메뉴 건너뛰기




Volumn 1868, Issue 2, 2017, Pages 341-358

Functional analyses of major cancer-related signaling pathways in Alzheimer's disease etiology

Author keywords

Aging; Alzheimer's disease; Amyloid; Cell signaling; Tau; Tumorigenesis

Indexed keywords

AMYLOID BETA PROTEIN; HYPOXIA INDUCIBLE FACTOR; LONG UNTRANSLATED RNA; MAMMALIAN TARGET OF RAPAMYCIN; MICRORNA; MITOGEN ACTIVATED PROTEIN KINASE; MITOGEN ACTIVATED PROTEIN KINASE INHIBITOR; PEPTIDYL PROLYL CIS TRANS ISOMERASE NIMA INTERACTING 1; SIRTUIN 1; TRANSCRIPTION FACTOR FOXO; SIRT1 PROTEIN, HUMAN; TARGET OF RAPAMYCIN KINASE; TRANSCRIPTION FACTOR FKHR;

EID: 85025671165     PISSN: 0304419X     EISSN: 18792561     Source Type: Journal    
DOI: 10.1016/j.bbcan.2017.07.001     Document Type: Review
Times cited : (52)

References (353)
  • 1
    • 84873638532 scopus 로고    scopus 로고
    • Aging, cellular senescence, and cancer
    • Campisi, J., Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 75 (2013), 685–705.
    • (2013) Annu. Rev. Physiol. , vol.75 , pp. 685-705
    • Campisi, J.1
  • 2
    • 84949555897 scopus 로고    scopus 로고
    • Cellular senescence in aging and age-related disease: from mechanisms to therapy
    • Childs, B.G. et al., Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat. Med. 21:12 (2015), 1424–1435.
    • (2015) Nat. Med. , vol.21 , Issue.12 , pp. 1424-1435
    • Childs, B.G.1
  • 3
    • 79957900395 scopus 로고    scopus 로고
    • Aging and chronic DNA damage response activate a regulatory pathway involving miR-29 and p53
    • Ugalde, A.P. et al., Aging and chronic DNA damage response activate a regulatory pathway involving miR-29 and p53. EMBO J. 30:11 (2011), 2219–2232.
    • (2011) EMBO J. , vol.30 , Issue.11 , pp. 2219-2232
    • Ugalde, A.P.1
  • 4
    • 84890085638 scopus 로고    scopus 로고
    • Role of protein misfolding and proteostasis deficiency in protein misfolding diseases and aging
    • Cuanalo-Contreras, K., Mukherjee, A., Soto, C., Role of protein misfolding and proteostasis deficiency in protein misfolding diseases and aging. Int. J. Cell. Biol., 2013, 2013, 638083.
    • (2013) Int. J. Cell. Biol. , vol.2013 , pp. 638083
    • Cuanalo-Contreras, K.1    Mukherjee, A.2    Soto, C.3
  • 5
    • 0037264120 scopus 로고    scopus 로고
    • Unfolding the role of protein misfolding in neurodegenerative diseases
    • Soto, C., Unfolding the role of protein misfolding in neurodegenerative diseases. Nat. Rev. Neurosci. 4:1 (2003), 49–60.
    • (2003) Nat. Rev. Neurosci. , vol.4 , Issue.1 , pp. 49-60
    • Soto, C.1
  • 6
    • 0035964315 scopus 로고    scopus 로고
    • Does oxidative damage to DNA increase with age?
    • Hamilton, M.L. et al., Does oxidative damage to DNA increase with age?. Proc. Natl. Acad. Sci. U. S. A. 98:18 (2001), 10469–10474.
    • (2001) Proc. Natl. Acad. Sci. U. S. A. , vol.98 , Issue.18 , pp. 10469-10474
    • Hamilton, M.L.1
  • 7
    • 84916941205 scopus 로고    scopus 로고
    • Inverse association between cancer and neurodegenerative disease: review of the epidemiologic and biological evidence
    • Driver, J.A., Inverse association between cancer and neurodegenerative disease: review of the epidemiologic and biological evidence. Biogerontology 15:6 (2014), 547–557.
    • (2014) Biogerontology , vol.15 , Issue.6 , pp. 547-557
    • Driver, J.A.1
  • 9
    • 0035522894 scopus 로고    scopus 로고
    • Two genetic hits (more or less) to cancer
    • Knudson, A.G., Two genetic hits (more or less) to cancer. Nat. Rev. Cancer 1:2 (2001), 157–162.
    • (2001) Nat. Rev. Cancer , vol.1 , Issue.2 , pp. 157-162
    • Knudson, A.G.1
  • 10
    • 20444504698 scopus 로고    scopus 로고
    • The genetic epidemiology of neurodegenerative disease
    • Bertram, L., Tanzi, R.E., The genetic epidemiology of neurodegenerative disease. J. Clin. Invest. 115:6 (2005), 1449–1457.
    • (2005) J. Clin. Invest. , vol.115 , Issue.6 , pp. 1449-1457
    • Bertram, L.1    Tanzi, R.E.2
  • 11
    • 0024244746 scopus 로고
    • Survival time, causes of death, and tumor/treatment-related morbidity in 100 women with ovarian cancer
    • Dvoretsky, P.M. et al., Survival time, causes of death, and tumor/treatment-related morbidity in 100 women with ovarian cancer. Hum. Pathol. 19:11 (1988), 1273–1279.
    • (1988) Hum. Pathol. , vol.19 , Issue.11 , pp. 1273-1279
    • Dvoretsky, P.M.1
  • 12
    • 64149093013 scopus 로고    scopus 로고
    • Neurodegenerative diseases target large-scale human brain networks
    • Seeley, W.W. et al., Neurodegenerative diseases target large-scale human brain networks. Neuron 62:1 (2009), 42–52.
    • (2009) Neuron , vol.62 , Issue.1 , pp. 42-52
    • Seeley, W.W.1
  • 13
    • 0035066332 scopus 로고    scopus 로고
    • Alzheimer's disease: genes, proteins, and therapy
    • Selkoe, D.J., Alzheimer's disease: genes, proteins, and therapy. Physiol. Rev. 81:2 (2001), 741–766.
    • (2001) Physiol. Rev. , vol.81 , Issue.2 , pp. 741-766
    • Selkoe, D.J.1
  • 14
    • 84860554747 scopus 로고    scopus 로고
    • New and emerging treatments for Alzheimer's disease
    • Corbett, A., Smith, J., Ballard, C., New and emerging treatments for Alzheimer's disease. Expert. Rev. Neurother. 12:5 (2012), 535–543.
    • (2012) Expert. Rev. Neurother. , vol.12 , Issue.5 , pp. 535-543
    • Corbett, A.1    Smith, J.2    Ballard, C.3
  • 15
    • 65449161780 scopus 로고    scopus 로고
    • 2009 Alzheimer's disease facts and figures
    • Alzheimer's, A., 2009 Alzheimer's disease facts and figures. Alzheimers Dement. 5:3 (2009), 234–270.
    • (2009) Alzheimers Dement. , vol.5 , Issue.3 , pp. 234-270
    • Alzheimer's, A.1
  • 16
    • 33947178563 scopus 로고    scopus 로고
    • Alzheimer disease, the two-hit hypothesis: an update
    • Zhu, X. et al., Alzheimer disease, the two-hit hypothesis: an update. Biochim. Biophys. Acta 1772:4 (2007), 494–502.
    • (2007) Biochim. Biophys. Acta , vol.1772 , Issue.4 , pp. 494-502
    • Zhu, X.1
  • 17
    • 0037135111 scopus 로고    scopus 로고
    • The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics
    • Hardy, J., Selkoe, D.J., The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297:5580 (2002), 353–356.
    • (2002) Science , vol.297 , Issue.5580 , pp. 353-356
    • Hardy, J.1    Selkoe, D.J.2
  • 18
    • 0029999787 scopus 로고    scopus 로고
    • Alzheimer's disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules
    • Alonso, A.C., Grundke-Iqbal, I., Iqbal, K., Alzheimer's disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules. Nat. Med. 2:7 (1996), 783–787.
    • (1996) Nat. Med. , vol.2 , Issue.7 , pp. 783-787
    • Alonso, A.C.1    Grundke-Iqbal, I.2    Iqbal, K.3
  • 19
    • 75149142375 scopus 로고    scopus 로고
    • The molecular biology of senile plaques and neurofibrillary tangles in Alzheimer's disease
    • Armstrong, R.A., The molecular biology of senile plaques and neurofibrillary tangles in Alzheimer's disease. Folia Neuropathol. 47:4 (2009), 289–299.
    • (2009) Folia Neuropathol. , vol.47 , Issue.4 , pp. 289-299
    • Armstrong, R.A.1
  • 20
    • 84858046578 scopus 로고    scopus 로고
    • Neuropathological alterations in Alzheimer disease
    • Serrano-Pozo, A. et al., Neuropathological alterations in Alzheimer disease. Cold Spring Harb. Perspect. Med., 1(1), 2011, a006189.
    • (2011) Cold Spring Harb. Perspect. Med. , vol.1 , Issue.1 , pp. a006189
    • Serrano-Pozo, A.1
  • 21
    • 0026145130 scopus 로고
    • Use of brief cognitive tests to identify individuals in the community with clinically diagnosed Alzheimer's disease
    • Albert, M. et al., Use of brief cognitive tests to identify individuals in the community with clinically diagnosed Alzheimer's disease. Int. J. Neurosci. 57:3-4 (1991), 167–178.
    • (1991) Int. J. Neurosci. , vol.57 , Issue.3-4 , pp. 167-178
    • Albert, M.1
  • 22
    • 0022414054 scopus 로고
    • Diagnosis of Alzheimer's disease
    • Khachaturian, Z.S., Diagnosis of Alzheimer's disease. Arch. Neurol. 42:11 (1985), 1097–1105.
    • (1985) Arch. Neurol. , vol.42 , Issue.11 , pp. 1097-1105
    • Khachaturian, Z.S.1
  • 23
    • 84879083786 scopus 로고    scopus 로고
    • Fluid biomarkers in Alzheimer's disease - current concepts
    • Rosen, C. et al., Fluid biomarkers in Alzheimer's disease - current concepts. Mol. Neurodegener., 8, 2013, 20.
    • (2013) Mol. Neurodegener. , vol.8 , pp. 20
    • Rosen, C.1
  • 24
    • 84988452783 scopus 로고    scopus 로고
    • Potential fluid biomarkers for pathological brain changes in Alzheimer's disease: Implication for the screening of cognitive frailty
    • Ruan, Q. et al., Potential fluid biomarkers for pathological brain changes in Alzheimer's disease: Implication for the screening of cognitive frailty. Mol. Med. Rep. 14:4 (2016), 3184–3198.
    • (2016) Mol. Med. Rep. , vol.14 , Issue.4 , pp. 3184-3198
    • Ruan, Q.1
  • 25
    • 16044373524 scopus 로고    scopus 로고
    • Secreted amyloid beta-protein similar to that in the senile plaques of Alzheimer's disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer's disease
    • Scheuner, D. et al., Secreted amyloid beta-protein similar to that in the senile plaques of Alzheimer's disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer's disease. Nat. Med. 2:8 (1996), 864–870.
    • (1996) Nat. Med. , vol.2 , Issue.8 , pp. 864-870
    • Scheuner, D.1
  • 26
    • 85018724643 scopus 로고    scopus 로고
    • Mouse models of Alzheimer's disease
    • Esquerda-Canals, G. et al., Mouse models of Alzheimer's disease. J. Alzheimers Dis. 57:4 (2017), 1171–1183.
    • (2017) J. Alzheimers Dis. , vol.57 , Issue.4 , pp. 1171-1183
    • Esquerda-Canals, G.1
  • 27
    • 0027327267 scopus 로고
    • Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer's disease
    • Saunders, A.M. et al., Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer's disease. Neurology 43:8 (1993), 1467–1472.
    • (1993) Neurology , vol.43 , Issue.8 , pp. 1467-1472
    • Saunders, A.M.1
  • 28
    • 0033966340 scopus 로고    scopus 로고
    • Increased plasma levels of interleukin-1, interleukin-6 and alpha-1-antichymotrypsin in patients with Alzheimer's disease: peripheral inflammation or signals from the brain?
    • Licastro, F. et al., Increased plasma levels of interleukin-1, interleukin-6 and alpha-1-antichymotrypsin in patients with Alzheimer's disease: peripheral inflammation or signals from the brain?. J. Neuroimmunol. 103:1 (2000), 97–102.
    • (2000) J. Neuroimmunol. , vol.103 , Issue.1 , pp. 97-102
    • Licastro, F.1
  • 29
    • 84898447902 scopus 로고    scopus 로고
    • Plasma phospholipids identify antecedent memory impairment in older adults
    • Mapstone, M. et al., Plasma phospholipids identify antecedent memory impairment in older adults. Nat. Med. 20:4 (2014), 415–418.
    • (2014) Nat. Med. , vol.20 , Issue.4 , pp. 415-418
    • Mapstone, M.1
  • 30
    • 84860460906 scopus 로고    scopus 로고
    • Mouse models of Alzheimer's disease
    • Hall, A.M., Roberson, E.D., Mouse models of Alzheimer's disease. Brain Res. Bull. 88:1 (2012), 3–12.
    • (2012) Brain Res. Bull. , vol.88 , Issue.1 , pp. 3-12
    • Hall, A.M.1    Roberson, E.D.2
  • 31
    • 33748331308 scopus 로고    scopus 로고
    • The mighty mouse: genetically engineered mouse models in cancer drug development
    • Sharpless, N.E., Depinho, R.A., The mighty mouse: genetically engineered mouse models in cancer drug development. Nat. Rev. Drug Discov. 5:9 (2006), 741–754.
    • (2006) Nat. Rev. Drug Discov. , vol.5 , Issue.9 , pp. 741-754
    • Sharpless, N.E.1    Depinho, R.A.2
  • 32
    • 84880187096 scopus 로고    scopus 로고
    • Mice as models: transgenic approaches and Alzheimer's disease
    • Games, D. et al., Mice as models: transgenic approaches and Alzheimer's disease. J. Alzheimers Dis. 9:3 Suppl (2006), 133–149.
    • (2006) J. Alzheimers Dis. , vol.9 , Issue.3 , pp. 133-149
    • Games, D.1
  • 33
    • 16944362157 scopus 로고    scopus 로고
    • Mutant presenilins of Alzheimer's disease increase production of 42-residue amyloid beta-protein in both transfected cells and transgenic mice
    • Citron, M. et al., Mutant presenilins of Alzheimer's disease increase production of 42-residue amyloid beta-protein in both transfected cells and transgenic mice. Nat. Med. 3:1 (1997), 67–72.
    • (1997) Nat. Med. , vol.3 , Issue.1 , pp. 67-72
    • Citron, M.1
  • 34
    • 0031914718 scopus 로고    scopus 로고
    • Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes
    • Holcomb, L. et al., Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes. Nat. Med. 4:1 (1998), 97–100.
    • (1998) Nat. Med. , vol.4 , Issue.1 , pp. 97-100
    • Holcomb, L.1
  • 35
    • 84863939887 scopus 로고    scopus 로고
    • Presenilins and gamma-secretase: structure, function, and role in Alzheimer disease
    • De Strooper, B., Iwatsubo, T., Wolfe, M.S., Presenilins and gamma-secretase: structure, function, and role in Alzheimer disease. Cold Spring Harb. Perspect. Med., 2(1), 2012, a006304.
    • (2012) Cold Spring Harb. Perspect. Med. , vol.2 , Issue.1 , pp. a006304
    • De Strooper, B.1    Iwatsubo, T.2    Wolfe, M.S.3
  • 36
    • 0028965635 scopus 로고
    • Somatodendritic localization and hyperphosphorylation of tau protein in transgenic mice expressing the longest human brain tau isoform
    • Gotz, J. et al., Somatodendritic localization and hyperphosphorylation of tau protein in transgenic mice expressing the longest human brain tau isoform. EMBO J. 14:7 (1995), 1304–1313.
    • (1995) EMBO J. , vol.14 , Issue.7 , pp. 1304-1313
    • Gotz, J.1
  • 37
    • 0034426011 scopus 로고    scopus 로고
    • Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein
    • Lewis, J. et al., Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat. Genet. 25:4 (2000), 402–405.
    • (2000) Nat. Genet. , vol.25 , Issue.4 , pp. 402-405
    • Lewis, J.1
  • 38
    • 0035943436 scopus 로고    scopus 로고
    • Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils
    • Gotz, J. et al., Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science 293:5534 (2001), 1491–1495.
    • (2001) Science , vol.293 , Issue.5534 , pp. 1491-1495
    • Gotz, J.1
  • 39
    • 33846538660 scopus 로고    scopus 로고
    • Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model
    • Yoshiyama, Y. et al., Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 53:3 (2007), 337–351.
    • (2007) Neuron , vol.53 , Issue.3 , pp. 337-351
    • Yoshiyama, Y.1
  • 40
    • 0042697305 scopus 로고    scopus 로고
    • Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction
    • Oddo, S. et al., Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron 39:3 (2003), 409–421.
    • (2003) Neuron , vol.39 , Issue.3 , pp. 409-421
    • Oddo, S.1
  • 41
    • 4243215848 scopus 로고    scopus 로고
    • Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer's disease
    • Holtzman, D.M. et al., Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer's disease. Proc. Natl. Acad. Sci. U. S. A. 97:6 (2000), 2892–2897.
    • (2000) Proc. Natl. Acad. Sci. U. S. A. , vol.97 , Issue.6 , pp. 2892-2897
    • Holtzman, D.M.1
  • 42
    • 0033593027 scopus 로고    scopus 로고
    • Apolipoprotein E is essential for amyloid deposition in the APP(V717F) transgenic mouse model of Alzheimer's disease
    • Bales, K.R. et al., Apolipoprotein E is essential for amyloid deposition in the APP(V717F) transgenic mouse model of Alzheimer's disease. Proc. Natl. Acad. Sci. U. S. A. 96:26 (1999), 15233–15238.
    • (1999) Proc. Natl. Acad. Sci. U. S. A. , vol.96 , Issue.26 , pp. 15233-15238
    • Bales, K.R.1
  • 43
    • 33645300736 scopus 로고    scopus 로고
    • The prolyl isomerase Pin1 regulates amyloid precursor protein processing and amyloid-beta production
    • Pastorino, L. et al., The prolyl isomerase Pin1 regulates amyloid precursor protein processing and amyloid-beta production. Nature 440:7083 (2006), 528–534.
    • (2006) Nature , vol.440 , Issue.7083 , pp. 528-534
    • Pastorino, L.1
  • 44
    • 85012907718 scopus 로고    scopus 로고
    • Mitophagy and Alzheimer's disease: cellular and molecular mechanisms
    • Kerr, J.S. et al., Mitophagy and Alzheimer's disease: cellular and molecular mechanisms. Trends Neurosci. 40:3 (2017), 151–166.
    • (2017) Trends Neurosci. , vol.40 , Issue.3 , pp. 151-166
    • Kerr, J.S.1
  • 45
    • 85014799386 scopus 로고    scopus 로고
    • Autophagy and neurodegeneration: pathogenic mechanisms and therapeutic opportunities
    • Menzies, F.M. et al., Autophagy and neurodegeneration: pathogenic mechanisms and therapeutic opportunities. Neuron 93:5 (2017), 1015–1034.
    • (2017) Neuron , vol.93 , Issue.5 , pp. 1015-1034
    • Menzies, F.M.1
  • 46
    • 85011263985 scopus 로고    scopus 로고
    • Autophagy regulates MAVS signaling activation in a phosphorylation-dependent manner in microglia
    • Cheng, J. et al., Autophagy regulates MAVS signaling activation in a phosphorylation-dependent manner in microglia. Cell Death Differ. 24:2 (2017), 276–287.
    • (2017) Cell Death Differ. , vol.24 , Issue.2 , pp. 276-287
    • Cheng, J.1
  • 47
    • 79551598347 scopus 로고    scopus 로고
    • AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1
    • Kim, J. et al., AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 13:2 (2011), 132–141.
    • (2011) Nat. Cell Biol. , vol.13 , Issue.2 , pp. 132-141
    • Kim, J.1
  • 48
    • 85015157838 scopus 로고    scopus 로고
    • Autophagy and microglia: novel partners in neurodegeneration and aging
    • Plaza-Zabala, A., Sierra-Torre, V., Sierra, A., Autophagy and microglia: novel partners in neurodegeneration and aging. Int. J. Mol. Sci., 2017, 18(3).
    • (2017) Int. J. Mol. Sci. , pp. 18(3)
    • Plaza-Zabala, A.1    Sierra-Torre, V.2    Sierra, A.3
  • 49
    • 85014348031 scopus 로고    scopus 로고
    • The autophagy machinery: a new player in chemotactic cell migration
    • Coly, P.M. et al., The autophagy machinery: a new player in chemotactic cell migration. Front. Neurosci., 11, 2017, 78.
    • (2017) Front. Neurosci. , vol.11 , pp. 78
    • Coly, P.M.1
  • 52
    • 85013932885 scopus 로고    scopus 로고
    • Autophagy and Alzheimer's disease
    • Li, Q., Liu, Y., Sun, M., Autophagy and Alzheimer's disease. Cell. Mol. Neurobiol. 37:3 (2017), 377–388.
    • (2017) Cell. Mol. Neurobiol. , vol.37 , Issue.3 , pp. 377-388
    • Li, Q.1    Liu, Y.2    Sun, M.3
  • 53
    • 84938257029 scopus 로고    scopus 로고
    • Autophagy in Alzheimer's disease
    • Zare-Shahabadi, A. et al., Autophagy in Alzheimer's disease. Rev. Neurosci. 26:4 (2015), 385–395.
    • (2015) Rev. Neurosci. , vol.26 , Issue.4 , pp. 385-395
    • Zare-Shahabadi, A.1
  • 54
    • 79957917512 scopus 로고    scopus 로고
    • A small-molecule enhancer of autophagy decreases levels of Abeta and APP-CTF via Atg5-dependent autophagy pathway
    • Tian, Y. et al., A small-molecule enhancer of autophagy decreases levels of Abeta and APP-CTF via Atg5-dependent autophagy pathway. FASEB J. 25:6 (2011), 1934–1942.
    • (2011) FASEB J. , vol.25 , Issue.6 , pp. 1934-1942
    • Tian, Y.1
  • 55
    • 79251556232 scopus 로고    scopus 로고
    • Novel synthetic small-molecule activators of AMPK as enhancers of autophagy and amyloid-beta peptide degradation
    • Vingtdeux, V. et al., Novel synthetic small-molecule activators of AMPK as enhancers of autophagy and amyloid-beta peptide degradation. FASEB J. 25:1 (2011), 219–231.
    • (2011) FASEB J. , vol.25 , Issue.1 , pp. 219-231
    • Vingtdeux, V.1
  • 56
    • 26444461505 scopus 로고    scopus 로고
    • A(beta) generation in autophagic vacuoles
    • Mizushima, N., A(beta) generation in autophagic vacuoles. J. Cell Biol. 171:1 (2005), 15–17.
    • (2005) J. Cell Biol. , vol.171 , Issue.1 , pp. 15-17
    • Mizushima, N.1
  • 57
    • 4344689871 scopus 로고    scopus 로고
    • Autophagic vacuoles are enriched in amyloid precursor protein-secretase activities: implications for beta-amyloid peptide over-production and localization in Alzheimer's disease
    • Yu, W.H. et al., Autophagic vacuoles are enriched in amyloid precursor protein-secretase activities: implications for beta-amyloid peptide over-production and localization in Alzheimer's disease. Int. J. Biochem. Cell Biol. 36:12 (2004), 2531–2540.
    • (2004) Int. J. Biochem. Cell Biol. , vol.36 , Issue.12 , pp. 2531-2540
    • Yu, W.H.1
  • 58
    • 51349125291 scopus 로고    scopus 로고
    • Neuronal apoptosis and autophagy cross talk in aging PS/APP mice, a model of Alzheimer's disease
    • Yang, D.S. et al., Neuronal apoptosis and autophagy cross talk in aging PS/APP mice, a model of Alzheimer's disease. Am. J. Pathol. 173:3 (2008), 665–681.
    • (2008) Am. J. Pathol. , vol.173 , Issue.3 , pp. 665-681
    • Yang, D.S.1
  • 59
    • 49049096562 scopus 로고    scopus 로고
    • Autophagy induction and autophagosome clearance in neurons: relationship to autophagic pathology in Alzheimer's disease
    • Boland, B. et al., Autophagy induction and autophagosome clearance in neurons: relationship to autophagic pathology in Alzheimer's disease. J. Neurosci. 28:27 (2008), 6926–6937.
    • (2008) J. Neurosci. , vol.28 , Issue.27 , pp. 6926-6937
    • Boland, B.1
  • 60
    • 84937874784 scopus 로고    scopus 로고
    • The role of PICALM in Alzheimer's disease
    • Xu, W., Tan, L., Yu, J.T., The role of PICALM in Alzheimer's disease. Mol. Neurobiol. 52:1 (2015), 399–413.
    • (2015) Mol. Neurobiol. , vol.52 , Issue.1 , pp. 399-413
    • Xu, W.1    Tan, L.2    Yu, J.T.3
  • 61
    • 84923350735 scopus 로고    scopus 로고
    • PICALM modulates autophagy activity and tau accumulation
    • Moreau, K. et al., PICALM modulates autophagy activity and tau accumulation. Nat. Commun., 5, 2014, 4998.
    • (2014) Nat. Commun. , vol.5 , pp. 4998
    • Moreau, K.1
  • 62
    • 45749114895 scopus 로고    scopus 로고
    • The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice
    • Pickford, F. et al., The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J. Clin. Invest. 118:6 (2008), 2190–2199.
    • (2008) J. Clin. Invest. , vol.118 , Issue.6 , pp. 2190-2199
    • Pickford, F.1
  • 63
    • 0342614209 scopus 로고    scopus 로고
    • Unified nomenclature for the winged helix/forkhead transcription factors
    • Kaestner, K.H., Knochel, W., Martinez, D.E., Unified nomenclature for the winged helix/forkhead transcription factors. Genes Dev. 14:2 (2000), 142–146.
    • (2000) Genes Dev. , vol.14 , Issue.2 , pp. 142-146
    • Kaestner, K.H.1    Knochel, W.2    Martinez, D.E.3
  • 64
    • 33846295218 scopus 로고    scopus 로고
    • FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis
    • Paik, J.H. et al., FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis. Cell 128:2 (2007), 309–323.
    • (2007) Cell , vol.128 , Issue.2 , pp. 309-323
    • Paik, J.H.1
  • 65
    • 84958765306 scopus 로고    scopus 로고
    • FOXO transcription factors in cancer development and therapy
    • Coomans de Brachene, A., Demoulin, J.B., FOXO transcription factors in cancer development and therapy. Cell. Mol. Life Sci. 73:6 (2016), 1159–1172.
    • (2016) Cell. Mol. Life Sci. , vol.73 , Issue.6 , pp. 1159-1172
    • Coomans de Brachene, A.1    Demoulin, J.B.2
  • 66
    • 11144357466 scopus 로고    scopus 로고
    • IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a
    • Hu, M.C. et al., IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a. Cell 117:2 (2004), 225–237.
    • (2004) Cell , vol.117 , Issue.2 , pp. 225-237
    • Hu, M.C.1
  • 67
    • 2442440607 scopus 로고    scopus 로고
    • FoxO: linking new signaling pathways
    • Arden, K.C., FoxO: linking new signaling pathways. Mol. Cell 14:4 (2004), 416–418.
    • (2004) Mol. Cell , vol.14 , Issue.4 , pp. 416-418
    • Arden, K.C.1
  • 68
    • 13244287966 scopus 로고    scopus 로고
    • Regulation of FoxO activity by CBP/p300-mediated acetylation
    • van der Heide, L.P., Smidt, M.P., Regulation of FoxO activity by CBP/p300-mediated acetylation. Trends Biochem. Sci. 30:2 (2005), 81–86.
    • (2005) Trends Biochem. Sci. , vol.30 , Issue.2 , pp. 81-86
    • van der Heide, L.P.1    Smidt, M.P.2
  • 69
    • 1342264308 scopus 로고    scopus 로고
    • Mammalian SIRT1 represses forkhead transcription factors
    • Motta, M.C. et al., Mammalian SIRT1 represses forkhead transcription factors. Cell 116:4 (2004), 551–563.
    • (2004) Cell , vol.116 , Issue.4 , pp. 551-563
    • Motta, M.C.1
  • 70
    • 84903769379 scopus 로고    scopus 로고
    • Prolyl hydroxylation by EglN2 destabilizes FOXO3a by blocking its interaction with the USP9x deubiquitinase
    • Zheng, X. et al., Prolyl hydroxylation by EglN2 destabilizes FOXO3a by blocking its interaction with the USP9x deubiquitinase. Genes Dev. 28:13 (2014), 1429–1444.
    • (2014) Genes Dev. , vol.28 , Issue.13 , pp. 1429-1444
    • Zheng, X.1
  • 71
    • 13444252337 scopus 로고    scopus 로고
    • Skp2 inhibits FOXO1 in tumor suppression through ubiquitin-mediated degradation
    • Huang, H. et al., Skp2 inhibits FOXO1 in tumor suppression through ubiquitin-mediated degradation. Proc. Natl. Acad. Sci. U. S. A. 102:5 (2005), 1649–1654.
    • (2005) Proc. Natl. Acad. Sci. U. S. A. , vol.102 , Issue.5 , pp. 1649-1654
    • Huang, H.1
  • 72
    • 53949087832 scopus 로고    scopus 로고
    • Arginine methylation of FOXO transcription factors inhibits their phosphorylation by Akt
    • Yamagata, K. et al., Arginine methylation of FOXO transcription factors inhibits their phosphorylation by Akt. Mol. Cell 32:2 (2008), 221–231.
    • (2008) Mol. Cell , vol.32 , Issue.2 , pp. 221-231
    • Yamagata, K.1
  • 73
    • 0037466652 scopus 로고    scopus 로고
    • DAF-16 target genes that control C. elegans life-span and metabolism
    • Lee, S.S. et al., DAF-16 target genes that control C. elegans life-span and metabolism. Science 300:5619 (2003), 644–647.
    • (2003) Science , vol.300 , Issue.5619 , pp. 644-647
    • Lee, S.S.1
  • 74
    • 0030713963 scopus 로고    scopus 로고
    • Genetics of aging
    • Finch, C.E., Tanzi, R.E., Genetics of aging. Science 278:5337 (1997), 407–411.
    • (1997) Science , vol.278 , Issue.5337 , pp. 407-411
    • Finch, C.E.1    Tanzi, R.E.2
  • 75
    • 41549135942 scopus 로고    scopus 로고
    • FoxO transcription factors in the maintenance of cellular homeostasis during aging
    • Salih, D.A., Brunet, A., FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr. Opin. Cell Biol. 20:2 (2008), 126–136.
    • (2008) Curr. Opin. Cell Biol. , vol.20 , Issue.2 , pp. 126-136
    • Salih, D.A.1    Brunet, A.2
  • 76
    • 79951720176 scopus 로고    scopus 로고
    • JNK regulates FoxO-dependent autophagy in neurons
    • Xu, P. et al., JNK regulates FoxO-dependent autophagy in neurons. Genes Dev. 25:4 (2011), 310–322.
    • (2011) Genes Dev. , vol.25 , Issue.4 , pp. 310-322
    • Xu, P.1
  • 77
    • 84872899284 scopus 로고    scopus 로고
    • FOXOs: signalling integrators for homeostasis maintenance
    • Eijkelenboom, A., Burgering, B.M., FOXOs: signalling integrators for homeostasis maintenance. Nat. Rev. Mol. Cell Biol. 14:2 (2013), 83–97.
    • (2013) Nat. Rev. Mol. Cell Biol. , vol.14 , Issue.2 , pp. 83-97
    • Eijkelenboom, A.1    Burgering, B.M.2
  • 78
    • 65249108225 scopus 로고    scopus 로고
    • FOXO3a-dependent regulation of Pink1 (Park6) mediates survival signaling in response to cytokine deprivation
    • Mei, Y. et al., FOXO3a-dependent regulation of Pink1 (Park6) mediates survival signaling in response to cytokine deprivation. Proc. Natl. Acad. Sci. U. S. A. 106:13 (2009), 5153–5158.
    • (2009) Proc. Natl. Acad. Sci. U. S. A. , vol.106 , Issue.13 , pp. 5153-5158
    • Mei, Y.1
  • 79
    • 0042477712 scopus 로고    scopus 로고
    • FOXO transcription factors directly activate bim gene expression and promote apoptosis in sympathetic neurons
    • Gilley, J., Coffer, P.J., Ham, J., FOXO transcription factors directly activate bim gene expression and promote apoptosis in sympathetic neurons. J. Cell Biol. 162:4 (2003), 613–622.
    • (2003) J. Cell Biol. , vol.162 , Issue.4 , pp. 613-622
    • Gilley, J.1    Coffer, P.J.2    Ham, J.3
  • 80
    • 85016052515 scopus 로고    scopus 로고
    • Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging
    • (e16)
    • Baar, M.P. et al., Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell 169:1 (2017), 132–147 (e16).
    • (2017) Cell , vol.169 , Issue.1 , pp. 132-147
    • Baar, M.P.1
  • 81
    • 0037136563 scopus 로고    scopus 로고
    • Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress
    • Kops, G.J. et al., Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature 419:6904 (2002), 316–321.
    • (2002) Nature , vol.419 , Issue.6904 , pp. 316-321
    • Kops, G.J.1
  • 82
    • 80052490840 scopus 로고    scopus 로고
    • FOXO1 is an essential regulator of pluripotency in human embryonic stem cells
    • Zhang, X. et al., FOXO1 is an essential regulator of pluripotency in human embryonic stem cells. Nat. Cell Biol. 13:9 (2011), 1092–1099.
    • (2011) Nat. Cell Biol. , vol.13 , Issue.9 , pp. 1092-1099
    • Zhang, X.1
  • 83
    • 85017102460 scopus 로고    scopus 로고
    • mTOR signaling in growth, metabolism, and disease
    • Saxton, R.A., Sabatini, D.M., mTOR signaling in growth, metabolism, and disease. Cell 169:2 (2017), 361–371.
    • (2017) Cell , vol.169 , Issue.2 , pp. 361-371
    • Saxton, R.A.1    Sabatini, D.M.2
  • 84
    • 84878532557 scopus 로고    scopus 로고
    • Signal integration by mTORC1 coordinates nutrient input with biosynthetic output
    • Dibble, C.C., Manning, B.D., Signal integration by mTORC1 coordinates nutrient input with biosynthetic output. Nat. Cell Biol. 15:6 (2013), 555–564.
    • (2013) Nat. Cell Biol. , vol.15 , Issue.6 , pp. 555-564
    • Dibble, C.C.1    Manning, B.D.2
  • 85
    • 70350418625 scopus 로고    scopus 로고
    • mTOR signaling at a glance
    • Laplante, M., Sabatini, D.M., mTOR signaling at a glance. J. Cell Sci. 122:Pt 20 (2009), 3589–3594.
    • (2009) J. Cell Sci. , vol.122 , pp. 3589-3594
    • Laplante, M.1    Sabatini, D.M.2
  • 86
  • 87
    • 84862179773 scopus 로고    scopus 로고
    • Mammalian target of rapamycin signaling in diabetic cardiovascular disease
    • Chong, Z.Z., Maiese, K., Mammalian target of rapamycin signaling in diabetic cardiovascular disease. Cardiovasc. Diabetol., 11, 2012, 45.
    • (2012) Cardiovasc. Diabetol. , vol.11 , pp. 45
    • Chong, Z.Z.1    Maiese, K.2
  • 88
    • 49749096430 scopus 로고    scopus 로고
    • Small molecule enhancers of autophagy for neurodegenerative diseases
    • Sarkar, S., Rubinsztein, D.C., Small molecule enhancers of autophagy for neurodegenerative diseases. Mol. BioSyst. 4:9 (2008), 895–901.
    • (2008) Mol. BioSyst. , vol.4 , Issue.9 , pp. 895-901
    • Sarkar, S.1    Rubinsztein, D.C.2
  • 89
    • 0032999830 scopus 로고    scopus 로고
    • Target of rapamycin (TOR): balancing the opposing forces of protein synthesis and degradation
    • Dennis, P.B., Fumagalli, S., Thomas, G., Target of rapamycin (TOR): balancing the opposing forces of protein synthesis and degradation. Curr. Opin. Genet. Dev. 9:1 (1999), 49–54.
    • (1999) Curr. Opin. Genet. Dev. , vol.9 , Issue.1 , pp. 49-54
    • Dennis, P.B.1    Fumagalli, S.2    Thomas, G.3
  • 90
    • 0642367846 scopus 로고    scopus 로고
    • Genetics: influence of TOR kinase on lifespan in C. elegans
    • Vellai, T. et al., Genetics: influence of TOR kinase on lifespan in C. elegans. Nature, 426(6967), 2003, 620.
    • (2003) Nature , vol.426 , Issue.6967 , pp. 620
    • Vellai, T.1
  • 91
    • 70349669095 scopus 로고    scopus 로고
    • Ribosomal protein S6 kinase 1 signaling regulates mammalian life span
    • Selman, C. et al., Ribosomal protein S6 kinase 1 signaling regulates mammalian life span. Science 326:5949 (2009), 140–144.
    • (2009) Science , vol.326 , Issue.5949 , pp. 140-144
    • Selman, C.1
  • 92
    • 30944458446 scopus 로고    scopus 로고
    • Extension of chronological life span in yeast by decreased TOR pathway signaling
    • Powers, R.W. 3rd et al., Extension of chronological life span in yeast by decreased TOR pathway signaling. Genes Dev. 20:2 (2006), 174–184.
    • (2006) Genes Dev. , vol.20 , Issue.2 , pp. 174-184
    • Powers, R.W.1
  • 93
    • 84860461929 scopus 로고    scopus 로고
    • TOR signaling and rapamycin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO
    • Robida-Stubbs, S. et al., TOR signaling and rapamycin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO. Cell Metab. 15:5 (2012), 713–724.
    • (2012) Cell Metab. , vol.15 , Issue.5 , pp. 713-724
    • Robida-Stubbs, S.1
  • 94
    • 72649091698 scopus 로고    scopus 로고
    • Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster
    • Bjedov, I. et al., Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster. Cell Metab. 11:1 (2010), 35–46.
    • (2010) Cell Metab. , vol.11 , Issue.1 , pp. 35-46
    • Bjedov, I.1
  • 95
    • 67650944993 scopus 로고    scopus 로고
    • Rapamycin fed late in life extends lifespan in genetically heterogeneous mice
    • Harrison, D.E. et al., Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460:7253 (2009), 392–395.
    • (2009) Nature , vol.460 , Issue.7253 , pp. 392-395
    • Harrison, D.E.1
  • 96
    • 84901403995 scopus 로고    scopus 로고
    • Rapamycinmediated lifespan increase in mice is dose and sex dependent and metabolically distinct from dietary restriction
    • Jun
    • Miller, R.A. et al., Rapamycinmediated lifespan increase in mice is dose and sex dependent and metabolically distinct from dietary restriction. Aging Cell 13:3 (2014 Jun), 468–477.
    • (2014) Aging Cell , vol.13 , Issue.3 , pp. 468-477
    • Miller, R.A.1
  • 97
    • 83755165458 scopus 로고    scopus 로고
    • Rapamycin increases lifespan and inhibits spontaneous tumorigenesis in inbred female mice
    • Anisimov, V.N. et al., Rapamycin increases lifespan and inhibits spontaneous tumorigenesis in inbred female mice. Cell Cycle 10:24 (2011), 4230–4236.
    • (2011) Cell Cycle , vol.10 , Issue.24 , pp. 4230-4236
    • Anisimov, V.N.1
  • 98
    • 79951794971 scopus 로고    scopus 로고
    • Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice
    • Miller, R.A. et al., Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice. J. Gerontol. A Biol. Sci. Med. Sci. 66:2 (2011), 191–201.
    • (2011) J. Gerontol. A Biol. Sci. Med. Sci. , vol.66 , Issue.2 , pp. 191-201
    • Miller, R.A.1
  • 99
    • 0027058857 scopus 로고
    • Modulation of the dynamic instability of tubulin assembly by the microtubule-associated protein tau
    • Drechsel, D.N. et al., Modulation of the dynamic instability of tubulin assembly by the microtubule-associated protein tau. Mol. Biol. Cell 3:10 (1992), 1141–1154.
    • (1992) Mol. Biol. Cell , vol.3 , Issue.10 , pp. 1141-1154
    • Drechsel, D.N.1
  • 100
    • 84877803859 scopus 로고    scopus 로고
    • mTOR regulates tau phosphorylation and degradation: implications for Alzheimer's disease and other tauopathies
    • Caccamo, A. et al., mTOR regulates tau phosphorylation and degradation: implications for Alzheimer's disease and other tauopathies. Aging Cell 12:3 (2013), 370–380.
    • (2013) Aging Cell , vol.12 , Issue.3 , pp. 370-380
    • Caccamo, A.1
  • 101
    • 84897401611 scopus 로고    scopus 로고
    • Protein phosphatase 2A dysfunction in Alzheimer's disease
    • Sontag, J.M., Sontag, E., Protein phosphatase 2A dysfunction in Alzheimer's disease. Front. Mol. Neurosci., 7, 2014, 16.
    • (2014) Front. Mol. Neurosci. , vol.7 , pp. 16
    • Sontag, J.M.1    Sontag, E.2
  • 102
    • 78650746798 scopus 로고    scopus 로고
    • Biguanide metformin acts on tau phosphorylation via mTOR/protein phosphatase 2A (PP2A) signaling
    • Kickstein, E. et al., Biguanide metformin acts on tau phosphorylation via mTOR/protein phosphatase 2A (PP2A) signaling. Proc. Natl. Acad. Sci. U. S. A. 107:50 (2010), 21830–21835.
    • (2010) Proc. Natl. Acad. Sci. U. S. A. , vol.107 , Issue.50 , pp. 21830-21835
    • Kickstein, E.1
  • 103
    • 29344460850 scopus 로고    scopus 로고
    • P70 S6 kinase mediates tau phosphorylation and synthesis
    • Pei, J.J. et al., P70 S6 kinase mediates tau phosphorylation and synthesis. FEBS Lett. 580:1 (2006), 107–114.
    • (2006) FEBS Lett. , vol.580 , Issue.1 , pp. 107-114
    • Pei, J.J.1
  • 104
    • 33847199335 scopus 로고    scopus 로고
    • eIF4E function in somatic cells modulates ageing in Caenorhabditis elegans
    • Syntichaki, P., Troulinaki, K., Tavernarakis, N., eIF4E function in somatic cells modulates ageing in Caenorhabditis elegans. Nature 445:7130 (2007), 922–926.
    • (2007) Nature , vol.445 , Issue.7130 , pp. 922-926
    • Syntichaki, P.1    Troulinaki, K.2    Tavernarakis, N.3
  • 105
    • 21344459656 scopus 로고    scopus 로고
    • mTOR/p70S6k signalling alteration by Abeta exposure as well as in APP-PS1 transgenic models and in patients with Alzheimer's disease
    • Lafay-Chebassier, C. et al., mTOR/p70S6k signalling alteration by Abeta exposure as well as in APP-PS1 transgenic models and in patients with Alzheimer's disease. J. Neurochem. 94:1 (2005), 215–225.
    • (2005) J. Neurochem. , vol.94 , Issue.1 , pp. 215-225
    • Lafay-Chebassier, C.1
  • 106
    • 80053243942 scopus 로고    scopus 로고
    • Inducing autophagy by rapamycin before, but not after, the formation of plaques and tangles ameliorates cognitive deficits
    • Majumder, S. et al., Inducing autophagy by rapamycin before, but not after, the formation of plaques and tangles ameliorates cognitive deficits. PLoS One, 6(9), 2011, e25416.
    • (2011) PLoS One , vol.6 , Issue.9
    • Majumder, S.1
  • 107
    • 77951227122 scopus 로고    scopus 로고
    • Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: effects on cognitive impairments
    • Caccamo, A. et al., Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: effects on cognitive impairments. J. Biol. Chem. 285:17 (2010), 13107–13120.
    • (2010) J. Biol. Chem. , vol.285 , Issue.17 , pp. 13107-13120
    • Caccamo, A.1
  • 108
    • 77956305343 scopus 로고    scopus 로고
    • Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer's disease
    • Spilman, P. et al., Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer's disease. PLoS One, 5(4), 2010, e9979.
    • (2010) PLoS One , vol.5 , Issue.4
    • Spilman, P.1
  • 109
    • 85011299376 scopus 로고    scopus 로고
    • MAPK pathway in melanoma part II-secondary and adaptive resistance mechanisms to BRAF inhibition
    • Amaral, T. et al., MAPK pathway in melanoma part II-secondary and adaptive resistance mechanisms to BRAF inhibition. Eur. J. Cancer 73 (2017), 93–101.
    • (2017) Eur. J. Cancer , vol.73 , pp. 93-101
    • Amaral, T.1
  • 110
    • 85012201477 scopus 로고    scopus 로고
    • Spatial and temporal signal processing and decision making by MAPK pathways
    • Atay, O., Skotheim, J.M., Spatial and temporal signal processing and decision making by MAPK pathways. J. Cell Biol. 216:2 (2017), 317–330.
    • (2017) J. Cell Biol. , vol.216 , Issue.2 , pp. 317-330
    • Atay, O.1    Skotheim, J.M.2
  • 111
    • 84930672166 scopus 로고    scopus 로고
    • Compromised MAPK signaling in human diseases: an update
    • Kim, E.K., Choi, E.J., Compromised MAPK signaling in human diseases: an update. Arch. Toxicol. 89:6 (2015), 867–882.
    • (2015) Arch. Toxicol. , vol.89 , Issue.6 , pp. 867-882
    • Kim, E.K.1    Choi, E.J.2
  • 112
    • 84962132644 scopus 로고    scopus 로고
    • The mitogen-activated protein kinase (MAPK) pathway: role in immune evasion by trypanosomatids
    • Soares-Silva, M. et al., The mitogen-activated protein kinase (MAPK) pathway: role in immune evasion by trypanosomatids. Front. Microbiol., 7, 2016, 183.
    • (2016) Front. Microbiol. , vol.7 , pp. 183
    • Soares-Silva, M.1
  • 113
    • 34248583886 scopus 로고    scopus 로고
    • MAP kinase signalling pathways in cancer
    • Dhillon, A.S. et al., MAP kinase signalling pathways in cancer. Oncogene 26:22 (2007), 3279–3290.
    • (2007) Oncogene , vol.26 , Issue.22 , pp. 3279-3290
    • Dhillon, A.S.1
  • 114
    • 84861147473 scopus 로고    scopus 로고
    • A comprehensive survey of Ras mutations in cancer
    • Prior, I.A., Lewis, P.D., Mattos, C., A comprehensive survey of Ras mutations in cancer. Cancer Res. 72:10 (2012), 2457–2467.
    • (2012) Cancer Res. , vol.72 , Issue.10 , pp. 2457-2467
    • Prior, I.A.1    Lewis, P.D.2    Mattos, C.3
  • 115
    • 33745913034 scopus 로고    scopus 로고
    • EGFR mutations in small-cell lung cancers in patients who have never smoked
    • Zakowski, M.F. et al., EGFR mutations in small-cell lung cancers in patients who have never smoked. N. Engl. J. Med. 355:2 (2006), 213–215.
    • (2006) N. Engl. J. Med. , vol.355 , Issue.2 , pp. 213-215
    • Zakowski, M.F.1
  • 116
    • 27744484054 scopus 로고    scopus 로고
    • The natural history of EGFR and EGFRvIII in glioblastoma patients
    • Heimberger, A.B. et al., The natural history of EGFR and EGFRvIII in glioblastoma patients. J. Transl. Med., 3, 2005, 38.
    • (2005) J. Transl. Med. , vol.3 , pp. 38
    • Heimberger, A.B.1
  • 117
    • 18444374405 scopus 로고    scopus 로고
    • Mutations of the BRAF gene in human cancer
    • Davies, H. et al., Mutations of the BRAF gene in human cancer. Nature 417:6892 (2002), 949–954.
    • (2002) Nature , vol.417 , Issue.6892 , pp. 949-954
    • Davies, H.1
  • 118
    • 56349145099 scopus 로고    scopus 로고
    • Oxidative stress hypothesis in Alzheimer's disease: a reappraisal
    • Pratico, D., Oxidative stress hypothesis in Alzheimer's disease: a reappraisal. Trends Pharmacol. Sci. 29:12 (2008), 609–615.
    • (2008) Trends Pharmacol. Sci. , vol.29 , Issue.12 , pp. 609-615
    • Pratico, D.1
  • 119
    • 84904093190 scopus 로고    scopus 로고
    • Abeta and tau toxicities in Alzheimer's are linked via oxidative stress-induced p38 activation: protective role of vitamin E
    • Giraldo, E. et al., Abeta and tau toxicities in Alzheimer's are linked via oxidative stress-induced p38 activation: protective role of vitamin E. Redox Biol. 2 (2014), 873–877.
    • (2014) Redox Biol. , vol.2 , pp. 873-877
    • Giraldo, E.1
  • 120
    • 80052843006 scopus 로고    scopus 로고
    • The role of CNI-1493 in the function of primary microglia with respect to amyloid-beta
    • Bach, J.P. et al., The role of CNI-1493 in the function of primary microglia with respect to amyloid-beta. J. Alzheimers Dis. 26:1 (2011), 69–80.
    • (2011) J. Alzheimers Dis. , vol.26 , Issue.1 , pp. 69-80
    • Bach, J.P.1
  • 121
    • 83755183781 scopus 로고    scopus 로고
    • Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and restores neuronal beta-catenin pathway function in an Alzheimer's disease model
    • Kitazawa, M. et al., Blocking IL-1 signaling rescues cognition, attenuates tau pathology, and restores neuronal beta-catenin pathway function in an Alzheimer's disease model. J. Immunol. 187:12 (2011), 6539–6549.
    • (2011) J. Immunol. , vol.187 , Issue.12 , pp. 6539-6549
    • Kitazawa, M.1
  • 122
    • 84890247564 scopus 로고    scopus 로고
    • Inhibition of ERK-DLP1 signaling and mitochondrial division alleviates mitochondrial dysfunction in Alzheimer's disease cybrid cell
    • Gan, X. et al., Inhibition of ERK-DLP1 signaling and mitochondrial division alleviates mitochondrial dysfunction in Alzheimer's disease cybrid cell. Biochim. Biophys. Acta 1842:2 (2014), 220–231.
    • (2014) Biochim. Biophys. Acta , vol.1842 , Issue.2 , pp. 220-231
    • Gan, X.1
  • 123
    • 84901228307 scopus 로고    scopus 로고
    • c-Jun N-terminal kinase has a key role in Alzheimer disease synaptic dysfunction in vivo
    • Sclip, A. et al., c-Jun N-terminal kinase has a key role in Alzheimer disease synaptic dysfunction in vivo. Cell Death Dis., 5, 2014, e1019.
    • (2014) Cell Death Dis. , vol.5
    • Sclip, A.1
  • 124
    • 0035968225 scopus 로고    scopus 로고
    • Effects of the beta-amyloid and carboxyl-terminal fragment of Alzheimer's amyloid precursor protein on the production of the tumor necrosis factor-alpha and matrix metalloproteinase-9 by human monocytic THP-1
    • Chong, Y.H. et al., Effects of the beta-amyloid and carboxyl-terminal fragment of Alzheimer's amyloid precursor protein on the production of the tumor necrosis factor-alpha and matrix metalloproteinase-9 by human monocytic THP-1. J. Biol. Chem. 276:26 (2001), 23511–23517.
    • (2001) J. Biol. Chem. , vol.276 , Issue.26 , pp. 23511-23517
    • Chong, Y.H.1
  • 125
    • 0037166068 scopus 로고    scopus 로고
    • Amyloid precursor protein processing is separately regulated by protein kinase C and tyrosine kinase in human astrocytes
    • Kim, C. et al., Amyloid precursor protein processing is separately regulated by protein kinase C and tyrosine kinase in human astrocytes. Neurosci. Lett. 324:3 (2002), 185–188.
    • (2002) Neurosci. Lett. , vol.324 , Issue.3 , pp. 185-188
    • Kim, C.1
  • 126
    • 2942572699 scopus 로고    scopus 로고
    • Beta-amyloid-activated cell cycle in SH-SY5Y neuroblastoma cells: correlation with the MAP kinase pathway
    • Frasca, G. et al., Beta-amyloid-activated cell cycle in SH-SY5Y neuroblastoma cells: correlation with the MAP kinase pathway. J. Mol. Neurosci. 22:3 (2004), 231–236.
    • (2004) J. Mol. Neurosci. , vol.22 , Issue.3 , pp. 231-236
    • Frasca, G.1
  • 127
    • 0037022288 scopus 로고    scopus 로고
    • Beta-amyloid precursor protein transgenic mice that harbor diffuse A beta deposits but do not form plaques show increased ischemic vulnerability: role of inflammation
    • Koistinaho, M. et al., Beta-amyloid precursor protein transgenic mice that harbor diffuse A beta deposits but do not form plaques show increased ischemic vulnerability: role of inflammation. Proc. Natl. Acad. Sci. U. S. A. 99:3 (2002), 1610–1615.
    • (2002) Proc. Natl. Acad. Sci. U. S. A. , vol.99 , Issue.3 , pp. 1610-1615
    • Koistinaho, M.1
  • 128
    • 0036746143 scopus 로고    scopus 로고
    • Pro-inflammatory effect of freshly solubilized beta-amyloid peptides in the brain
    • Paris, D. et al., Pro-inflammatory effect of freshly solubilized beta-amyloid peptides in the brain. Prostaglandins Other Lipid Mediat. 70:1-2 (2002), 1–12.
    • (2002) Prostaglandins Other Lipid Mediat. , vol.70 , Issue.1-2 , pp. 1-12
    • Paris, D.1
  • 129
    • 4344566337 scopus 로고    scopus 로고
    • Calpain mediates calcium-induced activation of the erk1,2 MAPK pathway and cytoskeletal phosphorylation in neurons: relevance to Alzheimer's disease
    • Veeranna et al., Calpain mediates calcium-induced activation of the erk1,2 MAPK pathway and cytoskeletal phosphorylation in neurons: relevance to Alzheimer's disease. Am. J. Pathol. 165:3 (2004), 795–805.
    • (2004) Am. J. Pathol. , vol.165 , Issue.3 , pp. 795-805
    • Veeranna1
  • 130
    • 84872246867 scopus 로고    scopus 로고
    • p38 MAPK: a mediator of hypoxia-induced cerebrovascular inflammation
    • Sanchez, A. et al., p38 MAPK: a mediator of hypoxia-induced cerebrovascular inflammation. J. Alzheimers Dis. 32:3 (2012), 587–597.
    • (2012) J. Alzheimers Dis. , vol.32 , Issue.3 , pp. 587-597
    • Sanchez, A.1
  • 131
    • 35348886061 scopus 로고    scopus 로고
    • A novel p38 alpha MAPK inhibitor suppresses brain proinflammatory cytokine up-regulation and attenuates synaptic dysfunction and behavioral deficits in an Alzheimer's disease mouse model
    • Munoz, L. et al., A novel p38 alpha MAPK inhibitor suppresses brain proinflammatory cytokine up-regulation and attenuates synaptic dysfunction and behavioral deficits in an Alzheimer's disease mouse model. J. Neuroinflammation, 4, 2007, 21.
    • (2007) J. Neuroinflammation , vol.4 , pp. 21
    • Munoz, L.1
  • 132
    • 84864012606 scopus 로고    scopus 로고
    • Inhibiting p38 mitogen-activated protein kinase attenuates cerebral ischemic injury in Swedish mutant amyloid precursor protein transgenic mice
    • Zou, L. et al., Inhibiting p38 mitogen-activated protein kinase attenuates cerebral ischemic injury in Swedish mutant amyloid precursor protein transgenic mice. Neural Regen. Res. 7:14 (2012), 1088–1094.
    • (2012) Neural Regen. Res. , vol.7 , Issue.14 , pp. 1088-1094
    • Zou, L.1
  • 133
    • 84983002115 scopus 로고    scopus 로고
    • Changes in insulin and insulin signaling in Alzheimer's disease: cause or consequence?
    • Stanley, M., Macauley, S.L., Holtzman, D.M., Changes in insulin and insulin signaling in Alzheimer's disease: cause or consequence?. J. Exp. Med. 213:8 (2016), 1375–1385.
    • (2016) J. Exp. Med. , vol.213 , Issue.8 , pp. 1375-1385
    • Stanley, M.1    Macauley, S.L.2    Holtzman, D.M.3
  • 134
    • 84963958179 scopus 로고    scopus 로고
    • Mitochondrial dysfunction precedes depression of AMPK/AKT signaling in insulin resistance induced by high glucose in primary cortical neurons
    • Peng, Y. et al., Mitochondrial dysfunction precedes depression of AMPK/AKT signaling in insulin resistance induced by high glucose in primary cortical neurons. J. Neurochem. 137:5 (2016), 701–713.
    • (2016) J. Neurochem. , vol.137 , Issue.5 , pp. 701-713
    • Peng, Y.1
  • 135
    • 85008517944 scopus 로고    scopus 로고
    • Insulin resistance in Alzheimer's disease
    • Diehl, T., Mullins, R., Kapogiannis, D., Insulin resistance in Alzheimer's disease. Transl. Res. 183 (2017), 26–40.
    • (2017) Transl. Res. , vol.183 , pp. 26-40
    • Diehl, T.1    Mullins, R.2    Kapogiannis, D.3
  • 136
    • 85014760200 scopus 로고    scopus 로고
    • Role of immune cells in obesity induced low grade inflammation and insulin resistance
    • Asghar, A., Sheikh, N., Role of immune cells in obesity induced low grade inflammation and insulin resistance. Cell. Immunol., 2017.
    • (2017) Cell. Immunol.
    • Asghar, A.1    Sheikh, N.2
  • 137
    • 85015707316 scopus 로고    scopus 로고
    • Skeletal muscle insulin resistance: role of mitochondria and other ROS sources
    • Di Meo, S., Iossa, S., Venditti, P., Skeletal muscle insulin resistance: role of mitochondria and other ROS sources. J. Endocrinol. 233:1 (2017), R15–R42.
    • (2017) J. Endocrinol. , vol.233 , Issue.1 , pp. R15-R42
    • Di Meo, S.1    Iossa, S.2    Venditti, P.3
  • 138
    • 85006186623 scopus 로고    scopus 로고
    • Polycystic ovary syndrome
    • Azziz, R. et al., Polycystic ovary syndrome. Nat. Rev. Dis. Primers, 2, 2016, 16057.
    • (2016) Nat. Rev. Dis. Primers , vol.2 , pp. 16057
    • Azziz, R.1
  • 139
    • 84991709714 scopus 로고    scopus 로고
    • The impact of insulin resistance on the kidney and vasculature
    • Artunc, F. et al., The impact of insulin resistance on the kidney and vasculature. Nat. Rev. Nephrol. 12:12 (2016), 721–737.
    • (2016) Nat. Rev. Nephrol. , vol.12 , Issue.12 , pp. 721-737
    • Artunc, F.1
  • 140
    • 85018502931 scopus 로고    scopus 로고
    • The ubiquitin ligase CHIP integrates proteostasis and aging by regulation of insulin receptor turnover
    • (e13)
    • Tawo, R. et al., The ubiquitin ligase CHIP integrates proteostasis and aging by regulation of insulin receptor turnover. Cell 169:3 (2017), 470–482 (e13).
    • (2017) Cell , vol.169 , Issue.3 , pp. 470-482
    • Tawo, R.1
  • 141
    • 85006410178 scopus 로고    scopus 로고
    • Insulin resistance and neurodegeneration: progress towards the development of new therapeutics for Alzheimer's disease
    • de la Monte, S.M., Insulin resistance and neurodegeneration: progress towards the development of new therapeutics for Alzheimer's disease. Drugs 77:1 (2017), 47–65.
    • (2017) Drugs , vol.77 , Issue.1 , pp. 47-65
    • de la Monte, S.M.1
  • 142
    • 84973428782 scopus 로고    scopus 로고
    • Targeting insulin signaling for the treatment of Alzheimer's disease
    • Chen, Y. et al., Targeting insulin signaling for the treatment of Alzheimer's disease. Curr. Top. Med. Chem. 16:5 (2016), 485–492.
    • (2016) Curr. Top. Med. Chem. , vol.16 , Issue.5 , pp. 485-492
    • Chen, Y.1
  • 144
    • 0000398342 scopus 로고    scopus 로고
    • Insulin-degrading enzyme regulates extracellular levels of amyloid beta-protein by degradation
    • Qiu, W.Q. et al., Insulin-degrading enzyme regulates extracellular levels of amyloid beta-protein by degradation. J. Biol. Chem. 273:49 (1998), 32730–32738.
    • (1998) J. Biol. Chem. , vol.273 , Issue.49 , pp. 32730-32738
    • Qiu, W.Q.1
  • 145
    • 84931271345 scopus 로고    scopus 로고
    • Aberrant insulin signaling in Alzheimer's disease: current knowledge
    • Bedse, G. et al., Aberrant insulin signaling in Alzheimer's disease: current knowledge. Front. Neurosci., 9, 2015, 204.
    • (2015) Front. Neurosci. , vol.9 , pp. 204
    • Bedse, G.1
  • 146
    • 67349280677 scopus 로고    scopus 로고
    • Erythropoietin protects PC12 cells from beta-amyloid(25–35)-induced apoptosis via PI3K/Akt signaling pathway
    • Ma, R. et al., Erythropoietin protects PC12 cells from beta-amyloid(25–35)-induced apoptosis via PI3K/Akt signaling pathway. Neuropharmacology 56:6-7 (2009), 1027–1034.
    • (2009) Neuropharmacology , vol.56 , Issue.6-7 , pp. 1027-1034
    • Ma, R.1
  • 147
    • 0029587224 scopus 로고
    • Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B
    • Cross, D.A. et al., Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature 378:6559 (1995), 785–789.
    • (1995) Nature , vol.378 , Issue.6559 , pp. 785-789
    • Cross, D.A.1
  • 148
    • 0027255817 scopus 로고
    • Glycogen synthase kinase 3 beta is identical to tau protein kinase I generating several epitopes of paired helical filaments
    • Ishiguro, K. et al., Glycogen synthase kinase 3 beta is identical to tau protein kinase I generating several epitopes of paired helical filaments. FEBS Lett. 325:3 (1993), 167–172.
    • (1993) FEBS Lett. , vol.325 , Issue.3 , pp. 167-172
    • Ishiguro, K.1
  • 149
    • 84893459319 scopus 로고    scopus 로고
    • SIRT1 in neurodevelopment and brain senescence
    • Herskovits, A.Z., Guarente, L., SIRT1 in neurodevelopment and brain senescence. Neuron 81:3 (2014), 471–483.
    • (2014) Neuron , vol.81 , Issue.3 , pp. 471-483
    • Herskovits, A.Z.1    Guarente, L.2
  • 150
    • 84874709843 scopus 로고    scopus 로고
    • SIRT1 and SIRT2: emerging targets in neurodegeneration
    • Donmez, G., Outeiro, T.F., SIRT1 and SIRT2: emerging targets in neurodegeneration. EMBO Mol. Med. 5:3 (2013), 344–352.
    • (2013) EMBO Mol. Med. , vol.5 , Issue.3 , pp. 344-352
    • Donmez, G.1    Outeiro, T.F.2
  • 151
    • 84978870848 scopus 로고    scopus 로고
    • mTORC1 and SIRT1 cooperate to foster expansion of gut adult stem cells during calorie restriction
    • Igarashi, M., Guarente, L., mTORC1 and SIRT1 cooperate to foster expansion of gut adult stem cells during calorie restriction. Cell 166:2 (2016), 436–450.
    • (2016) Cell , vol.166 , Issue.2 , pp. 436-450
    • Igarashi, M.1    Guarente, L.2
  • 152
    • 84880160092 scopus 로고    scopus 로고
    • Antagonistic crosstalk between NF-kappaB and SIRT1 in the regulation of inflammation and metabolic disorders
    • Kauppinen, A. et al., Antagonistic crosstalk between NF-kappaB and SIRT1 in the regulation of inflammation and metabolic disorders. Cell. Signal. 25:10 (2013), 1939–1948.
    • (2013) Cell. Signal. , vol.25 , Issue.10 , pp. 1939-1948
    • Kauppinen, A.1
  • 153
    • 37349110355 scopus 로고    scopus 로고
    • Metabolic adaptations through the PGC-1 alpha and SIRT1 pathways
    • Rodgers, J.T. et al., Metabolic adaptations through the PGC-1 alpha and SIRT1 pathways. FEBS Lett. 582:1 (2008), 46–53.
    • (2008) FEBS Lett. , vol.582 , Issue.1 , pp. 46-53
    • Rodgers, J.T.1
  • 154
    • 84893202518 scopus 로고    scopus 로고
    • Role of Sirt1 during the ageing process: relevance to protection of synapses in the brain
    • Godoy, J.A. et al., Role of Sirt1 during the ageing process: relevance to protection of synapses in the brain. Mol. Neurobiol. 50:3 (2014), 744–756.
    • (2014) Mol. Neurobiol. , vol.50 , Issue.3 , pp. 744-756
    • Godoy, J.A.1
  • 155
    • 84858750516 scopus 로고    scopus 로고
    • Metabolic actions of hypothalamic SIRT1
    • Coppari, R., Metabolic actions of hypothalamic SIRT1. Trends Endocrinol. Metab. 23:4 (2012), 179–185.
    • (2012) Trends Endocrinol. Metab. , vol.23 , Issue.4 , pp. 179-185
    • Coppari, R.1
  • 156
    • 79951686964 scopus 로고    scopus 로고
    • The sirtuin pathway in ageing and Alzheimer disease: mechanistic and therapeutic considerations
    • Bonda, D.J. et al., The sirtuin pathway in ageing and Alzheimer disease: mechanistic and therapeutic considerations. Lancet Neurol. 10:3 (2011), 275–279.
    • (2011) Lancet Neurol. , vol.10 , Issue.3 , pp. 275-279
    • Bonda, D.J.1
  • 157
    • 85003047354 scopus 로고    scopus 로고
    • Roles of resveratrol and other grape-derived polyphenols in Alzheimer's disease prevention and treatment
    • Pasinetti, G.M. et al., Roles of resveratrol and other grape-derived polyphenols in Alzheimer's disease prevention and treatment. Biochim. Biophys. Acta 1852:6 (2015), 1202–1208.
    • (2015) Biochim. Biophys. Acta , vol.1852 , Issue.6 , pp. 1202-1208
    • Pasinetti, G.M.1
  • 158
    • 84883497243 scopus 로고    scopus 로고
    • SIRT1 negatively regulates amyloid-beta-induced inflammation via the NF-kappaB pathway
    • Cao, L. et al., SIRT1 negatively regulates amyloid-beta-induced inflammation via the NF-kappaB pathway. Braz. J. Med. Biol. Res. 46:8 (2013), 659–669.
    • (2013) Braz. J. Med. Biol. Res. , vol.46 , Issue.8 , pp. 659-669
    • Cao, L.1
  • 159
    • 84875550716 scopus 로고    scopus 로고
    • Resveratrol inhibits beta-amyloid-induced neuronal apoptosis through regulation of SIRT1-ROCK1 signaling pathway
    • Feng, X. et al., Resveratrol inhibits beta-amyloid-induced neuronal apoptosis through regulation of SIRT1-ROCK1 signaling pathway. PLoS One, 8(3), 2013, e59888.
    • (2013) PLoS One , vol.8 , Issue.3
    • Feng, X.1
  • 160
    • 39749087530 scopus 로고    scopus 로고
    • SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization
    • Han, M.K. et al., SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization. Cell Stem Cell 2:3 (2008), 241–251.
    • (2008) Cell Stem Cell , vol.2 , Issue.3 , pp. 241-251
    • Han, M.K.1
  • 161
    • 12144290563 scopus 로고    scopus 로고
    • Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase
    • Brunet, A. et al., Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303:5666 (2004), 2011–2015.
    • (2004) Science , vol.303 , Issue.5666 , pp. 2011-2015
    • Brunet, A.1
  • 162
    • 77957001697 scopus 로고    scopus 로고
    • Acetylation of tau inhibits its degradation and contributes to tauopathy
    • Min, S.W. et al., Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron 67:6 (2010), 953–966.
    • (2010) Neuron , vol.67 , Issue.6 , pp. 953-966
    • Min, S.W.1
  • 163
    • 28844474597 scopus 로고    scopus 로고
    • SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling
    • Chen, J. et al., SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling. J. Biol. Chem. 280:48 (2005), 40364–40374.
    • (2005) J. Biol. Chem. , vol.280 , Issue.48 , pp. 40364-40374
    • Chen, J.1
  • 164
    • 0036359548 scopus 로고    scopus 로고
    • Hypoxia—a key regulatory factor in tumour growth
    • Harris, A.L., Hypoxia—a key regulatory factor in tumour growth. Nat. Rev. Cancer 2:1 (2002), 38–47.
    • (2002) Nat. Rev. Cancer , vol.2 , Issue.1 , pp. 38-47
    • Harris, A.L.1
  • 165
    • 85013697298 scopus 로고    scopus 로고
    • pVHL suppresses kinase activity of Akt in a proline-hydroxylation-dependent manner
    • Guo, J. et al., pVHL suppresses kinase activity of Akt in a proline-hydroxylation-dependent manner. Science 353:6302 (2016), 929–932.
    • (2016) Science , vol.353 , Issue.6302 , pp. 929-932
    • Guo, J.1
  • 166
    • 0037362890 scopus 로고    scopus 로고
    • HIF hydroxylation and the mammalian oxygen-sensing pathway
    • Safran, M., Kaelin, W.G. Jr., HIF hydroxylation and the mammalian oxygen-sensing pathway. J. Clin. Invest. 111:6 (2003), 779–783.
    • (2003) J. Clin. Invest. , vol.111 , Issue.6 , pp. 779-783
    • Safran, M.1    Kaelin, W.G.2
  • 167
    • 0035917313 scopus 로고    scopus 로고
    • 2 sensing
    • 2 sensing. Science 292:5516 (2001), 464–468.
    • (2001) Science , vol.292 , Issue.5516 , pp. 464-468
    • Ivan, M.1
  • 168
    • 17944375360 scopus 로고    scopus 로고
    • C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation
    • Epstein, A.C. et al., C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 107:1 (2001), 43–54.
    • (2001) Cell , vol.107 , Issue.1 , pp. 43-54
    • Epstein, A.C.1
  • 169
    • 0038037735 scopus 로고    scopus 로고
    • Regulation of angiogenesis by hypoxia: role of the HIF system
    • Pugh, C.W., Ratcliffe, P.J., Regulation of angiogenesis by hypoxia: role of the HIF system. Nat. Med. 9:6 (2003), 677–684.
    • (2003) Nat. Med. , vol.9 , Issue.6 , pp. 677-684
    • Pugh, C.W.1    Ratcliffe, P.J.2
  • 170
    • 0032581277 scopus 로고    scopus 로고
    • Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis
    • Carmeliet, P. et al., Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 394:6692 (1998), 485–490.
    • (1998) Nature , vol.394 , Issue.6692 , pp. 485-490
    • Carmeliet, P.1
  • 171
    • 84994626953 scopus 로고    scopus 로고
    • On-target efficacy of a HIF-2alpha antagonist in preclinical kidney cancer models
    • Cho, H. et al., On-target efficacy of a HIF-2alpha antagonist in preclinical kidney cancer models. Nature 539:7627 (2016), 107–111.
    • (2016) Nature , vol.539 , Issue.7627 , pp. 107-111
    • Cho, H.1
  • 172
    • 80054771537 scopus 로고    scopus 로고
    • Genetic and functional studies implicate HIF1alpha as a 14q kidney cancer suppressor gene
    • Shen, C. et al., Genetic and functional studies implicate HIF1alpha as a 14q kidney cancer suppressor gene. Cancer Discov. 1:3 (2011), 222–235.
    • (2011) Cancer Discov. , vol.1 , Issue.3 , pp. 222-235
    • Shen, C.1
  • 173
    • 84862776918 scopus 로고    scopus 로고
    • Transformation by the (R)-enantiomer of 2-hydroxyglutarate linked to EGLN activation
    • Koivunen, P. et al., Transformation by the (R)-enantiomer of 2-hydroxyglutarate linked to EGLN activation. Nature 483:7390 (2012), 484–488.
    • (2012) Nature , vol.483 , Issue.7390 , pp. 484-488
    • Koivunen, P.1
  • 174
    • 70350402494 scopus 로고    scopus 로고
    • Hypoxia and neurodegeneration
    • Peers, C. et al., Hypoxia and neurodegeneration. Ann. N. Y. Acad. Sci. 1177 (2009), 169–177.
    • (2009) Ann. N. Y. Acad. Sci. , vol.1177 , pp. 169-177
    • Peers, C.1
  • 175
    • 84919576857 scopus 로고    scopus 로고
    • Hypoxia-induced tau phosphorylation and memory deficit in rats
    • Zhang, C.E. et al., Hypoxia-induced tau phosphorylation and memory deficit in rats. Neurodegener. Dis. 14:3 (2014), 107–116.
    • (2014) Neurodegener. Dis. , vol.14 , Issue.3 , pp. 107-116
    • Zhang, C.E.1
  • 176
    • 0036773010 scopus 로고    scopus 로고
    • Hypoxia signaling to genes: significance in Alzheimer's disease
    • Bazan, N.G., Palacios-Pelaez, R., Lukiw, W.J., Hypoxia signaling to genes: significance in Alzheimer's disease. Mol. Neurobiol. 26:2-3 (2002), 283–298.
    • (2002) Mol. Neurobiol. , vol.26 , Issue.2-3 , pp. 283-298
    • Bazan, N.G.1    Palacios-Pelaez, R.2    Lukiw, W.J.3
  • 177
    • 0037098737 scopus 로고    scopus 로고
    • Hypoxia potentiates exocytosis and Ca2 + channels in PC12 cells via increased amyloid beta peptide formation and reactive oxygen species generation
    • Green, K.N., Boyle, J.P., Peers, C., Hypoxia potentiates exocytosis and Ca2 + channels in PC12 cells via increased amyloid beta peptide formation and reactive oxygen species generation. J. Physiol. 541:Pt 3 (2002), 1013–1023.
    • (2002) J. Physiol. , vol.541 , pp. 1013-1023
    • Green, K.N.1    Boyle, J.P.2    Peers, C.3
  • 178
    • 80055063239 scopus 로고    scopus 로고
    • Brain microvasculature and hypoxia-related proteins in Alzheimer's disease
    • Grammas, P. et al., Brain microvasculature and hypoxia-related proteins in Alzheimer's disease. Int. J. Clin. Exp. Pathol. 4:6 (2011), 616–627.
    • (2011) Int. J. Clin. Exp. Pathol. , vol.4 , Issue.6 , pp. 616-627
    • Grammas, P.1
  • 179
    • 54549089738 scopus 로고    scopus 로고
    • Hypoxia signalling through mTOR and the unfolded protein response in cancer
    • Wouters, B.G., Koritzinsky, M., Hypoxia signalling through mTOR and the unfolded protein response in cancer. Nat. Rev. Cancer 8:11 (2008), 851–864.
    • (2008) Nat. Rev. Cancer , vol.8 , Issue.11 , pp. 851-864
    • Wouters, B.G.1    Koritzinsky, M.2
  • 180
    • 70350767274 scopus 로고    scopus 로고
    • Oxidative stress and hypoxia contribute to Alzheimer's disease pathogenesis: two sides of the same coin
    • Guglielmotto, M., Tamagno, E., Danni, O., Oxidative stress and hypoxia contribute to Alzheimer's disease pathogenesis: two sides of the same coin. ScientificWorldJournal 9 (2009), 781–791.
    • (2009) ScientificWorldJournal , vol.9 , pp. 781-791
    • Guglielmotto, M.1    Tamagno, E.2    Danni, O.3
  • 181
    • 84970919297 scopus 로고    scopus 로고
    • Hypoxia signaling pathways in cancer metabolism: the importance of co-selecting interconnected physiological pathways
    • Masson, N., Ratcliffe, P.J., Hypoxia signaling pathways in cancer metabolism: the importance of co-selecting interconnected physiological pathways. Cancer Metab., 2(1), 2014, 3.
    • (2014) Cancer Metab. , vol.2 , Issue.1 , pp. 3
    • Masson, N.1    Ratcliffe, P.J.2
  • 182
    • 29044433048 scopus 로고    scopus 로고
    • Erythropoietin requires NF-kappaB and its nuclear translocation to prevent early and late apoptotic neuronal injury during beta-amyloid toxicity
    • Chong, Z.Z., Li, F., Maiese, K., Erythropoietin requires NF-kappaB and its nuclear translocation to prevent early and late apoptotic neuronal injury during beta-amyloid toxicity. Curr. Neurovasc. Res. 2:5 (2005), 387–399.
    • (2005) Curr. Neurovasc. Res. , vol.2 , Issue.5 , pp. 387-399
    • Chong, Z.Z.1    Li, F.2    Maiese, K.3
  • 183
    • 33846221802 scopus 로고    scopus 로고
    • Vascular endothelial growth factor overexpression delays neurodegeneration and prolongs survival in amyotrophic lateral sclerosis mice
    • Wang, Y. et al., Vascular endothelial growth factor overexpression delays neurodegeneration and prolongs survival in amyotrophic lateral sclerosis mice. J. Neurosci. 27:2 (2007), 304–307.
    • (2007) J. Neurosci. , vol.27 , Issue.2 , pp. 304-307
    • Wang, Y.1
  • 184
    • 33845505233 scopus 로고    scopus 로고
    • Hypoxia facilitates Alzheimer's disease pathogenesis by up-regulating BACE1 gene expression
    • Sun, X. et al., Hypoxia facilitates Alzheimer's disease pathogenesis by up-regulating BACE1 gene expression. Proc. Natl. Acad. Sci. U. S. A. 103:49 (2006), 18727–18732.
    • (2006) Proc. Natl. Acad. Sci. U. S. A. , vol.103 , Issue.49 , pp. 18727-18732
    • Sun, X.1
  • 185
    • 27144483873 scopus 로고    scopus 로고
    • Hypoxia inducible factor-1 and facilitative glucose transporters GLUT1 and GLUT3: putative molecular components of the oxygen and glucose sensing apparatus in articular chondrocytes
    • Mobasheri, A. et al., Hypoxia inducible factor-1 and facilitative glucose transporters GLUT1 and GLUT3: putative molecular components of the oxygen and glucose sensing apparatus in articular chondrocytes. Histol. Histopathol. 20:4 (2005), 1327–1338.
    • (2005) Histol. Histopathol. , vol.20 , Issue.4 , pp. 1327-1338
    • Mobasheri, A.1
  • 186
    • 38049050725 scopus 로고    scopus 로고
    • Decreased glucose transporters correlate to abnormal hyperphosphorylation of tau in Alzheimer disease
    • Liu, Y. et al., Decreased glucose transporters correlate to abnormal hyperphosphorylation of tau in Alzheimer disease. FEBS Lett. 582:2 (2008), 359–364.
    • (2008) FEBS Lett. , vol.582 , Issue.2 , pp. 359-364
    • Liu, Y.1
  • 187
    • 77951499897 scopus 로고    scopus 로고
    • Acute hypoxia promote the phosphorylation of tau via ERK pathway
    • Fang, H. et al., Acute hypoxia promote the phosphorylation of tau via ERK pathway. Neurosci. Lett. 474:3 (2010), 173–177.
    • (2010) Neurosci. Lett. , vol.474 , Issue.3 , pp. 173-177
    • Fang, H.1
  • 188
    • 33847217304 scopus 로고    scopus 로고
    • Evidence of oxidative stress-induced BNIP3 expression in amyloid beta neurotoxicity
    • Zhang, S. et al., Evidence of oxidative stress-induced BNIP3 expression in amyloid beta neurotoxicity. Brain Res. 1138 (2007), 221–230.
    • (2007) Brain Res. , vol.1138 , pp. 221-230
    • Zhang, S.1
  • 189
    • 0036728244 scopus 로고    scopus 로고
    • Oxidative stress, antioxidants and stress tolerance
    • Mittler, R., Oxidative stress, antioxidants and stress tolerance. Trends Plant Sci. 7:9 (2002), 405–410.
    • (2002) Trends Plant Sci. , vol.7 , Issue.9 , pp. 405-410
    • Mittler, R.1
  • 190
    • 0038799736 scopus 로고    scopus 로고
    • Oxidative DNA damage: mechanisms, mutation, and disease
    • Cooke, M.S. et al., Oxidative DNA damage: mechanisms, mutation, and disease. FASEB J. 17:10 (2003), 1195–1214.
    • (2003) FASEB J. , vol.17 , Issue.10 , pp. 1195-1214
    • Cooke, M.S.1
  • 191
    • 0034961888 scopus 로고    scopus 로고
    • Role of reactive oxygen species in cell signalling pathways
    • Hancock, J.T., Desikan, R., Neill, S.J., Role of reactive oxygen species in cell signalling pathways. Biochem. Soc. Trans. 29:Pt 2 (2001), 345–350.
    • (2001) Biochem. Soc. Trans. , vol.29 , pp. 345-350
    • Hancock, J.T.1    Desikan, R.2    Neill, S.J.3
  • 192
    • 78650894319 scopus 로고    scopus 로고
    • Crosstalk of reactive oxygen species and NF-kappaB signaling
    • Morgan, M.J., Liu, Z.G., Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 21:1 (2011), 103–115.
    • (2011) Cell Res. , vol.21 , Issue.1 , pp. 103-115
    • Morgan, M.J.1    Liu, Z.G.2
  • 193
    • 49449117608 scopus 로고    scopus 로고
    • Reactive oxygen species regulate hypoxia-inducible factor 1alpha differentially in cancer and ischemia
    • Qutub, A.A., Popel, A.S., Reactive oxygen species regulate hypoxia-inducible factor 1alpha differentially in cancer and ischemia. Mol. Cell. Biol. 28:16 (2008), 5106–5119.
    • (2008) Mol. Cell. Biol. , vol.28 , Issue.16 , pp. 5106-5119
    • Qutub, A.A.1    Popel, A.S.2
  • 194
    • 84857274709 scopus 로고    scopus 로고
    • Oxidative stress and antioxidant defense
    • Birben, E. et al., Oxidative stress and antioxidant defense. World Allergy Organ. J. 5:1 (2012), 9–19.
    • (2012) World Allergy Organ. J. , vol.5 , Issue.1 , pp. 9-19
    • Birben, E.1
  • 195
    • 64949138993 scopus 로고    scopus 로고
    • ROS and innate immunity
    • Kohchi, C. et al., ROS and innate immunity. Anticancer Res. 29:3 (2009), 817–821.
    • (2009) Anticancer Res. , vol.29 , Issue.3 , pp. 817-821
    • Kohchi, C.1
  • 196
    • 84892842130 scopus 로고    scopus 로고
    • Positive oxidative stress in aging and aging-related disease tolerance
    • Yan, L.J., Positive oxidative stress in aging and aging-related disease tolerance. Redox Biol. 2 (2014), 165–169.
    • (2014) Redox Biol. , vol.2 , pp. 165-169
    • Yan, L.J.1
  • 197
    • 76049083966 scopus 로고    scopus 로고
    • Reactive oxygen species, cellular redox systems, and apoptosis
    • Circu, M.L., Aw, T.Y., Reactive oxygen species, cellular redox systems, and apoptosis. Free Radic. Biol. Med. 48:6 (2010), 749–762.
    • (2010) Free Radic. Biol. Med. , vol.48 , Issue.6 , pp. 749-762
    • Circu, M.L.1    Aw, T.Y.2
  • 198
    • 80054993459 scopus 로고    scopus 로고
    • Mitogen-activated protein kinases and reactive oxygen species: how can ROS activate MAPK pathways?
    • Son, Y. et al., Mitogen-activated protein kinases and reactive oxygen species: how can ROS activate MAPK pathways?. J. Signal Transduct., 2011, 2011, 792639.
    • (2011) J. Signal Transduct. , vol.2011 , pp. 792639
    • Son, Y.1
  • 199
    • 0142137134 scopus 로고    scopus 로고
    • Redox regulation of PI 3-kinase signalling via inactivation of PTEN
    • Leslie, N.R. et al., Redox regulation of PI 3-kinase signalling via inactivation of PTEN. EMBO J. 22:20 (2003), 5501–5510.
    • (2003) EMBO J. , vol.22 , Issue.20 , pp. 5501-5510
    • Leslie, N.R.1
  • 200
    • 84878708044 scopus 로고    scopus 로고
    • Reactive oxygen species-mediated activation of the Akt/ASK1/p38 signaling cascade and p21(Cip1) downregulation are required for shikonin-induced apoptosis
    • Ahn, J. et al., Reactive oxygen species-mediated activation of the Akt/ASK1/p38 signaling cascade and p21(Cip1) downregulation are required for shikonin-induced apoptosis. Apoptosis 18:7 (2013), 870–881.
    • (2013) Apoptosis , vol.18 , Issue.7 , pp. 870-881
    • Ahn, J.1
  • 201
    • 0033729006 scopus 로고    scopus 로고
    • Influence of Helicobacter pylori on reactive oxygen-induced gastric epithelial cell injury
    • Smoot, D.T. et al., Influence of Helicobacter pylori on reactive oxygen-induced gastric epithelial cell injury. Carcinogenesis 21:11 (2000), 2091–2095.
    • (2000) Carcinogenesis , vol.21 , Issue.11 , pp. 2091-2095
    • Smoot, D.T.1
  • 202
    • 0034626735 scopus 로고    scopus 로고
    • Oxidants, oxidative stress and the biology of ageing
    • Finkel, T., Holbrook, N.J., Oxidants, oxidative stress and the biology of ageing. Nature 408:6809 (2000), 239–247.
    • (2000) Nature , vol.408 , Issue.6809 , pp. 239-247
    • Finkel, T.1    Holbrook, N.J.2
  • 203
    • 61449184625 scopus 로고    scopus 로고
    • Deletion of the mitochondrial superoxide dismutase sod-2 extends lifespan in Caenorhabditis elegans
    • Van Raamsdonk, J.M., Hekimi, S., Deletion of the mitochondrial superoxide dismutase sod-2 extends lifespan in Caenorhabditis elegans. PLoS Genet., 5(2), 2009, e1000361.
    • (2009) PLoS Genet. , vol.5 , Issue.2
    • Van Raamsdonk, J.M.1    Hekimi, S.2
  • 204
    • 0035503501 scopus 로고    scopus 로고
    • Lifespan extension and rescue of spongiform encephalopathy in superoxide dismutase 2 nullizygous mice treated with superoxide dismutase-catalase mimetics
    • Melov, S. et al., Lifespan extension and rescue of spongiform encephalopathy in superoxide dismutase 2 nullizygous mice treated with superoxide dismutase-catalase mimetics. J. Neurosci. 21:21 (2001), 8348–8353.
    • (2001) J. Neurosci. , vol.21 , Issue.21 , pp. 8348-8353
    • Melov, S.1
  • 205
    • 0034923434 scopus 로고    scopus 로고
    • Oxidative damage is the earliest event in Alzheimer disease
    • Nunomura, A. et al., Oxidative damage is the earliest event in Alzheimer disease. J. Neuropathol. Exp. Neurol. 60:8 (2001), 759–767.
    • (2001) J. Neuropathol. Exp. Neurol. , vol.60 , Issue.8 , pp. 759-767
    • Nunomura, A.1
  • 206
    • 33646152108 scopus 로고    scopus 로고
    • Mitochondria are a direct site of A beta accumulation in Alzheimer's disease neurons: implications for free radical generation and oxidative damage in disease progression
    • Manczak, M. et al., Mitochondria are a direct site of A beta accumulation in Alzheimer's disease neurons: implications for free radical generation and oxidative damage in disease progression. Hum. Mol. Genet. 15:9 (2006), 1437–1449.
    • (2006) Hum. Mol. Genet. , vol.15 , Issue.9 , pp. 1437-1449
    • Manczak, M.1
  • 207
    • 33750347347 scopus 로고    scopus 로고
    • Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases
    • Lin, M.T., Beal, M.F., Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443:7113 (2006), 787–795.
    • (2006) Nature , vol.443 , Issue.7113 , pp. 787-795
    • Lin, M.T.1    Beal, M.F.2
  • 208
    • 0032514165 scopus 로고    scopus 로고
    • Promotion of transition metal-induced reactive oxygen species formation by beta-amyloid
    • Bondy, S.C., Guo-Ross, S.X., Truong, A.T., Promotion of transition metal-induced reactive oxygen species formation by beta-amyloid. Brain Res. 799:1 (1998), 91–96.
    • (1998) Brain Res. , vol.799 , Issue.1 , pp. 91-96
    • Bondy, S.C.1    Guo-Ross, S.X.2    Truong, A.T.3
  • 209
    • 84874690163 scopus 로고    scopus 로고
    • Transition metals: a double edge sward in ROS generation and signaling
    • Rodrigo-Moreno, A., Poschenrieder, C., Shabala, S., Transition metals: a double edge sward in ROS generation and signaling. Plant Signal. Behav., 8(3), 2013, e23425.
    • (2013) Plant Signal. Behav. , vol.8 , Issue.3
    • Rodrigo-Moreno, A.1    Poschenrieder, C.2    Shabala, S.3
  • 210
    • 78650436415 scopus 로고    scopus 로고
    • Enhanced ROS generation mediated by Alzheimer's disease presenilin regulation of InsP3R Ca2 + signaling
    • Muller, M., Cheung, K.H., Foskett, J.K., Enhanced ROS generation mediated by Alzheimer's disease presenilin regulation of InsP3R Ca2 + signaling. Antioxid. Redox Signal. 14:7 (2011), 1225–1235.
    • (2011) Antioxid. Redox Signal. , vol.14 , Issue.7 , pp. 1225-1235
    • Muller, M.1    Cheung, K.H.2    Foskett, J.K.3
  • 211
    • 0035105715 scopus 로고    scopus 로고
    • Ratio of 8-hydroxyguanine in intact DNA to free 8-hydroxyguanine is increased in Alzheimer disease ventricular cerebrospinal fluid
    • Lovell, M.A., Markesbery, W.R., Ratio of 8-hydroxyguanine in intact DNA to free 8-hydroxyguanine is increased in Alzheimer disease ventricular cerebrospinal fluid. Arch. Neurol. 58:3 (2001), 392–396.
    • (2001) Arch. Neurol. , vol.58 , Issue.3 , pp. 392-396
    • Lovell, M.A.1    Markesbery, W.R.2
  • 212
    • 18344414746 scopus 로고    scopus 로고
    • The A beta peptide of Alzheimer's disease directly produces hydrogen peroxide through metal ion reduction
    • Huang, X. et al., The A beta peptide of Alzheimer's disease directly produces hydrogen peroxide through metal ion reduction. Biochemistry 38:24 (1999), 7609–7616.
    • (1999) Biochemistry , vol.38 , Issue.24 , pp. 7609-7616
    • Huang, X.1
  • 213
    • 0033788416 scopus 로고    scopus 로고
    • Vitamin E prevents Alzheimer's amyloid beta-peptide (1–42)-induced neuronal protein oxidation and reactive oxygen species production
    • Yatin, S.M., Varadarajan, S., Butterfield, D.A., Vitamin E prevents Alzheimer's amyloid beta-peptide (1–42)-induced neuronal protein oxidation and reactive oxygen species production. J. Alzheimers Dis. 2:2 (2000), 123–131.
    • (2000) J. Alzheimers Dis. , vol.2 , Issue.2 , pp. 123-131
    • Yatin, S.M.1    Varadarajan, S.2    Butterfield, D.A.3
  • 214
    • 24044472067 scopus 로고    scopus 로고
    • Vitamin E and donepezil for the treatment of mild cognitive impairment
    • (author reply 951-2)
    • Barnes, D.E., Yaffe, K., Vitamin E and donepezil for the treatment of mild cognitive impairment. N. Engl. J. Med. 353:9 (2005), 951–952 (author reply 951-2).
    • (2005) N. Engl. J. Med. , vol.353 , Issue.9 , pp. 951-952
    • Barnes, D.E.1    Yaffe, K.2
  • 215
    • 4444265582 scopus 로고    scopus 로고
    • Degradation of misfolded proteins prevents ER-derived oxidative stress and cell death
    • Haynes, C.M., Titus, E.A., Cooper, A.A., Degradation of misfolded proteins prevents ER-derived oxidative stress and cell death. Mol. Cell 15:5 (2004), 767–776.
    • (2004) Mol. Cell , vol.15 , Issue.5 , pp. 767-776
    • Haynes, C.M.1    Titus, E.A.2    Cooper, A.A.3
  • 216
    • 1842504323 scopus 로고    scopus 로고
    • Redox-active metals, oxidative stress, and Alzheimer's disease pathology
    • Huang, X. et al., Redox-active metals, oxidative stress, and Alzheimer's disease pathology. Ann. N. Y. Acad. Sci. 1012 (2004), 153–163.
    • (2004) Ann. N. Y. Acad. Sci. , vol.1012 , pp. 153-163
    • Huang, X.1
  • 217
    • 75149152568 scopus 로고    scopus 로고
    • Increased iron and free radical generation in preclinical Alzheimer disease and mild cognitive impairment
    • Smith, M.A. et al., Increased iron and free radical generation in preclinical Alzheimer disease and mild cognitive impairment. J. Alzheimers Dis. 19:1 (2010), 363–372.
    • (2010) J. Alzheimers Dis. , vol.19 , Issue.1 , pp. 363-372
    • Smith, M.A.1
  • 218
    • 3042666730 scopus 로고    scopus 로고
    • Aluminium, iron, zinc and copper influence the in vitro formation of amyloid fibrils of Abeta42 in a manner which may have consequences for metal chelation therapy in Alzheimer's disease
    • House, E. et al., Aluminium, iron, zinc and copper influence the in vitro formation of amyloid fibrils of Abeta42 in a manner which may have consequences for metal chelation therapy in Alzheimer's disease. J. Alzheimers Dis. 6:3 (2004), 291–301.
    • (2004) J. Alzheimers Dis. , vol.6 , Issue.3 , pp. 291-301
    • House, E.1
  • 219
    • 0034964390 scopus 로고    scopus 로고
    • Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimer's disease transgenic mice
    • Cherny, R.A. et al., Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimer's disease transgenic mice. Neuron 30:3 (2001), 665–676.
    • (2001) Neuron , vol.30 , Issue.3 , pp. 665-676
    • Cherny, R.A.1
  • 220
    • 84869080922 scopus 로고    scopus 로고
    • Intranasal deferoxamine reverses iron-induced memory deficits and inhibits amyloidogenic APP processing in a transgenic mouse model of Alzheimer's disease
    • Guo, C. et al., Intranasal deferoxamine reverses iron-induced memory deficits and inhibits amyloidogenic APP processing in a transgenic mouse model of Alzheimer's disease. Neurobiol. Aging 34:2 (2013), 562–575.
    • (2013) Neurobiol. Aging , vol.34 , Issue.2 , pp. 562-575
    • Guo, C.1
  • 221
    • 0036710931 scopus 로고    scopus 로고
    • Lipid peroxidation in aging brain and Alzheimer's disease
    • Montine, T.J. et al., Lipid peroxidation in aging brain and Alzheimer's disease. Free Radic. Biol. Med. 33:5 (2002), 620–626.
    • (2002) Free Radic. Biol. Med. , vol.33 , Issue.5 , pp. 620-626
    • Montine, T.J.1
  • 222
    • 79251517382 scopus 로고    scopus 로고
    • Regulation of cancer cell metabolism
    • Cairns, R.A., Harris, I.S., Mak, T.W., Regulation of cancer cell metabolism. Nat. Rev. Cancer 11:2 (2011), 85–95.
    • (2011) Nat. Rev. Cancer , vol.11 , Issue.2 , pp. 85-95
    • Cairns, R.A.1    Harris, I.S.2    Mak, T.W.3
  • 223
    • 84894523716 scopus 로고    scopus 로고
    • Making new contacts: the mTOR network in metabolism and signalling crosstalk
    • Shimobayashi, M., Hall, M.N., Making new contacts: the mTOR network in metabolism and signalling crosstalk. Nat. Rev. Mol. Cell Biol. 15:3 (2014), 155–162.
    • (2014) Nat. Rev. Mol. Cell Biol. , vol.15 , Issue.3 , pp. 155-162
    • Shimobayashi, M.1    Hall, M.N.2
  • 224
    • 67349276169 scopus 로고    scopus 로고
    • AMPK regulates energy expenditure by modulating NAD + metabolism and SIRT1 activity
    • Canto, C. et al., AMPK regulates energy expenditure by modulating NAD + metabolism and SIRT1 activity. Nature 458:7241 (2009), 1056–1060.
    • (2009) Nature , vol.458 , Issue.7241 , pp. 1056-1060
    • Canto, C.1
  • 225
    • 25144459980 scopus 로고    scopus 로고
    • The metabolic syndrome—a new worldwide definition
    • Alberti, K.G. et al., The metabolic syndrome—a new worldwide definition. Lancet 366:9491 (2005), 1059–1062.
    • (2005) Lancet , vol.366 , Issue.9491 , pp. 1059-1062
    • Alberti, K.G.1
  • 226
    • 84988345850 scopus 로고    scopus 로고
    • Metabolic control of epigenetics in cancer
    • Kinnaird, A. et al., Metabolic control of epigenetics in cancer. Nat. Rev. Cancer 16:11 (2016), 694–707.
    • (2016) Nat. Rev. Cancer , vol.16 , Issue.11 , pp. 694-707
    • Kinnaird, A.1
  • 227
    • 66249108601 scopus 로고    scopus 로고
    • Understanding the Warburg effect: the metabolic requirements of cell proliferation
    • Vander Heiden, M.G., Cantley, L.C., Thompson, C.B., Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324:5930 (2009), 1029–1033.
    • (2009) Science , vol.324 , Issue.5930 , pp. 1029-1033
    • Vander Heiden, M.G.1    Cantley, L.C.2    Thompson, C.B.3
  • 228
    • 84888009492 scopus 로고    scopus 로고
    • Total lesion glycolysis by 18F-FDG PET/CT is a reliable predictor of prognosis in soft-tissue sarcoma
    • Choi, E.S. et al., Total lesion glycolysis by 18F-FDG PET/CT is a reliable predictor of prognosis in soft-tissue sarcoma. Eur. J. Nucl. Med. Mol. Imaging 40:12 (2013), 1836–1842.
    • (2013) Eur. J. Nucl. Med. Mol. Imaging , vol.40 , Issue.12 , pp. 1836-1842
    • Choi, E.S.1
  • 229
    • 0028073917 scopus 로고
    • Pyruvate dehydrogenase deficiency
    • Brown, G.K. et al., Pyruvate dehydrogenase deficiency. J. Med. Genet. 31:11 (1994), 875–879.
    • (1994) J. Med. Genet. , vol.31 , Issue.11 , pp. 875-879
    • Brown, G.K.1
  • 230
    • 0027381941 scopus 로고
    • Mutations in the glucose-6-phosphatase gene that cause glycogen storage disease type 1a
    • Lei, K.J. et al., Mutations in the glucose-6-phosphatase gene that cause glycogen storage disease type 1a. Science 262:5133 (1993), 580–583.
    • (1993) Science , vol.262 , Issue.5133 , pp. 580-583
    • Lei, K.J.1
  • 231
    • 48249125862 scopus 로고    scopus 로고
    • Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice
    • Cani, P.D. et al., Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 57:6 (2008), 1470–1481.
    • (2008) Diabetes , vol.57 , Issue.6 , pp. 1470-1481
    • Cani, P.D.1
  • 232
    • 33746089859 scopus 로고    scopus 로고
    • Mitochondrial abnormalities and oxidative imbalance in Alzheimer disease
    • Zhu, X. et al., Mitochondrial abnormalities and oxidative imbalance in Alzheimer disease. J. Alzheimers Dis. 9:2 (2006), 147–153.
    • (2006) J. Alzheimers Dis. , vol.9 , Issue.2 , pp. 147-153
    • Zhu, X.1
  • 233
    • 28244445138 scopus 로고    scopus 로고
    • The mitochondrial complex I inhibitor rotenone triggers a cerebral tauopathy
    • Hoglinger, G.U. et al., The mitochondrial complex I inhibitor rotenone triggers a cerebral tauopathy. J. Neurochem. 95:4 (2005), 930–939.
    • (2005) J. Neurochem. , vol.95 , Issue.4 , pp. 930-939
    • Hoglinger, G.U.1
  • 234
    • 19944433571 scopus 로고    scopus 로고
    • Copper-dependent inhibition of human cytochrome c oxidase by a dimeric conformer of amyloid-beta1-42
    • Crouch, P.J. et al., Copper-dependent inhibition of human cytochrome c oxidase by a dimeric conformer of amyloid-beta1-42. J. Neurosci. 25:3 (2005), 672–679.
    • (2005) J. Neurosci. , vol.25 , Issue.3 , pp. 672-679
    • Crouch, P.J.1
  • 235
    • 0035341254 scopus 로고    scopus 로고
    • Mitochondrial abnormalities in Alzheimer's disease
    • Hirai, K. et al., Mitochondrial abnormalities in Alzheimer's disease. J. Neurosci. 21:9 (2001), 3017–3023.
    • (2001) J. Neurosci. , vol.21 , Issue.9 , pp. 3017-3023
    • Hirai, K.1
  • 236
    • 0025024024 scopus 로고
    • Cytochrome oxidase deficiency in Alzheimer's disease
    • Parker, W.D. Jr., Filley, C.M., Parks, J.K., Cytochrome oxidase deficiency in Alzheimer's disease. Neurology 40:8 (1990), 1302–1303.
    • (1990) Neurology , vol.40 , Issue.8 , pp. 1302-1303
    • Parker, W.D.1    Filley, C.M.2    Parks, J.K.3
  • 237
    • 80051665640 scopus 로고    scopus 로고
    • Neuroprotective strategies involving ROS in Alzheimer disease
    • Dumont, M., Beal, M.F., Neuroprotective strategies involving ROS in Alzheimer disease. Free Radic. Biol. Med. 51:5 (2011), 1014–1026.
    • (2011) Free Radic. Biol. Med. , vol.51 , Issue.5 , pp. 1014-1026
    • Dumont, M.1    Beal, M.F.2
  • 238
    • 3943071801 scopus 로고    scopus 로고
    • Sirtuin activators mimic caloric restriction and delay ageing in metazoans
    • Wood, J.G. et al., Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature 430:7000 (2004), 686–689.
    • (2004) Nature , vol.430 , Issue.7000 , pp. 686-689
    • Wood, J.G.1
  • 239
    • 84897524640 scopus 로고    scopus 로고
    • Caloric restriction reduces age-related and all-cause mortality in rhesus monkeys
    • Colman, R.J. et al., Caloric restriction reduces age-related and all-cause mortality in rhesus monkeys. Nat. Commun., 5, 2014, 3557.
    • (2014) Nat. Commun. , vol.5 , pp. 3557
    • Colman, R.J.1
  • 240
    • 67650439330 scopus 로고    scopus 로고
    • Caloric restriction delays disease onset and mortality in rhesus monkeys
    • Colman, R.J. et al., Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 325:5937 (2009), 201–204.
    • (2009) Science , vol.325 , Issue.5937 , pp. 201-204
    • Colman, R.J.1
  • 241
    • 84866163081 scopus 로고    scopus 로고
    • Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study
    • Mattison, J.A. et al., Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature 489:7415 (2012), 318–321.
    • (2012) Nature , vol.489 , Issue.7415 , pp. 318-321
    • Mattison, J.A.1
  • 242
    • 33947264635 scopus 로고    scopus 로고
    • Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer's disease
    • Halagappa, V.K. et al., Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer's disease. Neurobiol. Dis. 26:1 (2007), 212–220.
    • (2007) Neurobiol. Dis. , vol.26 , Issue.1 , pp. 212-220
    • Halagappa, V.K.1
  • 243
    • 84858782079 scopus 로고    scopus 로고
    • AMPK: a nutrient and energy sensor that maintains energy homeostasis
    • Hardie, D.G., Ross, F.A., Hawley, S.A., AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. 13:4 (2012), 251–262.
    • (2012) Nat. Rev. Mol. Cell Biol. , vol.13 , Issue.4 , pp. 251-262
    • Hardie, D.G.1    Ross, F.A.2    Hawley, S.A.3
  • 244
    • 84990876582 scopus 로고    scopus 로고
    • Functional characterization of AMP-activated protein kinase signaling in tumorigenesis
    • Cheng, J. et al., Functional characterization of AMP-activated protein kinase signaling in tumorigenesis. Biochim. Biophys. Acta 1866:2 (2016), 232–251.
    • (2016) Biochim. Biophys. Acta , vol.1866 , Issue.2 , pp. 232-251
    • Cheng, J.1
  • 245
    • 79952135798 scopus 로고    scopus 로고
    • AMP-activated protein kinase (AMPK) is a tau kinase, activated in response to amyloid beta-peptide exposure
    • Thornton, C. et al., AMP-activated protein kinase (AMPK) is a tau kinase, activated in response to amyloid beta-peptide exposure. Biochem. J. 434:3 (2011), 503–512.
    • (2011) Biochem. J. , vol.434 , Issue.3 , pp. 503-512
    • Thornton, C.1
  • 246
    • 79952135316 scopus 로고    scopus 로고
    • AMPK is abnormally activated in tangle- and pre-tangle-bearing neurons in Alzheimer's disease and other tauopathies
    • Vingtdeux, V. et al., AMPK is abnormally activated in tangle- and pre-tangle-bearing neurons in Alzheimer's disease and other tauopathies. Acta Neuropathol. 121:3 (2011), 337–349.
    • (2011) Acta Neuropathol. , vol.121 , Issue.3 , pp. 337-349
    • Vingtdeux, V.1
  • 247
    • 62649115914 scopus 로고    scopus 로고
    • Antidiabetic drug metformin (GlucophageR) increases biogenesis of Alzheimer's amyloid peptides via up-regulating BACE1 transcription
    • Chen, Y. et al., Antidiabetic drug metformin (GlucophageR) increases biogenesis of Alzheimer's amyloid peptides via up-regulating BACE1 transcription. Proc. Natl. Acad. Sci. U. S. A. 106:10 (2009), 3907–3912.
    • (2009) Proc. Natl. Acad. Sci. U. S. A. , vol.106 , Issue.10 , pp. 3907-3912
    • Chen, Y.1
  • 248
    • 79960566233 scopus 로고    scopus 로고
    • AMP-activated protein kinase: a potential player in Alzheimer's disease
    • Salminen, A. et al., AMP-activated protein kinase: a potential player in Alzheimer's disease. J. Neurochem. 118:4 (2011), 460–474.
    • (2011) J. Neurochem. , vol.118 , Issue.4 , pp. 460-474
    • Salminen, A.1
  • 249
    • 84907898151 scopus 로고    scopus 로고
    • Autophagy in microglia degrades extracellular beta-amyloid fibrils and regulates the NLRP3 inflammasome
    • Cho, M.H. et al., Autophagy in microglia degrades extracellular beta-amyloid fibrils and regulates the NLRP3 inflammasome. Autophagy 10:10 (2014), 1761–1775.
    • (2014) Autophagy , vol.10 , Issue.10 , pp. 1761-1775
    • Cho, M.H.1
  • 250
    • 76749098737 scopus 로고    scopus 로고
    • Overview of the immune response
    • Chaplin, D.D., Overview of the immune response. J. Allergy Clin. Immunol. 125:2 Suppl. 2 (2010), S3–23.
    • (2010) J. Allergy Clin. Immunol. , vol.125 , Issue.2 Suppl. 2 , pp. S3-23
    • Chaplin, D.D.1
  • 251
    • 33845866857 scopus 로고    scopus 로고
    • Inflammation and metabolic disorders
    • Hotamisligil, G.S., Inflammation and metabolic disorders. Nature 444:7121 (2006), 860–867.
    • (2006) Nature , vol.444 , Issue.7121 , pp. 860-867
    • Hotamisligil, G.S.1
  • 252
    • 0037180757 scopus 로고    scopus 로고
    • Inflammation and cancer
    • Coussens, L.M., Werb, Z., Inflammation and cancer. Nature 420:6917 (2002), 860–867.
    • (2002) Nature , vol.420 , Issue.6917 , pp. 860-867
    • Coussens, L.M.1    Werb, Z.2
  • 253
    • 65249138393 scopus 로고    scopus 로고
    • Myeloid-derived suppressor cells: linking inflammation and cancer
    • Ostrand-Rosenberg, S., Sinha, P., Myeloid-derived suppressor cells: linking inflammation and cancer. J. Immunol. 182:8 (2009), 4499–4506.
    • (2009) J. Immunol. , vol.182 , Issue.8 , pp. 4499-4506
    • Ostrand-Rosenberg, S.1    Sinha, P.2
  • 254
    • 0029858387 scopus 로고    scopus 로고
    • TNF- and cancer therapy-induced apoptosis: potentiation by inhibition of NF-kappaB
    • Wang, C.Y., Mayo, M.W., Baldwin, A.S. Jr., TNF- and cancer therapy-induced apoptosis: potentiation by inhibition of NF-kappaB. Science 274:5288 (1996), 784–787.
    • (1996) Science , vol.274 , Issue.5288 , pp. 784-787
    • Wang, C.Y.1    Mayo, M.W.2    Baldwin, A.S.3
  • 255
    • 0028234529 scopus 로고
    • Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins
    • Darnell, J.E. Jr., Kerr, I.M., Stark, G.R., Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 264:5164 (1994), 1415–1421.
    • (1994) Science , vol.264 , Issue.5164 , pp. 1415-1421
    • Darnell, J.E.1    Kerr, I.M.2    Stark, G.R.3
  • 256
    • 42649114059 scopus 로고    scopus 로고
    • The IRF family transcription factors in immunity and oncogenesis
    • Tamura, T. et al., The IRF family transcription factors in immunity and oncogenesis. Annu. Rev. Immunol. 26 (2008), 535–584.
    • (2008) Annu. Rev. Immunol. , vol.26 , pp. 535-584
    • Tamura, T.1
  • 257
    • 34249882777 scopus 로고    scopus 로고
    • Foxo3a is essential for maintenance of the hematopoietic stem cell pool
    • Miyamoto, K. et al., Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell 1:1 (2007), 101–112.
    • (2007) Cell Stem Cell , vol.1 , Issue.1 , pp. 101-112
    • Miyamoto, K.1
  • 258
    • 80053399756 scopus 로고    scopus 로고
    • Loss of memory B cells during chronic HIV infection is driven by Foxo3a- and TRAIL-mediated apoptosis
    • van Grevenynghe, J. et al., Loss of memory B cells during chronic HIV infection is driven by Foxo3a- and TRAIL-mediated apoptosis. J. Clin. Invest. 121:10 (2011), 3877–3888.
    • (2011) J. Clin. Invest. , vol.121 , Issue.10 , pp. 3877-3888
    • van Grevenynghe, J.1
  • 259
    • 84887891303 scopus 로고    scopus 로고
    • Kinase inhibitors and immune check-point blockade for the treatment of metastatic melanoma and advanced cancer: synergistic or antagonistic?
    • Luke, J.J., Ott, P.A., Kinase inhibitors and immune check-point blockade for the treatment of metastatic melanoma and advanced cancer: synergistic or antagonistic?. Expert. Opin. Pharmacother. 14:18 (2013), 2457–2462.
    • (2013) Expert. Opin. Pharmacother. , vol.14 , Issue.18 , pp. 2457-2462
    • Luke, J.J.1    Ott, P.A.2
  • 260
    • 84926370173 scopus 로고    scopus 로고
    • Neuroinflammation in Alzheimer's disease
    • Heneka, M.T. et al., Neuroinflammation in Alzheimer's disease. Lancet Neurol. 14:4 (2015), 388–405.
    • (2015) Lancet Neurol. , vol.14 , Issue.4 , pp. 388-405
    • Heneka, M.T.1
  • 261
    • 79251644774 scopus 로고    scopus 로고
    • Whole transcriptome sequencing reveals gene expression and splicing differences in brain regions affected by Alzheimer's disease
    • Twine, N.A. et al., Whole transcriptome sequencing reveals gene expression and splicing differences in brain regions affected by Alzheimer's disease. PLoS One, 6(1), 2011, e16266.
    • (2011) PLoS One , vol.6 , Issue.1
    • Twine, N.A.1
  • 262
    • 84885456046 scopus 로고    scopus 로고
    • The amyloid cascade-inflammatory hypothesis of Alzheimer disease: implications for therapy
    • McGeer, P.L., McGeer, E.G., The amyloid cascade-inflammatory hypothesis of Alzheimer disease: implications for therapy. Acta Neuropathol. 126:4 (2013), 479–497.
    • (2013) Acta Neuropathol. , vol.126 , Issue.4 , pp. 479-497
    • McGeer, P.L.1    McGeer, E.G.2
  • 263
    • 84882665749 scopus 로고    scopus 로고
    • Inflammatory process in Alzheimer's Disease
    • Meraz-Rios, M.A. et al., Inflammatory process in Alzheimer's Disease. Front. Integr. Neurosci., 7, 2013, 59.
    • (2013) Front. Integr. Neurosci. , vol.7 , pp. 59
    • Meraz-Rios, M.A.1
  • 264
    • 84862777666 scopus 로고    scopus 로고
    • ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models
    • Cramer, P.E. et al., ApoE-directed therapeutics rapidly clear beta-amyloid and reverse deficits in AD mouse models. Science 335:6075 (2012), 1503–1506.
    • (2012) Science , vol.335 , Issue.6075 , pp. 1503-1506
    • Cramer, P.E.1
  • 265
    • 0035067311 scopus 로고    scopus 로고
    • Inflammatory responses to amyloidosis in a transgenic mouse model of Alzheimer's disease
    • Matsuoka, Y. et al., Inflammatory responses to amyloidosis in a transgenic mouse model of Alzheimer's disease. Am. J. Pathol. 158:4 (2001), 1345–1354.
    • (2001) Am. J. Pathol. , vol.158 , Issue.4 , pp. 1345-1354
    • Matsuoka, Y.1
  • 266
    • 0037088914 scopus 로고    scopus 로고
    • Microglial activation and beta -amyloid deposit reduction caused by a nitric oxide-releasing nonsteroidal anti-inflammatory drug in amyloid precursor protein plus presenilin-1 transgenic mice
    • Jantzen, P.T. et al., Microglial activation and beta -amyloid deposit reduction caused by a nitric oxide-releasing nonsteroidal anti-inflammatory drug in amyloid precursor protein plus presenilin-1 transgenic mice. J. Neurosci. 22:6 (2002), 2246–2254.
    • (2002) J. Neurosci. , vol.22 , Issue.6 , pp. 2246-2254
    • Jantzen, P.T.1
  • 267
    • 70349323417 scopus 로고    scopus 로고
    • CD14 and toll-like receptors 2 and 4 are required for fibrillar A{beta}-stimulated microglial activation
    • Reed-Geaghan, E.G. et al., CD14 and toll-like receptors 2 and 4 are required for fibrillar A{beta}-stimulated microglial activation. J. Neurosci. 29:38 (2009), 11982–11992.
    • (2009) J. Neurosci. , vol.29 , Issue.38 , pp. 11982-11992
    • Reed-Geaghan, E.G.1
  • 268
    • 33645461302 scopus 로고    scopus 로고
    • NF-kappaB pathway: a target for preventing beta-amyloid (Abeta)-induced neuronal damage and Abeta42 production
    • Valerio, A. et al., NF-kappaB pathway: a target for preventing beta-amyloid (Abeta)-induced neuronal damage and Abeta42 production. Eur. J. Neurosci. 23:7 (2006), 1711–1720.
    • (2006) Eur. J. Neurosci. , vol.23 , Issue.7 , pp. 1711-1720
    • Valerio, A.1
  • 269
    • 0032510692 scopus 로고    scopus 로고
    • Amyloid beta-peptide stimulates nitric oxide production in astrocytes through an NFkappaB-dependent mechanism
    • Akama, K.T. et al., Amyloid beta-peptide stimulates nitric oxide production in astrocytes through an NFkappaB-dependent mechanism. Proc. Natl. Acad. Sci. U. S. A. 95:10 (1998), 5795–5800.
    • (1998) Proc. Natl. Acad. Sci. U. S. A. , vol.95 , Issue.10 , pp. 5795-5800
    • Akama, K.T.1
  • 270
    • 5444257609 scopus 로고    scopus 로고
    • MAPK recruitment by beta-amyloid in organotypic hippocampal slice cultures depends on physical state and exposure time
    • Bell, K.A. et al., MAPK recruitment by beta-amyloid in organotypic hippocampal slice cultures depends on physical state and exposure time. J. Neurochem. 91:2 (2004), 349–361.
    • (2004) J. Neurochem. , vol.91 , Issue.2 , pp. 349-361
    • Bell, K.A.1
  • 271
    • 84905685785 scopus 로고    scopus 로고
    • Modulating neurotoxicity through CX3CL1/CX3CR1 signaling
    • Limatola, C., Ransohoff, R.M., Modulating neurotoxicity through CX3CL1/CX3CR1 signaling. Front. Cell. Neurosci., 8, 2014, 229.
    • (2014) Front. Cell. Neurosci. , vol.8 , pp. 229
    • Limatola, C.1    Ransohoff, R.M.2
  • 272
    • 77957321869 scopus 로고    scopus 로고
    • Regulation of tau pathology by the microglial fractalkine receptor
    • Bhaskar, K. et al., Regulation of tau pathology by the microglial fractalkine receptor. Neuron 68:1 (2010), 19–31.
    • (2010) Neuron , vol.68 , Issue.1 , pp. 19-31
    • Bhaskar, K.1
  • 273
    • 84861078831 scopus 로고    scopus 로고
    • A review: inflammatory process in Alzheimer's disease, role of cytokines
    • Rubio-Perez, J.M., Morillas-Ruiz, J.M., A review: inflammatory process in Alzheimer's disease, role of cytokines. ScientificWorldJournal, 2012, 2012, 756357.
    • (2012) ScientificWorldJournal , vol.2012 , pp. 756357
    • Rubio-Perez, J.M.1    Morillas-Ruiz, J.M.2
  • 274
    • 85015484234 scopus 로고    scopus 로고
    • Role of pro-inflammatory cytokines released from microglia in Alzheimer's disease
    • Wang, W.Y. et al., Role of pro-inflammatory cytokines released from microglia in Alzheimer's disease. Ann. Transl. Med., 3(10), 2015, 136.
    • (2015) Ann. Transl. Med. , vol.3 , Issue.10 , pp. 136
    • Wang, W.Y.1
  • 275
    • 0037440474 scopus 로고    scopus 로고
    • Astrocytic expression of the Alzheimer's disease beta-secretase (BACE1) is stimulus-dependent
    • Hartlage-Rubsamen, M. et al., Astrocytic expression of the Alzheimer's disease beta-secretase (BACE1) is stimulus-dependent. Glia 41:2 (2003), 169–179.
    • (2003) Glia , vol.41 , Issue.2 , pp. 169-179
    • Hartlage-Rubsamen, M.1
  • 276
    • 7244226629 scopus 로고    scopus 로고
    • Non-steroidal anti-inflammatory drugs (NSAIDs) in Alzheimer's disease: old and new mechanisms of action
    • Gasparini, L., Ongini, E., Wenk, G., Non-steroidal anti-inflammatory drugs (NSAIDs) in Alzheimer's disease: old and new mechanisms of action. J. Neurochem. 91:3 (2004), 521–536.
    • (2004) J. Neurochem. , vol.91 , Issue.3 , pp. 521-536
    • Gasparini, L.1    Ongini, E.2    Wenk, G.3
  • 277
    • 0030897133 scopus 로고    scopus 로고
    • Risk of Alzheimer's disease and duration of NSAID use
    • Stewart, W.F. et al., Risk of Alzheimer's disease and duration of NSAID use. Neurology 48:3 (1997), 626–632.
    • (1997) Neurology , vol.48 , Issue.3 , pp. 626-632
    • Stewart, W.F.1
  • 278
    • 0035689690 scopus 로고    scopus 로고
    • Nasal vaccination with beta-amyloid peptide for the treatment of Alzheimer's disease
    • Lemere, C.A. et al., Nasal vaccination with beta-amyloid peptide for the treatment of Alzheimer's disease. DNA Cell Biol. 20:11 (2001), 705–711.
    • (2001) DNA Cell Biol. , vol.20 , Issue.11 , pp. 705-711
    • Lemere, C.A.1
  • 279
    • 84984755327 scopus 로고    scopus 로고
    • A beta peptide vaccination prevents memory loss in an animal model of Alzheimer's disease
    • Morgan, D. et al., A beta peptide vaccination prevents memory loss in an animal model of Alzheimer's disease. Nature 408:6815 (2000), 982–985.
    • (2000) Nature , vol.408 , Issue.6815 , pp. 982-985
    • Morgan, D.1
  • 280
    • 0036240395 scopus 로고    scopus 로고
    • Immunization reverses memory deficits without reducing brain Abeta burden in Alzheimer's disease model
    • Dodart, J.C. et al., Immunization reverses memory deficits without reducing brain Abeta burden in Alzheimer's disease model. Nat. Neurosci. 5:5 (2002), 452–457.
    • (2002) Nat. Neurosci. , vol.5 , Issue.5 , pp. 452-457
    • Dodart, J.C.1
  • 281
    • 85020432663 scopus 로고    scopus 로고
    • miRNAs: novel regulators of autoimmunity-mediated pancreatic beta-cell destruction in type 1 diabetes
    • Zheng, Y., Wang, Z., Zhou, Z., miRNAs: novel regulators of autoimmunity-mediated pancreatic beta-cell destruction in type 1 diabetes. Cell. Mol. Immunol., 2017.
    • (2017) Cell. Mol. Immunol.
    • Zheng, Y.1    Wang, Z.2    Zhou, Z.3
  • 282
    • 84985956947 scopus 로고    scopus 로고
    • Alzheimer's disease: presence and role of microRNAs
    • Basavaraju, M., de Lencastre, A., Alzheimer's disease: presence and role of microRNAs. Biomol. Concepts 7:4 (2016), 241–252.
    • (2016) Biomol. Concepts , vol.7 , Issue.4 , pp. 241-252
    • Basavaraju, M.1    de Lencastre, A.2
  • 283
    • 85007481150 scopus 로고    scopus 로고
    • Getting miRNA therapeutics into the target cells for neurodegenerative diseases: a mini-review
    • Wen, M.M., Getting miRNA therapeutics into the target cells for neurodegenerative diseases: a mini-review. Front. Mol. Neurosci., 9, 2016, 129.
    • (2016) Front. Mol. Neurosci. , vol.9 , pp. 129
    • Wen, M.M.1
  • 284
    • 84890121012 scopus 로고    scopus 로고
    • Alternative polyadenylation and miR-34 family members regulate tau expression
    • Dickson, J.R. et al., Alternative polyadenylation and miR-34 family members regulate tau expression. J. Neurochem. 127:6 (2013), 739–749.
    • (2013) J. Neurochem. , vol.127 , Issue.6 , pp. 739-749
    • Dickson, J.R.1
  • 286
    • 44049108170 scopus 로고    scopus 로고
    • Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer's disease correlates with increased BACE1/beta-secretase expression
    • Hebert, S.S. et al., Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer's disease correlates with increased BACE1/beta-secretase expression. Proc. Natl. Acad. Sci. U. S. A. 105:17 (2008), 6415–6420.
    • (2008) Proc. Natl. Acad. Sci. U. S. A. , vol.105 , Issue.17 , pp. 6415-6420
    • Hebert, S.S.1
  • 287
    • 84928138931 scopus 로고    scopus 로고
    • MicroRNA-29a is a candidate biomarker for Alzheimer's disease in cell-free cerebrospinal fluid
    • Muller, M. et al., MicroRNA-29a is a candidate biomarker for Alzheimer's disease in cell-free cerebrospinal fluid. Mol. Neurobiol. 53:5 (2016), 2894–2899.
    • (2016) Mol. Neurobiol. , vol.53 , Issue.5 , pp. 2894-2899
    • Muller, M.1
  • 288
    • 84875590101 scopus 로고    scopus 로고
    • Long noncoding RNAs: past, present, and future
    • Kung, J.T., Colognori, D., Lee, J.T., Long noncoding RNAs: past, present, and future. Genetics 193:3 (2013), 651–669.
    • (2013) Genetics , vol.193 , Issue.3 , pp. 651-669
    • Kung, J.T.1    Colognori, D.2    Lee, J.T.3
  • 289
    • 85015882308 scopus 로고    scopus 로고
    • Targeting noncoding RNAs in disease
    • Adams, B.D. et al., Targeting noncoding RNAs in disease. J. Clin. Invest. 127:3 (2017), 761–771.
    • (2017) J. Clin. Invest. , vol.127 , Issue.3 , pp. 761-771
    • Adams, B.D.1
  • 290
    • 84959078512 scopus 로고    scopus 로고
    • The role of long noncoding RNAs in neurodegenerative diseases
    • Wan, P., Su, W., Zhuo, Y., The role of long noncoding RNAs in neurodegenerative diseases. Mol. Neurobiol. 54:3 (2017), 2012–2021.
    • (2017) Mol. Neurobiol. , vol.54 , Issue.3 , pp. 2012-2021
    • Wan, P.1    Su, W.2    Zhuo, Y.3
  • 291
    • 85011536754 scopus 로고    scopus 로고
    • LncRNAs: macromolecules with big roles in neurobiology and neurological diseases
    • Chen, Y., Zhou, J., LncRNAs: macromolecules with big roles in neurobiology and neurological diseases. Metab. Brain Dis. 32:2 (2017), 281–291.
    • (2017) Metab. Brain Dis. , vol.32 , Issue.2 , pp. 281-291
    • Chen, Y.1    Zhou, J.2
  • 292
    • 84949952099 scopus 로고    scopus 로고
    • Unique features of long non-coding RNA biogenesis and function
    • Quinn, J.J., Chang, H.Y., Unique features of long non-coding RNA biogenesis and function. Nat. Rev. Genet. 17:1 (2016), 47–62.
    • (2016) Nat. Rev. Genet. , vol.17 , Issue.1 , pp. 47-62
    • Quinn, J.J.1    Chang, H.Y.2
  • 293
    • 80052678741 scopus 로고    scopus 로고
    • Knockdown of BACE1-AS nonprotein-coding transcript modulates beta-amyloid-related hippocampal neurogenesis
    • Modarresi, F. et al., Knockdown of BACE1-AS nonprotein-coding transcript modulates beta-amyloid-related hippocampal neurogenesis. Int. J. Alzheimers Dis., 2011, 2011, 929042.
    • (2011) Int. J. Alzheimers Dis. , vol.2011 , pp. 929042
    • Modarresi, F.1
  • 294
    • 46749083733 scopus 로고    scopus 로고
    • Expression of a noncoding RNA is elevated in Alzheimer's disease and drives rapid feed-forward regulation of beta-secretase
    • Faghihi, M.A. et al., Expression of a noncoding RNA is elevated in Alzheimer's disease and drives rapid feed-forward regulation of beta-secretase. Nat. Med. 14:7 (2008), 723–730.
    • (2008) Nat. Med. , vol.14 , Issue.7 , pp. 723-730
    • Faghihi, M.A.1
  • 295
    • 34547515964 scopus 로고    scopus 로고
    • Dendritic BC200 RNA in aging and in Alzheimer's disease
    • Mus, E., Hof, P.R., Tiedge, H., Dendritic BC200 RNA in aging and in Alzheimer's disease. Proc. Natl. Acad. Sci. U. S. A. 104:25 (2007), 10679–10684.
    • (2007) Proc. Natl. Acad. Sci. U. S. A. , vol.104 , Issue.25 , pp. 10679-10684
    • Mus, E.1    Hof, P.R.2    Tiedge, H.3
  • 296
    • 84879823671 scopus 로고    scopus 로고
    • Roles of long noncoding RNAs in brain development, functional diversification and neurodegenerative diseases
    • Wu, P. et al., Roles of long noncoding RNAs in brain development, functional diversification and neurodegenerative diseases. Brain Res. Bull. 97 (2013), 69–80.
    • (2013) Brain Res. Bull. , vol.97 , pp. 69-80
    • Wu, P.1
  • 297
    • 69349098474 scopus 로고    scopus 로고
    • Epigenetics: definition, mechanisms and clinical perspective
    • Dupont, C., Armant, D.R., Brenner, C.A., Epigenetics: definition, mechanisms and clinical perspective. Semin. Reprod. Med. 27:5 (2009), 351–357.
    • (2009) Semin. Reprod. Med. , vol.27 , Issue.5 , pp. 351-357
    • Dupont, C.1    Armant, D.R.2    Brenner, C.A.3
  • 298
    • 85042330844 scopus 로고    scopus 로고
    • Epigenetics in Alzheimer's disease: perspective of DNA methylation
    • Qazi, T.J. et al., Epigenetics in Alzheimer's disease: perspective of DNA methylation. Mol. Neurobiol., 2017.
    • (2017) Mol. Neurobiol.
    • Qazi, T.J.1
  • 299
    • 84984653344 scopus 로고    scopus 로고
    • The epigenome in Alzheimer's disease: current state and approaches for a new path to gene discovery and understanding disease mechanism
    • Klein, H.U., Bennett, D.A., De Jager, P.L., The epigenome in Alzheimer's disease: current state and approaches for a new path to gene discovery and understanding disease mechanism. Acta Neuropathol. 132:4 (2016), 503–514.
    • (2016) Acta Neuropathol. , vol.132 , Issue.4 , pp. 503-514
    • Klein, H.U.1    Bennett, D.A.2    De Jager, P.L.3
  • 300
    • 84957807598 scopus 로고    scopus 로고
    • Epigenetic alterations in Alzheimer's disease
    • Sanchez-Mut, J.V., Graff, J., Epigenetic alterations in Alzheimer's disease. Front. Behav. Neurosci., 9, 2015, 347.
    • (2015) Front. Behav. Neurosci. , vol.9 , pp. 347
    • Sanchez-Mut, J.V.1    Graff, J.2
  • 301
    • 84938293899 scopus 로고    scopus 로고
    • The epigenetics of aging and neurodegeneration
    • Lardenoije, R. et al., The epigenetics of aging and neurodegeneration. Prog. Neurobiol. 131 (2015), 21–64.
    • (2015) Prog. Neurobiol. , vol.131 , pp. 21-64
    • Lardenoije, R.1
  • 302
    • 84878856081 scopus 로고    scopus 로고
    • Soluble oligomers of amyloid-beta cause changes in redox state, DNA methylation, and gene transcription by inhibiting EAAT3 mediated cysteine uptake
    • Hodgson, N. et al., Soluble oligomers of amyloid-beta cause changes in redox state, DNA methylation, and gene transcription by inhibiting EAAT3 mediated cysteine uptake. J. Alzheimers Dis. 36:1 (2013), 197–209.
    • (2013) J. Alzheimers Dis. , vol.36 , Issue.1 , pp. 197-209
    • Hodgson, N.1
  • 303
    • 80555122780 scopus 로고    scopus 로고
    • Amyloid protein-mediated differential DNA methylation status regulates gene expression in Alzheimer's disease model cell line
    • Sung, H.Y. et al., Amyloid protein-mediated differential DNA methylation status regulates gene expression in Alzheimer's disease model cell line. Biochem. Biophys. Res. Commun. 414:4 (2011), 700–705.
    • (2011) Biochem. Biophys. Res. Commun. , vol.414 , Issue.4 , pp. 700-705
    • Sung, H.Y.1
  • 304
    • 84906716826 scopus 로고    scopus 로고
    • Methylomic profiling implicates cortical deregulation of ANK1 in Alzheimer's disease
    • Lunnon, K. et al., Methylomic profiling implicates cortical deregulation of ANK1 in Alzheimer's disease. Nat. Neurosci. 17:9 (2014), 1164–1170.
    • (2014) Nat. Neurosci. , vol.17 , Issue.9 , pp. 1164-1170
    • Lunnon, K.1
  • 305
    • 84919784465 scopus 로고    scopus 로고
    • Alzheimer's disease: early alterations in brain DNA methylation at ANK1, BIN1, RHBDF2 and other loci
    • De Jager, P.L. et al., Alzheimer's disease: early alterations in brain DNA methylation at ANK1, BIN1, RHBDF2 and other loci. Nat. Neurosci. 17:9 (2014), 1156–1163.
    • (2014) Nat. Neurosci. , vol.17 , Issue.9 , pp. 1156-1163
    • De Jager, P.L.1
  • 306
    • 84939251632 scopus 로고    scopus 로고
    • Pharmacological selectivity within class I histone deacetylases Predicts effects on synaptic function and memory rescue
    • Rumbaugh, G. et al., Pharmacological selectivity within class I histone deacetylases Predicts effects on synaptic function and memory rescue. Neuropsychopharmacology 40:10 (2015), 2307–2316.
    • (2015) Neuropsychopharmacology , vol.40 , Issue.10 , pp. 2307-2316
    • Rumbaugh, G.1
  • 307
    • 84862777943 scopus 로고    scopus 로고
    • An epigenetic blockade of cognitive functions in the neurodegenerating brain
    • Graff, J. et al., An epigenetic blockade of cognitive functions in the neurodegenerating brain. Nature 483:7388 (2012), 222–226.
    • (2012) Nature , vol.483 , Issue.7388 , pp. 222-226
    • Graff, J.1
  • 308
    • 84973128266 scopus 로고    scopus 로고
    • The isomerase PIN1 controls numerous cancer-driving pathways and is a unique drug target
    • Zhou, X.Z., Lu, K.P., The isomerase PIN1 controls numerous cancer-driving pathways and is a unique drug target. Nat. Rev. Cancer 16:7 (2016), 463–478.
    • (2016) Nat. Rev. Cancer , vol.16 , Issue.7 , pp. 463-478
    • Zhou, X.Z.1    Lu, K.P.2
  • 309
    • 84994758800 scopus 로고    scopus 로고
    • PIN1 in breast development and cancer: a clinical perspective
    • Rustighi, A. et al., PIN1 in breast development and cancer: a clinical perspective. Cell Death Differ. 24:2 (2017), 200–211.
    • (2017) Cell Death Differ. , vol.24 , Issue.2 , pp. 200-211
    • Rustighi, A.1
  • 310
    • 84975904934 scopus 로고    scopus 로고
    • Prolyl isomerase Pin1 in cancer
    • Lu, Z., Hunter, T., Prolyl isomerase Pin1 in cancer. Cell Res. 24:9 (2014), 1033–1049.
    • (2014) Cell Res. , vol.24 , Issue.9 , pp. 1033-1049
    • Lu, Z.1    Hunter, T.2
  • 311
    • 84925308777 scopus 로고    scopus 로고
    • Landscape of Pin1 in the cell cycle
    • Lin, C.H. et al., Landscape of Pin1 in the cell cycle. Exp. Biol. Med. (Maywood) 240:3 (2015), 403–408.
    • (2015) Exp. Biol. Med. (Maywood) , vol.240 , Issue.3 , pp. 403-408
    • Lin, C.H.1
  • 312
    • 84924536720 scopus 로고    scopus 로고
    • The emerging role of peptidyl-prolyl isomerase chaperones in tau oligomerization, amyloid processing, and Alzheimer's disease
    • Blair, L.J. et al., The emerging role of peptidyl-prolyl isomerase chaperones in tau oligomerization, amyloid processing, and Alzheimer's disease. J. Neurochem. 133:1 (2015), 1–13.
    • (2015) J. Neurochem. , vol.133 , Issue.1 , pp. 1-13
    • Blair, L.J.1
  • 313
    • 0033600242 scopus 로고    scopus 로고
    • The prolyl isomerase Pin1 restores the function of Alzheimer-associated phosphorylated tau protein
    • Lu, P.J. et al., The prolyl isomerase Pin1 restores the function of Alzheimer-associated phosphorylated tau protein. Nature 399:6738 (1999), 784–788.
    • (1999) Nature , vol.399 , Issue.6738 , pp. 784-788
    • Lu, P.J.1
  • 314
    • 84863230253 scopus 로고    scopus 로고
    • Prolyl isomerase Pin1 promotes amyloid precursor protein (APP) turnover by inhibiting glycogen synthase kinase-3beta (GSK3beta) activity: novel mechanism for Pin1 to protect against Alzheimer disease
    • Ma, S.L. et al., Prolyl isomerase Pin1 promotes amyloid precursor protein (APP) turnover by inhibiting glycogen synthase kinase-3beta (GSK3beta) activity: novel mechanism for Pin1 to protect against Alzheimer disease. J. Biol. Chem. 287:10 (2012), 6969–6973.
    • (2012) J. Biol. Chem. , vol.287 , Issue.10 , pp. 6969-6973
    • Ma, S.L.1
  • 315
    • 84983273959 scopus 로고    scopus 로고
    • Perspectives for treating Alzheimer's disease: a review on promising pharmacological substances
    • Cazarim Mde, S. et al., Perspectives for treating Alzheimer's disease: a review on promising pharmacological substances. Sao Paulo Med. J. 134:4 (2016), 342–354.
    • (2016) Sao Paulo Med. J. , vol.134 , Issue.4 , pp. 342-354
    • Cazarim Mde, S.1
  • 316
    • 85019217957 scopus 로고    scopus 로고
    • Brain aging and neurodegeneration: from a mitochondrial point of view
    • Grimm, A., Eckert, A., Brain aging and neurodegeneration: from a mitochondrial point of view. J. Neurochem., 2017.
    • (2017) J. Neurochem.
    • Grimm, A.1    Eckert, A.2
  • 317
    • 74549205329 scopus 로고    scopus 로고
    • Validation of anti-aging drugs by treating age-related diseases
    • Blagosklonny, M.V., Validation of anti-aging drugs by treating age-related diseases. Aging (Albany NY) 1:3 (2009), 281–288.
    • (2009) Aging (Albany NY) , vol.1 , Issue.3 , pp. 281-288
    • Blagosklonny, M.V.1
  • 318
    • 84874611570 scopus 로고    scopus 로고
    • Rapalogs and mTOR inhibitors as anti-aging therapeutics
    • Lamming, D.W. et al., Rapalogs and mTOR inhibitors as anti-aging therapeutics. J. Clin. Invest. 123:3 (2013), 980–989.
    • (2013) J. Clin. Invest. , vol.123 , Issue.3 , pp. 980-989
    • Lamming, D.W.1
  • 319
    • 84938061050 scopus 로고    scopus 로고
    • How longevity research can lead to therapies for Alzheimer's disease: the rapamycin story
    • Richardson, A. et al., How longevity research can lead to therapies for Alzheimer's disease: the rapamycin story. Exp. Gerontol. 68 (2015), 51–58.
    • (2015) Exp. Gerontol. , vol.68 , pp. 51-58
    • Richardson, A.1
  • 320
    • 84959010623 scopus 로고    scopus 로고
    • Alzheimer's disease: an overview of amyloid beta dependent pathogenesis and its therapeutic implications along with in silico approaches emphasizing the role of natural products
    • Awasthi, M. et al., Alzheimer's disease: an overview of amyloid beta dependent pathogenesis and its therapeutic implications along with in silico approaches emphasizing the role of natural products. J. Neurol. Sci. 361 (2016), 256–271.
    • (2016) J. Neurol. Sci. , vol.361 , pp. 256-271
    • Awasthi, M.1
  • 321
    • 84859429591 scopus 로고    scopus 로고
    • Sirtuins in cognitive ageing and Alzheimer's disease
    • Braidy, N. et al., Sirtuins in cognitive ageing and Alzheimer's disease. Curr. Opin. Psychiatry 25:3 (2012), 226–230.
    • (2012) Curr. Opin. Psychiatry , vol.25 , Issue.3 , pp. 226-230
    • Braidy, N.1
  • 322
    • 78650902065 scopus 로고    scopus 로고
    • Neuroprotective properties of resveratrol in different neurodegenerative disorders
    • Albani, D. et al., Neuroprotective properties of resveratrol in different neurodegenerative disorders. Biofactors 36:5 (2010), 370–376.
    • (2010) Biofactors , vol.36 , Issue.5 , pp. 370-376
    • Albani, D.1
  • 323
    • 85019707192 scopus 로고    scopus 로고
    • Drugs in clinical trials for Alzheimer's disease: the major trends
    • Bachurin, S.O., Bovina, E.V., Ustyugov, A.A., Drugs in clinical trials for Alzheimer's disease: the major trends. Med. Res. Rev., 2017.
    • (2017) Med. Res. Rev.
    • Bachurin, S.O.1    Bovina, E.V.2    Ustyugov, A.A.3
  • 324
    • 84994492966 scopus 로고    scopus 로고
    • The BACE1 inhibitor verubecestat (MK-8931) reduces CNS beta-amyloid in animal models and in Alzheimer's disease patients
    • Kennedy, M.E. et al., The BACE1 inhibitor verubecestat (MK-8931) reduces CNS beta-amyloid in animal models and in Alzheimer's disease patients. Sci. Transl. Med., 8(363), 2016, 363ra150.
    • (2016) Sci. Transl. Med. , vol.8 , Issue.363 , pp. 363ra150
    • Kennedy, M.E.1
  • 325
    • 85016405305 scopus 로고    scopus 로고
    • Merck ends trial of potential Alzheimer's drug verubecestat
    • Hawkes, N., Merck ends trial of potential Alzheimer's drug verubecestat. BMJ, 356, 2017, j845.
    • (2017) BMJ , vol.356 , pp. j845
    • Hawkes, N.1
  • 326
    • 85005978119 scopus 로고    scopus 로고
    • AZD3293: pharmacokinetic and pharmacodynamic effects in healthy subjects and patients with Alzheimer's disease
    • Cebers, G. et al., AZD3293: pharmacokinetic and pharmacodynamic effects in healthy subjects and patients with Alzheimer's disease. J. Alzheimers Dis. 55:3 (2017), 1039–1053.
    • (2017) J. Alzheimers Dis. , vol.55 , Issue.3 , pp. 1039-1053
    • Cebers, G.1
  • 327
    • 85019087699 scopus 로고    scopus 로고
    • Clinical activity of the gamma-secretase inhibitor PF-03084014 in adults with Desmoid tumors (aggressive fibromatosis)
    • Kummar, S. et al., Clinical activity of the gamma-secretase inhibitor PF-03084014 in adults with Desmoid tumors (aggressive fibromatosis). J. Clin. Oncol., 2017, JCO2016711994.
    • (2017) J. Clin. Oncol.
    • Kummar, S.1
  • 328
    • 84989350415 scopus 로고    scopus 로고
    • A Phase 1 study of the novel gamma-secretase inhibitor PF-03084014 in patients with T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma
    • Papayannidis, C. et al., A Phase 1 study of the novel gamma-secretase inhibitor PF-03084014 in patients with T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma. Blood Cancer J., 5, 2015, e350.
    • (2015) Blood Cancer J. , vol.5
    • Papayannidis, C.1
  • 329
    • 84958582155 scopus 로고    scopus 로고
    • Validating Immunotherapy in Alzheimer's disease: The EB101 Vaccine
    • Carrera, I. et al., Validating Immunotherapy in Alzheimer's disease: The EB101 Vaccine. Curr. Pharm. Des. 22:7 (2016), 849–858.
    • (2016) Curr. Pharm. Des. , vol.22 , Issue.7 , pp. 849-858
    • Carrera, I.1
  • 330
    • 85020894520 scopus 로고    scopus 로고
    • Amyloid-beta immunotherapy: the hope for Alzheimer disease?
    • Barrera-Ocampo, A., Lopera, F., Amyloid-beta immunotherapy: the hope for Alzheimer disease?. Colomb. Med. 47:4 (2016), 203–212.
    • (2016) Colomb. Med. , vol.47 , Issue.4 , pp. 203-212
    • Barrera-Ocampo, A.1    Lopera, F.2
  • 331
    • 84949092821 scopus 로고    scopus 로고
    • Decreased amyloid-beta and increased neuronal hyperactivity by immunotherapy in Alzheimer's models
    • Busche, M.A. et al., Decreased amyloid-beta and increased neuronal hyperactivity by immunotherapy in Alzheimer's models. Nat. Neurosci. 18:12 (2015), 1725–1727.
    • (2015) Nat. Neurosci. , vol.18 , Issue.12 , pp. 1725-1727
    • Busche, M.A.1
  • 332
    • 84957439174 scopus 로고    scopus 로고
    • Tau-directed immunotherapy: a promising strategy for treating Alzheimer's disease and other tauopathies
    • Schroeder, S.K. et al., Tau-directed immunotherapy: a promising strategy for treating Alzheimer's disease and other tauopathies. J. NeuroImmune Pharmacol. 11:1 (2016), 9–25.
    • (2016) J. NeuroImmune Pharmacol. , vol.11 , Issue.1 , pp. 9-25
    • Schroeder, S.K.1
  • 333
    • 84982860160 scopus 로고    scopus 로고
    • Tau-centric targets and drugs in clinical development for the treatment of Alzheimer's disease
    • Panza, F. et al., Tau-centric targets and drugs in clinical development for the treatment of Alzheimer's disease. Biomed. Res. Int., 2016, 2016, 3245935.
    • (2016) Biomed. Res. Int. , vol.2016 , pp. 3245935
    • Panza, F.1
  • 334
    • 84926354826 scopus 로고    scopus 로고
    • Treatment for Alzheimer's disease: has anything changed?
    • Waite, L.M., Treatment for Alzheimer's disease: has anything changed?. Aust. Prescr. 38:2 (2015), 60–63.
    • (2015) Aust. Prescr. , vol.38 , Issue.2 , pp. 60-63
    • Waite, L.M.1
  • 335
    • 84925758089 scopus 로고    scopus 로고
    • Phosphodiesterase inhibitors as a target for cognition enhancement in aging and Alzheimer's disease: a translational overview
    • Heckman, P.R., Wouters, C., Prickaerts, J., Phosphodiesterase inhibitors as a target for cognition enhancement in aging and Alzheimer's disease: a translational overview. Curr. Pharm. Des. 21:3 (2015), 317–331.
    • (2015) Curr. Pharm. Des. , vol.21 , Issue.3 , pp. 317-331
    • Heckman, P.R.1    Wouters, C.2    Prickaerts, J.3
  • 336
    • 84938528109 scopus 로고    scopus 로고
    • The role of Cdk5 in Alzheimer's disease
    • Liu, S.L. et al., The role of Cdk5 in Alzheimer's disease. Mol. Neurobiol. 53:7 (2016), 4328–4342.
    • (2016) Mol. Neurobiol. , vol.53 , Issue.7 , pp. 4328-4342
    • Liu, S.L.1
  • 337
    • 84935004225 scopus 로고    scopus 로고
    • Synthetic and natural inhibitors of phospholipases A2: their importance for understanding and treatment of neurological disorders
    • Ong, W.Y. et al., Synthetic and natural inhibitors of phospholipases A2: their importance for understanding and treatment of neurological disorders. ACS Chem. Neurosci. 6:6 (2015), 814–831.
    • (2015) ACS Chem. Neurosci. , vol.6 , Issue.6 , pp. 814-831
    • Ong, W.Y.1
  • 338
    • 0028985574 scopus 로고
    • Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor protein
    • Games, D. et al., Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor protein. Nature 373:6514 (1995), 523–527.
    • (1995) Nature , vol.373 , Issue.6514 , pp. 523-527
    • Games, D.1
  • 339
    • 0011444914 scopus 로고    scopus 로고
    • Two amyloid precursor protein transgenic mouse models with Alzheimer disease-like pathology
    • Sturchler-Pierrat, C. et al., Two amyloid precursor protein transgenic mouse models with Alzheimer disease-like pathology. Proc. Natl. Acad. Sci. U. S. A. 94:24 (1997), 13287–13292.
    • (1997) Proc. Natl. Acad. Sci. U. S. A. , vol.94 , Issue.24 , pp. 13287-13292
    • Sturchler-Pierrat, C.1
  • 340
    • 0034213718 scopus 로고    scopus 로고
    • High-level neuronal expression of abeta 1–42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation
    • Mucke, L. et al., High-level neuronal expression of abeta 1–42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J. Neurosci. 20:11 (2000), 4050–4058.
    • (2000) J. Neurosci. , vol.20 , Issue.11 , pp. 4050-4058
    • Mucke, L.1
  • 341
    • 0034700471 scopus 로고    scopus 로고
    • A beta peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer's disease
    • Janus, C. et al., A beta peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer's disease. Nature 408:6815 (2000), 979–982.
    • (2000) Nature , vol.408 , Issue.6815 , pp. 979-982
    • Janus, C.1
  • 342
    • 0035889404 scopus 로고    scopus 로고
    • Early formation of mature amyloid-beta protein deposits in a mutant APP transgenic model depends on levels of Abeta(1–42)
    • Rockenstein, E. et al., Early formation of mature amyloid-beta protein deposits in a mutant APP transgenic model depends on levels of Abeta(1–42). J. Neurosci. Res. 66:4 (2001), 573–582.
    • (2001) J. Neurosci. Res. , vol.66 , Issue.4 , pp. 573-582
    • Rockenstein, E.1
  • 343
    • 16044366039 scopus 로고    scopus 로고
    • Increased amyloid-beta42(43) in brains of mice expressing mutant presenilin 1
    • Duff, K. et al., Increased amyloid-beta42(43) in brains of mice expressing mutant presenilin 1. Nature 383:6602 (1996), 710–713.
    • (1996) Nature , vol.383 , Issue.6602 , pp. 710-713
    • Duff, K.1
  • 344
    • 0041803006 scopus 로고    scopus 로고
    • Hyperphosphorylation and aggregation of tau in mice expressing normal human tau isoforms
    • Andorfer, C. et al., Hyperphosphorylation and aggregation of tau in mice expressing normal human tau isoforms. J. Neurochem. 86:3 (2003), 582–590.
    • (2003) J. Neurochem. , vol.86 , Issue.3 , pp. 582-590
    • Andorfer, C.1
  • 345
    • 2442642836 scopus 로고    scopus 로고
    • Retarded axonal transport of R406W mutant tau in transgenic mice with a neurodegenerative tauopathy
    • Zhang, B. et al., Retarded axonal transport of R406W mutant tau in transgenic mice with a neurodegenerative tauopathy. J. Neurosci. 24:19 (2004), 4657–4667.
    • (2004) J. Neurosci. , vol.24 , Issue.19 , pp. 4657-4667
    • Zhang, B.1
  • 346
    • 0036850725 scopus 로고    scopus 로고
    • Abundant tau filaments and nonapoptotic neurodegeneration in transgenic mice expressing human P301S tau protein
    • Allen, B. et al., Abundant tau filaments and nonapoptotic neurodegeneration in transgenic mice expressing human P301S tau protein. J. Neurosci. 22:21 (2002), 9340–9351.
    • (2002) J. Neurosci. , vol.22 , Issue.21 , pp. 9340-9351
    • Allen, B.1
  • 347
    • 22344438508 scopus 로고    scopus 로고
    • Tau suppression in a neurodegenerative mouse model improves memory function
    • Santacruz, K. et al., Tau suppression in a neurodegenerative mouse model improves memory function. Science 309:5733 (2005), 476–481.
    • (2005) Science , vol.309 , Issue.5733 , pp. 476-481
    • Santacruz, K.1
  • 348
    • 0031278270 scopus 로고    scopus 로고
    • Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition
    • Bales, K.R. et al., Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition. Nat. Genet. 17:3 (1997), 263–264.
    • (1997) Nat. Genet. , vol.17 , Issue.3 , pp. 263-264
    • Bales, K.R.1
  • 349
    • 12844273475 scopus 로고    scopus 로고
    • Gene delivery of human apolipoprotein E alters brain Abeta burden in a mouse model of Alzheimer's disease
    • Dodart, J.C. et al., Gene delivery of human apolipoprotein E alters brain Abeta burden in a mouse model of Alzheimer's disease. Proc. Natl. Acad. Sci. U. S. A. 102:4 (2005), 1211–1216.
    • (2005) Proc. Natl. Acad. Sci. U. S. A. , vol.102 , Issue.4 , pp. 1211-1216
    • Dodart, J.C.1
  • 350
    • 0043069764 scopus 로고    scopus 로고
    • Role of the prolyl isomerase Pin1 in protecting against age-dependent neurodegeneration
    • Liou, Y.C. et al., Role of the prolyl isomerase Pin1 in protecting against age-dependent neurodegeneration. Nature 424:6948 (2003), 556–561.
    • (2003) Nature , vol.424 , Issue.6948 , pp. 556-561
    • Liou, Y.C.1
  • 351
    • 0030833055 scopus 로고    scopus 로고
    • Accelerated amyloid deposition in the brains of transgenic mice coexpressing mutant presenilin 1 and amyloid precursor proteins
    • Borchelt, D.R. et al., Accelerated amyloid deposition in the brains of transgenic mice coexpressing mutant presenilin 1 and amyloid precursor proteins. Neuron 19:4 (1997), 939–945.
    • (1997) Neuron , vol.19 , Issue.4 , pp. 939-945
    • Borchelt, D.R.1
  • 352
    • 17944382037 scopus 로고    scopus 로고
    • Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP
    • Lewis, J. et al., Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 293:5534 (2001), 1487–1491.
    • (2001) Science , vol.293 , Issue.5534 , pp. 1487-1491
    • Lewis, J.1
  • 353
    • 84914706406 scopus 로고    scopus 로고
    • Human ApoE epsilon4 alters circadian rhythm activity, IL-1beta, and GFAP in CRND8 mice
    • Graybeal, J.J. et al., Human ApoE epsilon4 alters circadian rhythm activity, IL-1beta, and GFAP in CRND8 mice. J. Alzheimers Dis. 43:3 (2015), 823–834.
    • (2015) J. Alzheimers Dis. , vol.43 , Issue.3 , pp. 823-834
    • Graybeal, J.J.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.