    • Yamada, T.1    Nishiyama, T.2    Ohno, H.3    Nakane, S.4
  • 306
    • 84896851428 scopus 로고
    • Reproduction studies of ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report II): study of administration to rats during organogenesis
    • Yamada T, Uchida H, Inoue T, Ohba Y, Nakane S. Reproduction studies of ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report II): study of administration to rats during organogenesis. Iyakuhin Kenkyu (1986) 14:617-31.
    • (1986) Iyakuhin Kenkyu , vol.14 , pp. 617-631
    • Yamada, T.1    Uchida, H.2    Inoue, T.3    Ohba, Y.4    Nakane, S.5
  • 307
    • 84896805010 scopus 로고
    • Reproduction studies of ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report I): study of administration to rats prior to and in early stage of gestation
    • Yamada T, Nishiyama T, Nakane S. Reproduction studies of ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report I): study of administration to rats prior to and in early stage of gestation. Iyakuhin Kenkyu (1986) 17:609-16.
    • (1986) Iyakuhin Kenkyu , vol.17 , pp. 609-616
    • Yamada, T.1    Nishiyama, T.2    Nakane, S.3
  • 308
    • 84896805010 scopus 로고
    • Reproduction studies of ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report IV): study of administration to rats during perinatal and postnatal periods
    • Yamada T, Uchida H, Ohno H, Matsuzawa N, Nakane S. Reproduction studies of ethyl(+)-(2S,2S)-3[(S)-3-methyl-1-(3-methylbutylcarbamoyl)butylcarbamoyl]-2-oxiranecarboxylate (EST) (report IV): study of administration to rats during perinatal and postnatal periods. Iyakuhin Kenkyu (1986) 17:639-51.
    • (1986) Iyakuhin Kenkyu , vol.17 , pp. 639-651
    • Yamada, T.1    Uchida, H.2    Ohno, H.3    Matsuzawa, N.4    Nakane, S.5
  • 309
    • 74249093601 scopus 로고    scopus 로고
    • The identification, characterization and optimization of small molecule probes of cysteine proteases: experiences of the Penn Center for Molecular Discovery with cathepsin B and cathepsin L
    • Huryn DM, Smith AB III. The identification, characterization and optimization of small molecule probes of cysteine proteases: experiences of the Penn Center for Molecular Discovery with cathepsin B and cathepsin L. Curr Top Med Chem (2009) 9:1206-16. doi:10.2174/156802609789753653
    • (2009) Curr Top Med Chem , vol.9 , pp. 1206-1216
    • Huryn, D.M.1    Smith, A.B.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.