메뉴 건너뛰기




Volumn 5, Issue OCT, 2014, Pages

Insulin in the brain: Its pathophysiological implications for states related with central insulin resistance, type 2 diabetes and alzheimer's disease

Author keywords

Alzheimer's disease; Biological actions; Brain; Central insulin resistance; Insulin; Pathophysiological implications; Receptors; Type 2 diabetes

Indexed keywords

C PEPTIDE; GASTRIC INHIBITORY POLYPEPTIDE; GLUCAGON LIKE PEPTIDE 1; GLUCOSE TRANSPORTER; IMMUNOREACTIVE INSULIN; INSULIN; METFORMIN; PEROXISOME PROLIFERATOR ACTIVATED RECEPTOR GAMMA AGONIST;

EID: 84922693194     PISSN: None     EISSN: 16642392     Source Type: Journal    
DOI: 10.3389/fendo.2014.00161     Document Type: Review
Times cited : (393)

References (292)
  • 1
    • 84877794459 scopus 로고    scopus 로고
    • Insulin and the brain
    • Derakhshan F, Toth C. Insulin and the brain. Curr Diabetes Rev (2013) 9(2):102-16. doi: 10.2174/1573399811309020002
    • (2013) Curr Diabetes Rev , vol.9 , Issue.2 , pp. 102-116
    • Derakhshan, F.1    Toth, C.2
  • 3
    • 0018773152 scopus 로고
    • Concentrations of insulin and insulin receptors in the brain are independent of peripheral insulin levels. Studies of obese and streptozotocin-treated rodents
    • Havrankova J, Roth J, Brownstein MJ. Concentrations of insulin and insulin receptors in the brain are independent of peripheral insulin levels. Studies of obese and streptozotocin-treated rodents. J Clin Invest (1979) 64(2):636-42. doi:10.1172/JCI109504
    • (1979) J Clin Invest , vol.64 , Issue.2 , pp. 636-642
    • Havrankova, J.1    Roth, J.2    Brownstein, M.J.3
  • 5
    • 0033391691 scopus 로고    scopus 로고
    • Blood-brain barrier biology and methodology
    • Pardridge WM. Blood-brain barrier biology and methodology. J Neurovirol (1999) 5(6):556-69. doi:10.3109/13550289909021285
    • (1999) J Neurovirol , vol.5 , Issue.6 , pp. 556-569
    • Pardridge, W.M.1
  • 6
    • 0030664286 scopus 로고    scopus 로고
    • Transport of insulin across the blood-brain barrier: saturability at euglycemic doses of insulin
    • Banks WA, Jaspan JB, Huang W, Kastin AJ. Transport of insulin across the blood-brain barrier: saturability at euglycemic doses of insulin. Peptides (1997) 18(9):1423-9. doi:10.1016/S0196-9781(97)00231-3
    • (1997) Peptides , vol.18 , Issue.9 , pp. 1423-1429
    • Banks, W.A.1    Jaspan, J.B.2    Huang, W.3    Kastin, A.J.4
  • 7
    • 0014214923 scopus 로고
    • Insulin in the cerebrospinal fluid
    • Margolis RU, Altszuler N. Insulin in the cerebrospinal fluid. Nature (1967) 215(5108):1375-6. doi:10.1038/2151375a0
    • (1967) Nature , vol.215 , Issue.5108 , pp. 1375-1376
    • Margolis, R.U.1    Altszuler, N.2
  • 8
    • 0017745414 scopus 로고
    • Relationship between plasma and cerebrospinal fluid insulin levels of dogs
    • Woods SC, Porte D Jr. Relationship between plasma and cerebrospinal fluid insulin levels of dogs. Am J Physiol (1977) 233(4):E331-4.
    • (1977) Am J Physiol , vol.233 , Issue.4 , pp. E331-E334
    • Woods, S.C.1    Porte Jr, D.2
  • 9
    • 0023636719 scopus 로고
    • Blood-brain barrier transcytosis of insulin in developing rabbits
    • Duffy KR, Pardridge WM. Blood-brain barrier transcytosis of insulin in developing rabbits. Brain Res (1987) 420(1):32-8. doi:10.1016/0006-8993(87)90236-8
    • (1987) Brain Res , vol.420 , Issue.1 , pp. 32-38
    • Duffy, K.R.1    Pardridge, W.M.2
  • 10
    • 0242351798 scopus 로고    scopus 로고
    • Cholesterol modulates amyloid beta-peptide's membrane interactions
    • Eckert GP, Kirsch C, Leutz S, Wood WG, Muller WE. Cholesterol modulates amyloid beta-peptide's membrane interactions. Pharmacopsychiatry (2003) 36(Suppl 2):S136-43. doi:10.1055/s-2003-43059
    • (2003) Pharmacopsychiatry , vol.36 , pp. S136-S143
    • Eckert, G.P.1    Kirsch, C.2    Leutz, S.3    Wood, W.G.4    Muller, W.E.5
  • 11
    • 0027361094 scopus 로고
    • Saturable transport of insulin from plasma into the central nervous system of dogs in vivo. A mechanism for regulated insulin delivery to the brain
    • Baura GD, Foster DM, Porte D Jr, Kahn SE, Bergman RN, Cobelli C, et al. Saturable transport of insulin from plasma into the central nervous system of dogs in vivo. A mechanism for regulated insulin delivery to the brain. J Clin Invest (1993) 92(4):1824-30. doi:10.1172/JCI116773
    • (1993) J Clin Invest , vol.92 , Issue.4 , pp. 1824-1830
    • Baura, G.D.1    Foster, D.M.2    Porte Jr, D.3    Kahn, S.E.4    Bergman, R.N.5    Cobelli, C.6
  • 12
    • 0031837461 scopus 로고    scopus 로고
    • Differential permeability of the blood-brain barrier to two pancreatic peptides: insulin and amylin
    • Banks WA, Kastin AJ. Differential permeability of the blood-brain barrier to two pancreatic peptides: insulin and amylin. Peptides (1998) 19(5):883-9. doi:10.1016/S0196-9781(98)00018-7
    • (1998) Peptides , vol.19 , Issue.5 , pp. 883-889
    • Banks, W.A.1    Kastin, A.J.2
  • 13
    • 0030069591 scopus 로고    scopus 로고
    • Insulin transport from plasma into the central nervous system is inhibited by dexamethasone in dogs
    • Baura GD, Foster DM, Kaiyala K, Porte D Jr, Kahn SE, Schwartz MW. Insulin transport from plasma into the central nervous system is inhibited by dexamethasone in dogs. Diabetes (1996) 45(1):86-90. doi:10.2337/diab.45.1.86
    • (1996) Diabetes , vol.45 , Issue.1 , pp. 86-90
    • Baura, G.D.1    Foster, D.M.2    Kaiyala, K.3    Porte Jr, D.4    Kahn, S.E.5    Schwartz, M.W.6
  • 14
    • 0023722820 scopus 로고
    • Insulin responses and glucose levels in plasma and cerebrospinal fluid during fasting and refeeding in the rat
    • Strubbe JH, Porte D Jr, Woods SC. Insulin responses and glucose levels in plasma and cerebrospinal fluid during fasting and refeeding in the rat. Physiol Behav (1988) 44(2):205-8. doi:10.1016/0031-9384(88)90139-4
    • (1988) Physiol Behav , vol.44 , Issue.2 , pp. 205-208
    • Strubbe, J.H.1    Porte Jr, D.2    Woods, S.C.3
  • 15
    • 0033857975 scopus 로고    scopus 로고
    • Obesity induced by a high-fat diet is associated with reduced brain insulin transport in dogs
    • Kaiyala KJ, Prigeon RL, Kahn SE, Woods SC, Schwartz MW. Obesity induced by a high-fat diet is associated with reduced brain insulin transport in dogs. Diabetes (2000) 49(9):1525-33. doi:10.2337/diabetes.49.9.1525
    • (2000) Diabetes , vol.49 , Issue.9 , pp. 1525-1533
    • Kaiyala, K.J.1    Prigeon, R.L.2    Kahn, S.E.3    Woods, S.C.4    Schwartz, M.W.5
  • 16
    • 0025911235 scopus 로고
    • Seasonal changes in CSF insulin levels in marmots: insulin may not be a satiety signal for fasting in winter
    • Florant GL, Richardson RD, Mahan S, Singer L, Woods SC. Seasonal changes in CSF insulin levels in marmots: insulin may not be a satiety signal for fasting in winter. Am J Physiol (1991) 260(4 Pt 2):R712-6.
    • (1991) Am J Physiol , vol.260 , Issue.4 , pp. R712-R716
    • Florant, G.L.1    Richardson, R.D.2    Mahan, S.3    Singer, L.4    Woods, S.C.5
  • 17
    • 84865575373 scopus 로고    scopus 로고
    • Insulin in the brain: there and back again
    • Banks WA, Owen JB, Erickson MA. Insulin in the brain: there and back again. Pharmacol Ther (2012) 136(1):82-93. doi:10.1016/j.pharmthera.2012.07.006
    • (2012) Pharmacol Ther , vol.136 , Issue.1 , pp. 82-93
    • Banks, W.A.1    Owen, J.B.2    Erickson, M.A.3
  • 18
    • 84872501218 scopus 로고    scopus 로고
    • Insulin in the brain: sources, localization and functions
    • Ghasemi R, Haeri A, Dargahi L, Mohamed Z, Ahmadiani A. Insulin in the brain: sources, localization and functions. Mol Neurobiol (2013) 47(1):145-71. doi:10.1007/s12035-012-8339-9
    • (2013) Mol Neurobiol , vol.47 , Issue.1 , pp. 145-171
    • Ghasemi, R.1    Haeri, A.2    Dargahi, L.3    Mohamed, Z.4    Ahmadiani, A.5
  • 19
    • 0021088485 scopus 로고
    • Insulin and C-peptide in human brain neurons (insulin/C-peptide/brain peptides/immunohistochemistry/radioimmunoassay)
    • Dorn A, Rinne A, Bernstein HG, Hahn HJ, Ziegler M. Insulin and C-peptide in human brain neurons (insulin/C-peptide/brain peptides/immunohistochemistry/radioimmunoassay). J Hirnforsch (1983) 24(5):495-9.
    • (1983) J Hirnforsch , vol.24 , Issue.5 , pp. 495-499
    • Dorn, A.1    Rinne, A.2    Bernstein, H.G.3    Hahn, H.J.4    Ziegler, M.5
  • 20
    • 0022296020 scopus 로고
    • C-peptide-like material in rat brain: response to fasting and glucose ingestion
    • Jezova D, Vigas M, Sadlon J. C-peptide-like material in rat brain: response to fasting and glucose ingestion. Endocrinol Exp (1985) 19(4):261-6.
    • (1985) Endocrinol Exp , vol.19 , Issue.4 , pp. 261-266
    • Jezova, D.1    Vigas, M.2    Sadlon, J.3
  • 21
  • 22
    • 0022976762 scopus 로고
    • Periventricular hypothalamic cells in the rat brain contain insulin mRNA
    • Young WS III. Periventricular hypothalamic cells in the rat brain contain insulin mRNA. Neuropeptides (1986) 8(2):93-7. doi:10.1016/0143-4179(86)90035-1
    • (1986) Neuropeptides , vol.8 , Issue.2 , pp. 93-97
    • Young W.S, I.I.I.1
  • 25
    • 0020623935 scopus 로고
    • Localization of insulin-like immunoreactivity in the neurons from primary cultures of rat brain
    • Raizada MK. Localization of insulin-like immunoreactivity in the neurons from primary cultures of rat brain. Exp Cell Res (1983) 143(2):351-7. doi:10.1016/0014-4827(83)90061-7
    • (1983) Exp Cell Res , vol.143 , Issue.2 , pp. 351-357
    • Raizada, M.K.1
  • 26
    • 0021264577 scopus 로고
    • Proinsulin-like material in mouse foetal brain cell cultures
    • Birch NP, Christie DL, Renwick AG. Proinsulin-like material in mouse foetal brain cell cultures. FEBS Lett (1984) 168(2):299-302. doi:10.1016/0014-5793(84)80266-5
    • (1984) FEBS Lett , vol.168 , Issue.2 , pp. 299-302
    • Birch, N.P.1    Christie, D.L.2    Renwick, A.G.3
  • 27
    • 0028179038 scopus 로고
    • Immunohistochemical and in situ hybridization study of an insulin-like substance in fetal neuron cell cultures
    • Schechter R, Whitmire J, Wheet GS, Beju D, Jackson KW, Harlow R, et al. Immunohistochemical and in situ hybridization study of an insulin-like substance in fetal neuron cell cultures. Brain Res (1994) 636(1):9-27. doi:10.1016/0006-8993(94)90170-8
    • (1994) Brain Res , vol.636 , Issue.1 , pp. 9-27
    • Schechter, R.1    Whitmire, J.2    Wheet, G.S.3    Beju, D.4    Jackson, K.W.5    Harlow, R.6
  • 28
    • 0037303635 scopus 로고    scopus 로고
    • Brain insulin: regulation, mechanisms of action and functions
    • Gerozissis K. Brain insulin: regulation, mechanisms of action and functions. Cell Mol Neurobiol (2003) 23(1):1-25. doi:10.1023/A:1025021529347
    • (2003) Cell Mol Neurobiol , vol.23 , Issue.1 , pp. 1-25
    • Gerozissis, K.1
  • 29
    • 0022516633 scopus 로고
    • Insulin is released from rat brain neuronal cells in culture
    • Clarke DW, Mudd L, Boyd FT Jr, Fields M, Raizada MK. Insulin is released from rat brain neuronal cells in culture. J Neurochem (1986) 47(3):831-6. doi:10.1111/j.1471-4159.1986.tb00686.x
    • (1986) J Neurochem , vol.47 , Issue.3 , pp. 831-836
    • Clarke, D.W.1    Mudd, L.2    Boyd F.T, Jr.3    Fields, M.4    Raizada, M.K.5
  • 30
    • 0025217801 scopus 로고
    • Release of immunoreactive insulin from rat brain synaptosomes under depolarizing conditions
    • Wei LT, Matsumoto H, Rhoads DE. Release of immunoreactive insulin from rat brain synaptosomes under depolarizing conditions. J Neurochem (1990) 54(5):1661-5. doi:10.1111/j.1471-4159.1990.tb01219.x
    • (1990) J Neurochem , vol.54 , Issue.5 , pp. 1661-1665
    • Wei, L.T.1    Matsumoto, H.2    Rhoads, D.E.3
  • 31
    • 0032956405 scopus 로고    scopus 로고
    • Stimulation of immunoreactive insulin release by glucose in rat brain synaptosomes
    • Santos MS, Pereira EM, Carvaho AP. Stimulation of immunoreactive insulin release by glucose in rat brain synaptosomes. Neurochem Res (1999) 24(1):33-6. doi:10.1023/A:1020971812098
    • (1999) Neurochem Res , vol.24 , Issue.1 , pp. 33-36
    • Santos, M.S.1    Pereira, E.M.2    Carvaho, A.P.3
  • 32
    • 42249107130 scopus 로고    scopus 로고
    • Brain insulin, energy and glucose homeostasis; genes, environment and metabolic pathologies
    • Gerozissis K. Brain insulin, energy and glucose homeostasis; genes, environment and metabolic pathologies. Eur J Pharmacol (2008) 585(1):38-49. doi:10.1016/j.ejphar.2008.01.050
    • (2008) Eur J Pharmacol , vol.585 , Issue.1 , pp. 38-49
    • Gerozissis, K.1
  • 33
    • 84857410355 scopus 로고    scopus 로고
    • Differential insulin receptor substrate-1 (IRS1)-related modulation of neuropeptide Y and proopiomelanocortin expression in nondiabetic and diabetic IRS2-/- mice
    • Burgos-Ramos E, Gonzalez-Rodriguez A, Canelles S, Baquedano E, Frago LM, Revuelta-Cervantes J, et al. Differential insulin receptor substrate-1 (IRS1)-related modulation of neuropeptide Y and proopiomelanocortin expression in nondiabetic and diabetic IRS2-/- mice. Endocrinology (2012) 153(3):1129-40. doi:10.1210/en.2011-1278
    • (2012) Endocrinology , vol.153 , Issue.3 , pp. 1129-1140
    • Burgos-Ramos, E.1    Gonzalez-Rodriguez, A.2    Canelles, S.3    Baquedano, E.4    Frago, L.M.5    Revuelta-Cervantes, J.6
  • 34
    • 0027967822 scopus 로고
    • Identification and distribution of insulin receptors on cultured bovine brain microvessel endothelial cells: possible function in insulin processing in the blood-brain barrier
    • Miller DW, Keller BT, Borchardt RT. Identification and distribution of insulin receptors on cultured bovine brain microvessel endothelial cells: possible function in insulin processing in the blood-brain barrier. J Cell Physiol (1994) 161(2):333-41. doi:10.1002/jcp.1041610218
    • (1994) J Cell Physiol , vol.161 , Issue.2 , pp. 333-341
    • Miller, D.W.1    Keller, B.T.2    Borchardt, R.T.3
  • 35
    • 0016908973 scopus 로고
    • Possible role of insulin in the transport of tyrosine and tryptophan from blood to brain
    • Tagliamonte A, DeMontis MG, Olianas M, Onali PL, Gessa GL. Possible role of insulin in the transport of tyrosine and tryptophan from blood to brain. Adv Exp Med Biol (1976) 69:89-94. doi:10.1007/978-1-4684-3264-0_7
    • (1976) Adv Exp Med Biol , vol.69 , pp. 89-94
    • Tagliamonte, A.1    DeMontis, M.G.2    Olianas, M.3    Onali, P.L.4    Gessa, G.L.5
  • 36
    • 0024533951 scopus 로고
    • Blood-brain barrier passage of azidothymidine in rats: effect of insulin
    • Ayre SG, Skaletski B, Mosnaim AD. Blood-brain barrier passage of azidothymidine in rats: effect of insulin. Res Commun Chem Pathol Pharmacol (1989) 63(1):45-52.
    • (1989) Res Commun Chem Pathol Pharmacol , vol.63 , Issue.1 , pp. 45-52
    • Ayre, S.G.1    Skaletski, B.2    Mosnaim, A.D.3
  • 37
    • 0035016703 scopus 로고    scopus 로고
    • Glucose and insulin increase the transport of leptin through the blood-brain barrier in normal mice but not in streptozotocin-diabetic mice
    • Kastin AJ, Akerstrom V. Glucose and insulin increase the transport of leptin through the blood-brain barrier in normal mice but not in streptozotocin-diabetic mice. Neuroendocrinology (2001) 73(4):237-42. doi:10.1159/000054640
    • (2001) Neuroendocrinology , vol.73 , Issue.4 , pp. 237-242
    • Kastin, A.J.1    Akerstrom, V.2
  • 38
    • 58149175658 scopus 로고    scopus 로고
    • Insulin regulates P-glycoprotein in rat brain microvessel endothelial cells via an insulin receptor-mediated PKC/NF-kappaB pathway but not a PI3K/Akt pathway
    • Liu H, Yang H, Wang D, Liu Y, Liu X, Li Y, et al. Insulin regulates P-glycoprotein in rat brain microvessel endothelial cells via an insulin receptor-mediated PKC/NF-kappaB pathway but not a PI3K/Akt pathway. Eur J Pharmacol (2009) 602(2-3):277-82. doi:10.1016/j.ejphar.2008.11.026
    • (2009) Eur J Pharmacol , vol.602 , Issue.2-3 , pp. 277-282
    • Liu, H.1    Yang, H.2    Wang, D.3    Liu, Y.4    Liu, X.5    Li, Y.6
  • 39
    • 79955930291 scopus 로고    scopus 로고
    • Insulin suppresses the expression and function of breast cancer resistance protein in primary cultures of rat brain microvessel endothelial cells
    • Liu X, Jing XY, Jin S, Li Y, Liu L, Yu YL, et al. Insulin suppresses the expression and function of breast cancer resistance protein in primary cultures of rat brain microvessel endothelial cells. Pharmacol Rep (2011) 63(2):487-93. doi:10.1016/S1734-1140(11)70515-1
    • (2011) Pharmacol Rep , vol.63 , Issue.2 , pp. 487-493
    • Liu, X.1    Jing, X.Y.2    Jin, S.3    Li, Y.4    Liu, L.5    Yu, Y.L.6
  • 41
    • 80053583532 scopus 로고    scopus 로고
    • Activation of promoter activity of the catalytic subunit of gamma-glutamylcysteine ligase (GCL) in brain endothelial cells by insulin requires antioxidant response element 4 and altered glycemic status: implication for GCL expression and GSH synthesis
    • Langston JW, Li W, Harrison L, Aw TY. Activation of promoter activity of the catalytic subunit of gamma-glutamylcysteine ligase (GCL) in brain endothelial cells by insulin requires antioxidant response element 4 and altered glycemic status: implication for GCL expression and GSH synthesis. Free Radic Biol Med (2011) 51(9):1749-57. doi:10.1016/j.freeradbiomed.2011.08.004
    • (2011) Free Radic Biol Med , vol.51 , Issue.9 , pp. 1749-1757
    • Langston, J.W.1    Li, W.2    Harrison, L.3    Aw, T.Y.4
  • 43
    • 39149099122 scopus 로고    scopus 로고
    • Regional and cellular distribution patterns of insulin-degrading enzyme in the adult human brain and pituitary
    • Bernstein HG, Lendeckel U, Bukowska A, Ansorge S, Ernst T, Stauch R, et al. Regional and cellular distribution patterns of insulin-degrading enzyme in the adult human brain and pituitary. J Chem Neuroanat (2008) 35(2):216-24. doi:10.1016/j.jchemneu.2007.12.001
    • (2008) J Chem Neuroanat , vol.35 , Issue.2 , pp. 216-224
    • Bernstein, H.G.1    Lendeckel, U.2    Bukowska, A.3    Ansorge, S.4    Ernst, T.5    Stauch, R.6
  • 44
    • 33646363798 scopus 로고    scopus 로고
    • Insulin degrading enzyme is localized predominantly at the cell surface of polarized and unpolarized human cerebrovascular endothelial cell cultures
    • Lynch JA, George AM, Eisenhauer PB, Conn K, Gao W, Carreras I, et al. Insulin degrading enzyme is localized predominantly at the cell surface of polarized and unpolarized human cerebrovascular endothelial cell cultures. J Neurosci Res (2006) 83(7):1262-70. doi:10.1002/jnr.20809
    • (2006) J Neurosci Res , vol.83 , Issue.7 , pp. 1262-1270
    • Lynch, J.A.1    George, A.M.2    Eisenhauer, P.B.3    Conn, K.4    Gao, W.5    Carreras, I.6
  • 45
    • 0024548162 scopus 로고
    • Structure of the human insulin receptor gene and characterization of its promoter
    • Seino S, Seino M, Nishi S, Bell GI. Structure of the human insulin receptor gene and characterization of its promoter. Proc Natl Acad Sci U S A (1989) 86(1):114-8. doi:10.1073/pnas.86.1.114
    • (1989) Proc Natl Acad Sci U S A , vol.86 , Issue.1 , pp. 114-118
    • Seino, S.1    Seino, M.2    Nishi, S.3    Bell, G.I.4
  • 46
    • 0024403208 scopus 로고
    • Tissue-specific expression of two alternatively spliced insulin receptor mRNAs in man
    • Moller DE, Yokota A, Caro JF, Flier JS. Tissue-specific expression of two alternatively spliced insulin receptor mRNAs in man. Mol Endocrinol (1989) 3(8):1263-9. doi:10.1210/mend-3-8-1263
    • (1989) Mol Endocrinol , vol.3 , Issue.8 , pp. 1263-1269
    • Moller, D.E.1    Yokota, A.2    Caro, J.F.3    Flier, J.S.4
  • 47
    • 0024539198 scopus 로고
    • Alternative splicing of human insulin receptor messenger RNA
    • Seino S, Bell GI. Alternative splicing of human insulin receptor messenger RNA. Biochem Biophys Res Commun (1989) 159(1):312-6. doi:10.1016/0006-291X(89)92439-X
    • (1989) Biochem Biophys Res Commun , vol.159 , Issue.1 , pp. 312-316
    • Seino, S.1    Bell, G.I.2
  • 48
    • 82455185011 scopus 로고    scopus 로고
    • Receptor-isoform-selective insulin analogues give tissue-preferential effects
    • Vienberg SG, Bouman SD, Sorensen H, Stidsen CE, Kjeldsen T, Glendorf T, et al. Receptor-isoform-selective insulin analogues give tissue-preferential effects. Biochem J (2011) 440(3):301-8. doi:10.1042/BJ20110880
    • (2011) Biochem J , vol.440 , Issue.3 , pp. 301-308
    • Vienberg, S.G.1    Bouman, S.D.2    Sorensen, H.3    Stidsen, C.E.4    Kjeldsen, T.5    Glendorf, T.6
  • 49
    • 0033664539 scopus 로고    scopus 로고
    • Insulin receptors and insulin action in the brain: review and clinical implications
    • Schulingkamp RJ, Pagano TC, Hung D, Raffa RB. Insulin receptors and insulin action in the brain: review and clinical implications. Neurosci Biobehav Rev (2000) 24(8):855-72. doi:10.1016/S0149-7634(00)00040-3
    • (2000) Neurosci Biobehav Rev , vol.24 , Issue.8 , pp. 855-872
    • Schulingkamp, R.J.1    Pagano, T.C.2    Hung, D.3    Raffa, R.B.4
  • 50
    • 0015451256 scopus 로고
    • Evidence for an insulin-sensitive receptor in the central nervous system
    • Szabo O, Szabo AJ. Evidence for an insulin-sensitive receptor in the central nervous system. Am J Physiol (1972) 223(6):1349-53.
    • (1972) Am J Physiol , vol.223 , Issue.6 , pp. 1349-1353
    • Szabo, O.1    Szabo, A.J.2
  • 51
    • 0016212059 scopus 로고
    • Studies of insulin, growth hormone and prolactin binding: tissue distribution, species variation and characterization
    • Posner BI, Kelly PA, Shiu RP, Friesen HG. Studies of insulin, growth hormone and prolactin binding: tissue distribution, species variation and characterization. Endocrinology (1974) 95(2):521-31. doi:10.1210/endo-95-2-521
    • (1974) Endocrinology , vol.95 , Issue.2 , pp. 521-531
    • Posner, B.I.1    Kelly, P.A.2    Shiu, R.P.3    Friesen, H.G.4
  • 52
    • 0018090354 scopus 로고
    • Insulin receptors are widely distributed in the central nervous system of the rat
    • Havrankova J, Roth J, Brownstein M. Insulin receptors are widely distributed in the central nervous system of the rat. Nature (1978) 272(5656):827-9. doi:10.1038/272827a0
    • (1978) Nature , vol.272 , Issue.5656 , pp. 827-829
    • Havrankova, J.1    Roth, J.2    Brownstein, M.3
  • 53
    • 0022588853 scopus 로고
    • Development of brain insulin receptors: structural and functional studies of insulin receptors from whole brain and primary cell cultures
    • Lowe WL Jr, Boyd FT, Clarke DW, Raizada MK, Hart C, LeRoith D. Development of brain insulin receptors: structural and functional studies of insulin receptors from whole brain and primary cell cultures. Endocrinology (1986) 119(1):25-35. doi:10.1210/endo-119-1-25
    • (1986) Endocrinology , vol.119 , Issue.1 , pp. 25-35
    • Lowe W.L, Jr.1    Boyd, F.T.2    Clarke, D.W.3    Raizada, M.K.4    Hart, C.5    LeRoith, D.6
  • 55
    • 0022510796 scopus 로고
    • Autoradiographic localization of insulin receptors in rat brain: prominence in olfactory and limbic areas
    • Hill JM, Lesniak MA, Pert CB, Roth J. Autoradiographic localization of insulin receptors in rat brain: prominence in olfactory and limbic areas. Neuroscience (1986) 17(4):1127-38. doi:10.1016/0306-4522(86)90082-5
    • (1986) Neuroscience , vol.17 , Issue.4 , pp. 1127-1138
    • Hill, J.M.1    Lesniak, M.A.2    Pert, C.B.3    Roth, J.4
  • 56
    • 0023271871 scopus 로고
    • Localization and characterization of insulin receptors in rat brain and pituitary gland using in vitro autoradiography and computerized densitometry
    • Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Allen AM, et al. Localization and characterization of insulin receptors in rat brain and pituitary gland using in vitro autoradiography and computerized densitometry. Endocrinology (1987) 121(4):1562-70. doi:10.1210/endo-121-4-1562
    • (1987) Endocrinology , vol.121 , Issue.4 , pp. 1562-1570
    • Werther, G.A.1    Hogg, A.2    Oldfield, B.J.3    McKinley, M.J.4    Figdor, R.5    Allen, A.M.6
  • 57
    • 0025612094 scopus 로고
    • Localization of insulin receptor mRNA in rat brain by in situ hybridization
    • Marks JL, Porte D Jr, Stahl WL, Baskin DG. Localization of insulin receptor mRNA in rat brain by in situ hybridization. Endocrinology (1990) 127(6):3234-6. doi:10.1210/endo-127-6-3234
    • (1990) Endocrinology , vol.127 , Issue.6 , pp. 3234-3236
    • Marks, J.L.1    Porte Jr, D.2    Stahl, W.L.3    Baskin, D.G.4
  • 58
    • 77953292778 scopus 로고    scopus 로고
    • Age-related changes in insulin receptor mRNA and protein expression in genetically obese Zucker rats
    • Amessou M, Tahiri K, Chauvet G, Desbuquois B. Age-related changes in insulin receptor mRNA and protein expression in genetically obese Zucker rats. Diabetes Metab (2010) 36(2):120-8. doi:10.1016/j.diabet.2009.09.004
    • (2010) Diabetes Metab , vol.36 , Issue.2 , pp. 120-128
    • Amessou, M.1    Tahiri, K.2    Chauvet, G.3    Desbuquois, B.4
  • 59
    • 0018638092 scopus 로고
    • Central nervous system insulin receptors in normal and diabetic rats
    • Pacold ST, Blackard WG. Central nervous system insulin receptors in normal and diabetic rats. Endocrinology (1979) 105(6):1452-7.
    • (1979) Endocrinology , vol.105 , Issue.6 , pp. 1452-1457
    • Pacold, S.T.1    Blackard, W.G.2
  • 60
    • 0024326827 scopus 로고
    • Localization and characterization of insulin-like growth factor-I receptors in rat brain and pituitary gland using in vitro autoradiography and computerized densitometry* A distinct distribution from insulin receptors
    • Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Mendelsohn FA. Localization and characterization of insulin-like growth factor-I receptors in rat brain and pituitary gland using in vitro autoradiography and computerized densitometry* A distinct distribution from insulin receptors. J Neuroendocrinol (1989) 1(5):369-77. doi:10.1111/j.1365-2826.1989.tb00131.x
    • (1989) J Neuroendocrinol , vol.1 , Issue.5 , pp. 369-377
    • Werther, G.A.1    Hogg, A.2    Oldfield, B.J.3    McKinley, M.J.4    Figdor, R.5    Mendelsohn, F.A.6
  • 61
    • 84862902924 scopus 로고    scopus 로고
    • Sex differences and left-right asymmetries in expression of insulin and insulin-like growth factor-1 receptors in developing rat hippocampus
    • Hami J, Sadr-Nabavi A, Sankian M, Haghir H. Sex differences and left-right asymmetries in expression of insulin and insulin-like growth factor-1 receptors in developing rat hippocampus. Brain Struct Funct (2012) 217(2):293-302. doi:10.1007/s00429-011-0358-1
    • (2012) Brain Struct Funct , vol.217 , Issue.2 , pp. 293-302
    • Hami, J.1    Sadr-Nabavi, A.2    Sankian, M.3    Haghir, H.4
  • 62
    • 0031454481 scopus 로고    scopus 로고
    • Insulin receptors are widely distributed in human brain and bind human and porcine insulin with equal affinity
    • Hopkins DF, Williams G. Insulin receptors are widely distributed in human brain and bind human and porcine insulin with equal affinity. Diabet Med (1997) 14(12):1044-50. doi:10.1002/(SICI)1096-9136(199712)14:12<1044::AID-DIA508>3.3.CO;2-6
    • (1997) Diabet Med , vol.14 , Issue.12 , pp. 1044-1050
    • Hopkins, D.F.1    Williams, G.2
  • 63
    • 0026099276 scopus 로고
    • Ontogenesis of insulin receptors in human cerebral cortex
    • Potau N, Escofet MA, Martinez MC. Ontogenesis of insulin receptors in human cerebral cortex. J Endocrinol Invest (1991) 14(1):53-8. doi:10.1007/BF03350263
    • (1991) J Endocrinol Invest , vol.14 , Issue.1 , pp. 53-58
    • Potau, N.1    Escofet, M.A.2    Martinez, M.C.3
  • 64
    • 0021326129 scopus 로고
    • Characterization and regulation of insulin receptors in rat brain
    • Zahniser NR, Goens MB, Hanaway PJ, Vinych JV. Characterization and regulation of insulin receptors in rat brain. J Neurochem (1984) 42(5):1354-62. doi:10.1111/j.1471-4159.1984.tb02795.x
    • (1984) J Neurochem , vol.42 , Issue.5 , pp. 1354-1362
    • Zahniser, N.R.1    Goens, M.B.2    Hanaway, P.J.3    Vinych, J.V.4
  • 65
    • 0020962084 scopus 로고
    • Structural differences between insulin receptors in the brain and peripheral target tissues
    • Heidenreich KA, Zahniser NR, Berhanu P, Brandenburg D, Olefsky JM. Structural differences between insulin receptors in the brain and peripheral target tissues. J Biol Chem (1983) 258(14):8527-30.
    • (1983) J Biol Chem , vol.258 , Issue.14 , pp. 8527-8530
    • Heidenreich, K.A.1    Zahniser, N.R.2    Berhanu, P.3    Brandenburg, D.4    Olefsky, J.M.5
  • 66
    • 0028957007 scopus 로고
    • Structural and functional heterogeneity of insulin receptors
    • Joost HG. Structural and functional heterogeneity of insulin receptors. Cell Signal (1995) 7(2):85-91. doi:10.1016/0898-6568(94)00071-I
    • (1995) Cell Signal , vol.7 , Issue.2 , pp. 85-91
    • Joost, H.G.1
  • 67
    • 0027314389 scopus 로고
    • Quantitative autoradiographic localization of [125I]insulin-like growth factor I, [125I]insulin-like growth factor II, and [125I]insulin receptor binding sites in developing and adult rat brain
    • Kar S, Chabot JG, Quirion R. Quantitative autoradiographic localization of [125I]insulin-like growth factor I, [125I]insulin-like growth factor II, and [125I]insulin receptor binding sites in developing and adult rat brain. J Comp Neurol (1993) 333(3):375-97. doi:10.1002/cne.903330306
    • (1993) J Comp Neurol , vol.333 , Issue.3 , pp. 375-397
    • Kar, S.1    Chabot, J.G.2    Quirion, R.3
  • 68
    • 0024848790 scopus 로고
    • Insulin and IGF-I stimulate phosphorylation of their respective receptors in intact neuronal and glial cells in primary culture
    • Shemer J, Adamo M, Raizada MK, Heffez D, Zick Y, LeRoith D. Insulin and IGF-I stimulate phosphorylation of their respective receptors in intact neuronal and glial cells in primary culture. J Mol Neurosci (1989) 1(1):3-8. doi:10.1007/BF02918899
    • (1989) J Mol Neurosci , vol.1 , Issue.1 , pp. 3-8
    • Shemer, J.1    Adamo, M.2    Raizada, M.K.3    Heffez, D.4    Zick, Y.5    LeRoith, D.6
  • 69
    • 79952111041 scopus 로고    scopus 로고
    • Docking proteins
    • Brummer T, Schmitz-Peiffer C, Daly RJ. Docking proteins. FEBS J (2010) 277(21):4356-69. doi:10.1111/j.1742-4658.2010.07865.x
    • (2010) FEBS J , vol.277 , Issue.21 , pp. 4356-4369
    • Brummer, T.1    Schmitz-Peiffer, C.2    Daly, R.J.3
  • 70
    • 43549122104 scopus 로고    scopus 로고
    • Insulin-like signaling, nutrient homeostasis, and life span
    • Taguchi A, White MF. Insulin-like signaling, nutrient homeostasis, and life span. Annu Rev Physiol (2008) 70:191-212. doi:10.1146/annurev.physiol.70.113006.100533
    • (2008) Annu Rev Physiol , vol.70 , pp. 191-212
    • Taguchi, A.1    White, M.F.2
  • 71
    • 0036470213 scopus 로고    scopus 로고
    • Insulin signaling pathways in time and space
    • Saltiel AR, Pessin JE. Insulin signaling pathways in time and space. Trends Cell Biol (2002) 12(2):65-71. doi:10.1016/S0962-8924(01)02207-3
    • (2002) Trends Cell Biol , vol.12 , Issue.2 , pp. 65-71
    • Saltiel, A.R.1    Pessin, J.E.2
  • 72
    • 0020695047 scopus 로고
    • Coated vesicles participate in the receptor-mediated endocytosis of insulin
    • Pilch PF, Shia MA, Benson RJ, Fine RE. Coated vesicles participate in the receptor-mediated endocytosis of insulin. J Cell Biol (1983) 96(1):133-8. doi:10.1083/jcb.96.1.133
    • (1983) J Cell Biol , vol.96 , Issue.1 , pp. 133-138
    • Pilch, P.F.1    Shia, M.A.2    Benson, R.J.3    Fine, R.E.4
  • 73
    • 0022580601 scopus 로고
    • Receptor aggregation is necessary for activation of the soluble insulin receptor kinase
    • Heffetz D, Zick Y. Receptor aggregation is necessary for activation of the soluble insulin receptor kinase. J Biol Chem (1986) 261(2):889-94.
    • (1986) J Biol Chem , vol.261 , Issue.2 , pp. 889-894
    • Heffetz, D.1    Zick, Y.2
  • 74
    • 0344305782 scopus 로고    scopus 로고
    • Insulin signaling in health and disease
    • White MF. Insulin signaling in health and disease. Science (2003) 302(5651):1710-1. doi:10.1126/science.1092952
    • (2003) Science , vol.302 , Issue.5651 , pp. 1710-1711
    • White, M.F.1
  • 75
    • 0028287467 scopus 로고
    • The IRS-1 signaling system
    • Myers MG Jr, Sun XJ, White MF. The IRS-1 signaling system. Trends Biochem Sci (1994) 19(7):289-93. doi:10.1016/0968-0004(94)90007-8
    • (1994) Trends Biochem Sci , vol.19 , Issue.7 , pp. 289-293
    • Myers M.G, Jr.1    Sun, X.J.2    White, M.F.3
  • 76
    • 0037059330 scopus 로고    scopus 로고
    • Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action
    • Aguirre V, Werner ED, Giraud J, Lee YH, Shoelson SE, White MF. Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action. J Biol Chem (2002) 277(2):1531-7. doi:10.1074/jbc.M101521200
    • (2002) J Biol Chem , vol.277 , Issue.2 , pp. 1531-1537
    • Aguirre, V.1    Werner, E.D.2    Giraud, J.3    Lee, Y.H.4    Shoelson, S.E.5    White, M.F.6
  • 77
    • 17044451410 scopus 로고    scopus 로고
    • A new role for SOCS in insulin action. Suppressor of cytokine signaling
    • Krebs DL, Hilton DJ. A new role for SOCS in insulin action. Suppressor of cytokine signaling. Sci STKE (2003) 2003(169):e6. doi:10.1126/stke.2003.169.pe6
    • (2003) Sci STKE , vol.2003 , Issue.169 , pp. e6
    • Krebs, D.L.1    Hilton, D.J.2
  • 78
    • 0038339191 scopus 로고    scopus 로고
    • cAMP promotes pancreatic beta-cell survival via CREB-mediated induction of IRS2
    • Jhala US, Canettieri G, Screaton RA, Kulkarni RN, Krajewski S, Reed J, et al. cAMP promotes pancreatic beta-cell survival via CREB-mediated induction of IRS2. Genes Dev (2003) 17(13):1575-80. doi:10.1101/gad.1097103
    • (2003) Genes Dev , vol.17 , Issue.13 , pp. 1575-1580
    • Jhala, U.S.1    Canettieri, G.2    Screaton, R.A.3    Kulkarni, R.N.4    Krajewski, S.5    Reed, J.6
  • 79
    • 0033197996 scopus 로고    scopus 로고
    • The insulin receptor tyrosine kinase substrate p58/53 and the insulin receptor are components of CNS synapses
    • Abbott MA, Wells DG, Fallon JR. The insulin receptor tyrosine kinase substrate p58/53 and the insulin receptor are components of CNS synapses. J Neurosci (1999) 19(17):7300-8.
    • (1999) J Neurosci , vol.19 , Issue.17 , pp. 7300-7308
    • Abbott, M.A.1    Wells, D.G.2    Fallon, J.R.3
  • 80
    • 0035930903 scopus 로고    scopus 로고
    • Distinctive tissue distribution and phosphorylation of IRSp53 isoforms
    • Okamura-Oho Y, Miyashita T, Yamada M. Distinctive tissue distribution and phosphorylation of IRSp53 isoforms. Biochem Biophys Res Commun (2001) 289(5):957-60. doi:10.1006/bbrc.2001.6102
    • (2001) Biochem Biophys Res Commun , vol.289 , Issue.5 , pp. 957-960
    • Okamura-Oho, Y.1    Miyashita, T.2    Yamada, M.3
  • 81
    • 0141533149 scopus 로고    scopus 로고
    • Genomic structure and alternative splicing of the insulin receptor tyrosine kinase substrate of 53-kDa protein
    • Miyahara A, Okamura-Oho Y, Miyashita T, Hoshika A, Yamada M. Genomic structure and alternative splicing of the insulin receptor tyrosine kinase substrate of 53-kDa protein. J Hum Genet (2003) 48(8):410-4. doi:10.1007/s10038-003-0047-x
    • (2003) J Hum Genet , vol.48 , Issue.8 , pp. 410-414
    • Miyahara, A.1    Okamura-Oho, Y.2    Miyashita, T.3    Hoshika, A.4    Yamada, M.5
  • 82
    • 19944433966 scopus 로고    scopus 로고
    • Regulation of dendritic spine morphogenesis by insulin receptor substrate 53, a downstream effector of Rac1 and Cdc42 small GTPases
    • Choi J, Ko J, Racz B, Burette A, Lee JR, Kim S, et al. Regulation of dendritic spine morphogenesis by insulin receptor substrate 53, a downstream effector of Rac1 and Cdc42 small GTPases. J Neurosci (2005) 25(4):869-79. doi:10.1523/JNEUROSCI.3212-04.2005
    • (2005) J Neurosci , vol.25 , Issue.4 , pp. 869-879
    • Choi, J.1    Ko, J.2    Racz, B.3    Burette, A.4    Lee, J.R.5    Kim, S.6
  • 83
    • 23844532570 scopus 로고    scopus 로고
    • Novel brain 14-3-3 interacting proteins involved in neurodegenerative disease
    • Mackie S, Aitken A. Novel brain 14-3-3 interacting proteins involved in neurodegenerative disease. FEBS J (2005) 272(16):4202-10. doi:10.1111/j.1742-4658.2005.04832.x
    • (2005) FEBS J , vol.272 , Issue.16 , pp. 4202-4210
    • Mackie, S.1    Aitken, A.2
  • 84
    • 66149124468 scopus 로고    scopus 로고
    • The insulin receptor substrate of 53 kDa (IRSp53) limits hippocampal synaptic plasticity
    • Sawallisch C, Berhorster K, Disanza A, Mantoani S, Kintscher M, Stoenica L, et al. The insulin receptor substrate of 53 kDa (IRSp53) limits hippocampal synaptic plasticity. J Biol Chem (2009) 284(14):9225-36. doi:10.1074/jbc.M808425200
    • (2009) J Biol Chem , vol.284 , Issue.14 , pp. 9225-9236
    • Sawallisch, C.1    Berhorster, K.2    Disanza, A.3    Mantoani, S.4    Kintscher, M.5    Stoenica, L.6
  • 85
    • 0037007124 scopus 로고    scopus 로고
    • The insulin signalling pathway
    • Lizcano JM, Alessi DR. The insulin signalling pathway. Curr Biol (2002) 12(7):R236-8. doi:10.1016/S0960-9822(02)00777-7
    • (2002) Curr Biol , vol.12 , Issue.7 , pp. R236-R238
    • Lizcano, J.M.1    Alessi, D.R.2
  • 86
    • 84884676252 scopus 로고    scopus 로고
    • Nutrient signaling to mTOR and cell growth
    • Jewell JL, Guan KL. Nutrient signaling to mTOR and cell growth. Trends Biochem Sci (2013) 38(5):233-42. doi:10.1016/j.tibs.2013.01.004
    • (2013) Trends Biochem Sci , vol.38 , Issue.5 , pp. 233-242
    • Jewell, J.L.1    Guan, K.L.2
  • 87
    • 0005887162 scopus 로고    scopus 로고
    • The role of Raf kinases in malignant transformation
    • Kolch W, Kotwaliwale A, Vass K, Janosch P. The role of Raf kinases in malignant transformation. Expert Rev Mol Med (2002) 4(8):1-18. doi:10.1017/S1462399402004386
    • (2002) Expert Rev Mol Med , vol.4 , Issue.8 , pp. 1-18
    • Kolch, W.1    Kotwaliwale, A.2    Vass, K.3    Janosch, P.4
  • 88
    • 2442684094 scopus 로고    scopus 로고
    • Cholesterol and APOE genotype interact to influence Alzheimer disease progression
    • Evans RM, Hui S, Perkins A, Lahiri DK, Poirier J, Farlow MR. Cholesterol and APOE genotype interact to influence Alzheimer disease progression. Neurology (2004) 62(10):1869-71. doi:10.1212/01.WNL.0000125323.15458.3F
    • (2004) Neurology , vol.62 , Issue.10 , pp. 1869-1871
    • Evans, R.M.1    Hui, S.2    Perkins, A.3    Lahiri, D.K.4    Poirier, J.5    Farlow, M.R.6
  • 89
    • 0040175067 scopus 로고    scopus 로고
    • Role and regulation of 90 kDa ribosomal S6 kinase (RSK) in signal transduction
    • Frodin M, Gammeltoft S. Role and regulation of 90 kDa ribosomal S6 kinase (RSK) in signal transduction. Mol Cell Endocrinol (1999) 151(1-2):65-77. doi:10.1016/S0303-7207(99)00061-1
    • (1999) Mol Cell Endocrinol , vol.151 , Issue.1-2 , pp. 65-77
    • Frodin, M.1    Gammeltoft, S.2
  • 90
    • 84862739428 scopus 로고    scopus 로고
    • Clinical review: ketones and brain injury
    • White H, Venkatesh B. Clinical review: ketones and brain injury. Crit Care (2011) 15(2):219. doi:10.1186/cc10020
    • (2011) Crit Care , vol.15 , Issue.2 , pp. 219
    • White, H.1    Venkatesh, B.2
  • 91
    • 34548207654 scopus 로고    scopus 로고
    • Brain glucose sensing, counterregulation, and energy homeostasis
    • Marty N, Dallaporta M, Thorens B. Brain glucose sensing, counterregulation, and energy homeostasis. Physiology (Bethesda) (2007) 22:241-51. doi:10.1152/physiol.00010.2007
    • (2007) Physiology (Bethesda) , vol.22 , pp. 241-251
    • Marty, N.1    Dallaporta, M.2    Thorens, B.3
  • 92
    • 0030056878 scopus 로고    scopus 로고
    • Expression of the glucagon-like peptide-1 receptor gene in rat brain
    • Alvarez E, Roncero I, Chowen JA, Thorens B, Blazquez E. Expression of the glucagon-like peptide-1 receptor gene in rat brain. J Neurochem (1996) 66(3):920-7. doi:10.1046/j.1471-4159.1996.66030920.x
    • (1996) J Neurochem , vol.66 , Issue.3 , pp. 920-927
    • Alvarez, E.1    Roncero, I.2    Chowen, J.A.3    Thorens, B.4    Blazquez, E.5
  • 93
    • 10244241840 scopus 로고    scopus 로고
    • Colocalization of glucagon-like peptide-1 (GLP-1) receptors, glucose transporter GLUT-2, and glucokinase mRNAs in rat hypothalamic cells: evidence for a role of GLP-1 receptor agonists as an inhibitory signal for food and water intake
    • Navarro M, Rodriquez de Fonseca F, Alvarez E, Chowen JA, Zueco JA, Gomez R, et al. Colocalization of glucagon-like peptide-1 (GLP-1) receptors, glucose transporter GLUT-2, and glucokinase mRNAs in rat hypothalamic cells: evidence for a role of GLP-1 receptor agonists as an inhibitory signal for food and water intake. J Neurochem (1996) 67(5):1982-91. doi:10.1046/j.1471-4159.1996.67051982.x
    • (1996) J Neurochem , vol.67 , Issue.5 , pp. 1982-1991
    • Navarro, M.1    Rodriquez de Fonseca, F.2    Alvarez, E.3    Chowen, J.A.4    Zueco, J.A.5    Gomez, R.6
  • 94
    • 0034090708 scopus 로고    scopus 로고
    • Functional glucokinase isoforms are expressed in rat brain
    • Roncero I, Alvarez E, Vazquez P, Blazquez E. Functional glucokinase isoforms are expressed in rat brain. J Neurochem (2000) 74(5):1848-57. doi:10.1046/j.1471-4159.2000.0741848.x
    • (2000) J Neurochem , vol.74 , Issue.5 , pp. 1848-1857
    • Roncero, I.1    Alvarez, E.2    Vazquez, P.3    Blazquez, E.4
  • 95
    • 74949118966 scopus 로고    scopus 로고
    • Glucose transporters in the 21st century
    • Thorens B, Mueckler M. Glucose transporters in the 21st century. Am J Physiol Endocrinol Metab (2010) 298(2):E141-5. doi:10.1152/ajpendo.00712.2009
    • (2010) Am J Physiol Endocrinol Metab , vol.298 , Issue.2 , pp. E141-E145
    • Thorens, B.1    Mueckler, M.2
  • 96
    • 0032898973 scopus 로고    scopus 로고
    • Blood-brain barrier glucose transporter: effects of hypo- and hyper-glycemia revisited
    • Simpson IA, Appel NM, Hokari M, Oki J, Holman GD, Maher F, et al. Blood-brain barrier glucose transporter: effects of hypo- and hyper-glycemia revisited. J Neurochem (1999) 72(1):238-47. doi:10.1046/j.1471-4159.1999.0720238.x
    • (1999) J Neurochem , vol.72 , Issue.1 , pp. 238-247
    • Simpson, I.A.1    Appel, N.M.2    Hokari, M.3    Oki, J.4    Holman, G.D.5    Maher, F.6
  • 97
    • 1442276966 scopus 로고    scopus 로고
    • Physiological and molecular characteristics of rat hypothalamic ventromedial nucleus glucosensing neurons
    • Kang L, Routh VH, Kuzhikandathil EV, Gaspers LD, Levin BE. Physiological and molecular characteristics of rat hypothalamic ventromedial nucleus glucosensing neurons. Diabetes (2004) 53(3):549-59. doi:10.2337/diabetes.53.3.549
    • (2004) Diabetes , vol.53 , Issue.3 , pp. 549-559
    • Kang, L.1    Routh, V.H.2    Kuzhikandathil, E.V.3    Gaspers, L.D.4    Levin, B.E.5
  • 98
    • 4644253021 scopus 로고    scopus 로고
    • Neuronal glucosensing: what do we know after 50 years?
    • Levin BE, Routh VH, Kang L, Sanders NM, Dunn-Meynell AA. Neuronal glucosensing: what do we know after 50 years? Diabetes (2004) 53(10):2521-8. doi:10.2337/diabetes.53.10.2521
    • (2004) Diabetes , vol.53 , Issue.10 , pp. 2521-2528
    • Levin, B.E.1    Routh, V.H.2    Kang, L.3    Sanders, N.M.4    Dunn-Meynell, A.A.5
  • 99
    • 0038582455 scopus 로고    scopus 로고
    • Distribution of glucokinase, glucose transporter GLUT2, sulfonylurea receptor-1, glucagon-like peptide-1 receptor and neuropeptide Y messenger RNAs in rat brain by quantitative real time RT-PCR
    • Li B, Xi X, Roane DS, Ryan DH, Martin RJ. Distribution of glucokinase, glucose transporter GLUT2, sulfonylurea receptor-1, glucagon-like peptide-1 receptor and neuropeptide Y messenger RNAs in rat brain by quantitative real time RT-PCR. Brain Res Mol Brain Res (2003) 113(1-2):139-42. doi:10.1016/S0169-328X(03)00125-6
    • (2003) Brain Res Mol Brain Res , vol.113 , Issue.1-2 , pp. 139-142
    • Li, B.1    Xi, X.2    Roane, D.S.3    Ryan, D.H.4    Martin, R.J.5
  • 100
    • 0026501334 scopus 로고
    • Glucose transporter expression in brain. cDNA sequence of mouse GLUT3, the brain facilitative glucose transporter isoform, and identification of sites of expression by in situ hybridization
    • Nagamatsu S, Kornhauser JM, Burant CF, Seino S, Mayo KE, Bell GI. Glucose transporter expression in brain. cDNA sequence of mouse GLUT3, the brain facilitative glucose transporter isoform, and identification of sites of expression by in situ hybridization. J Biol Chem (1992) 267(1):467-72.
    • (1992) J Biol Chem , vol.267 , Issue.1 , pp. 467-472
    • Nagamatsu, S.1    Kornhauser, J.M.2    Burant, C.F.3    Seino, S.4    Mayo, K.E.5    Bell, G.I.6
  • 101
    • 0026549486 scopus 로고
    • Expression of the brain-type glucose transporter is restricted to brain and neuronal cells in mice
    • Gould GW, Brant AM, Kahn BB, Shepherd PR, McCoid SC, Gibbs EM. Expression of the brain-type glucose transporter is restricted to brain and neuronal cells in mice. Diabetologia (1992) 35(4):304-9. doi:10.1007/BF00401196
    • (1992) Diabetologia , vol.35 , Issue.4 , pp. 304-309
    • Gould, G.W.1    Brant, A.M.2    Kahn, B.B.3    Shepherd, P.R.4    McCoid, S.C.5    Gibbs, E.M.6
  • 102
    • 0032487335 scopus 로고    scopus 로고
    • Immunocytochemical localization of the insulin-responsive glucose transporter 4 (Glut4) in the rat central nervous system
    • El Messari S, Leloup C, Quignon M, Brisorgueil MJ, Penicaud L, Arluison M. Immunocytochemical localization of the insulin-responsive glucose transporter 4 (Glut4) in the rat central nervous system. J Comp Neurol (1998) 399(4):492-512. doi:10.1002/(SICI)1096-9861(19981005)399:4<492::AID-CNE4>3.0.CO;2-X
    • (1998) J Comp Neurol , vol.399 , Issue.4 , pp. 492-512
    • El Messari, S.1    Leloup, C.2    Quignon, M.3    Brisorgueil, M.J.4    Penicaud, L.5    Arluison, M.6
  • 103
    • 0032557956 scopus 로고    scopus 로고
    • GLUT4 glucose transporter expression in rodent brain: effect of diabetes
    • Vannucci SJ, Koehler-Stec EM, Li K, Reynolds TH, Clark R, Simpson IA. GLUT4 glucose transporter expression in rodent brain: effect of diabetes. Brain Res (1998) 797(1):1-11. doi:10.1016/S0006-8993(98)00103-6
    • (1998) Brain Res , vol.797 , Issue.1 , pp. 1-11
    • Vannucci, S.J.1    Koehler-Stec, E.M.2    Li, K.3    Reynolds, T.H.4    Clark, R.5    Simpson, I.A.6
  • 104
    • 84859718265 scopus 로고    scopus 로고
    • Demonstrated brain insulin resistance in Alzheimer's disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline
    • Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, et al. Demonstrated brain insulin resistance in Alzheimer's disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Invest (2012) 122(4):1316-38. doi:10.1172/JCI59903
    • (2012) J Clin Invest , vol.122 , Issue.4 , pp. 1316-1338
    • Talbot, K.1    Wang, H.Y.2    Kazi, H.3    Han, L.Y.4    Bakshi, K.P.5    Stucky, A.6
  • 105
    • 0028890116 scopus 로고
    • Hypothalamic GLUT 4 expression: a glucose- and insulin-sensing mechanism?
    • Livingstone C, Lyall H, Gould GW. Hypothalamic GLUT 4 expression: a glucose- and insulin-sensing mechanism? Mol Cell Endocrinol (1995) 107(1):67-70. doi:10.1016/0303-7207(94)03423-Q
    • (1995) Mol Cell Endocrinol , vol.107 , Issue.1 , pp. 67-70
    • Livingstone, C.1    Lyall, H.2    Gould, G.W.3
  • 106
    • 3242765290 scopus 로고    scopus 로고
    • The regulation of glucose-excited neurons in the hypothalamic arcuate nucleus by glucose and feeding-relevant peptides
    • Wang R, Liu X, Hentges ST, Dunn-Meynell AA, Levin BE, Wang W, et al. The regulation of glucose-excited neurons in the hypothalamic arcuate nucleus by glucose and feeding-relevant peptides. Diabetes (2004) 53(8):1959-65. doi:10.2337/diabetes.53.8.1959
    • (2004) Diabetes , vol.53 , Issue.8 , pp. 1959-1965
    • Wang, R.1    Liu, X.2    Hentges, S.T.3    Dunn-Meynell, A.A.4    Levin, B.E.5    Wang, W.6
  • 107
    • 0035956883 scopus 로고    scopus 로고
    • Localization and regulation of GLUTx1 glucose transporter in the hippocampus of streptozotocin diabetic rats
    • Reagan LP, Gorovits N, Hoskin EK, Alves SE, Katz EB, Grillo CA, et al. Localization and regulation of GLUTx1 glucose transporter in the hippocampus of streptozotocin diabetic rats. Proc Natl Acad Sci U S A (2001) 98(5):2820-5. doi:10.1073/pnas.051629798
    • (2001) Proc Natl Acad Sci U S A , vol.98 , Issue.5 , pp. 2820-2825
    • Reagan, L.P.1    Gorovits, N.2    Hoskin, E.K.3    Alves, S.E.4    Katz, E.B.5    Grillo, C.A.6
  • 108
    • 0036141095 scopus 로고    scopus 로고
    • Immunolocalization of GLUTX1 in the testis and to specific brain areas and vasopressin-containing neurons
    • Ibberson M, Riederer BM, Uldry M, Guhl B, Roth J, Thorens B. Immunolocalization of GLUTX1 in the testis and to specific brain areas and vasopressin-containing neurons. Endocrinology (2002) 143(1):276-84. doi:10.1210/endo.143.1.8587
    • (2002) Endocrinology , vol.143 , Issue.1 , pp. 276-284
    • Ibberson, M.1    Riederer, B.M.2    Uldry, M.3    Guhl, B.4    Roth, J.5    Thorens, B.6
  • 109
    • 0037023202 scopus 로고    scopus 로고
    • GLUT8 glucose transporter is localized to excitatory and inhibitory neurons in the rat hippocampus
    • Reagan LP, Rosell DR, Alves SE, Hoskin EK, McCall AL, Charron MJ, et al. GLUT8 glucose transporter is localized to excitatory and inhibitory neurons in the rat hippocampus. Brain Res (2002) 932(1-2):129-34. doi:10.1016/S0006-8993(02)02308-9
    • (2002) Brain Res , vol.932 , Issue.1-2 , pp. 129-134
    • Reagan, L.P.1    Rosell, D.R.2    Alves, S.E.3    Hoskin, E.K.4    McCall, A.L.5    Charron, M.J.6
  • 110
    • 0037112093 scopus 로고    scopus 로고
    • Insulin-responsive glucose transporters-GLUT8 and GLUT4 are expressed in the developing mammalian brain
    • Sankar R, Thamotharan S, Shin D, Moley KH, Devaskar SU. Insulin-responsive glucose transporters-GLUT8 and GLUT4 are expressed in the developing mammalian brain. Brain Res Mol Brain Res (2002) 107(2):157-65. doi:10.1016/S0169-328X(02)00487-4
    • (2002) Brain Res Mol Brain Res , vol.107 , Issue.2 , pp. 157-165
    • Sankar, R.1    Thamotharan, S.2    Shin, D.3    Moley, K.H.4    Devaskar, S.U.5
  • 111
    • 1842734654 scopus 로고    scopus 로고
    • Glucose transporter expression in the central nervous system: relationship to synaptic function
    • McEwen BS, Reagan LP. Glucose transporter expression in the central nervous system: relationship to synaptic function. Eur J Pharmacol (2004) 490(1-3):13-24. doi:10.1016/j.ejphar.2004.02.041
    • (2004) Eur J Pharmacol , vol.490 , Issue.1-3 , pp. 13-24
    • McEwen, B.S.1    Reagan, L.P.2
  • 112
    • 0037078231 scopus 로고    scopus 로고
    • Peripheral glucose administration stimulates the translocation of GLUT8 glucose transporter to the endoplasmic reticulum in the rat hippocampus
    • Piroli GG, Grillo CA, Hoskin EK, Znamensky V, Katz EB, Milner TA, et al. Peripheral glucose administration stimulates the translocation of GLUT8 glucose transporter to the endoplasmic reticulum in the rat hippocampus. J Comp Neurol (2002) 452(2):103-14. doi:10.1002/cne.10368
    • (2002) J Comp Neurol , vol.452 , Issue.2 , pp. 103-114
    • Piroli, G.G.1    Grillo, C.A.2    Hoskin, E.K.3    Znamensky, V.4    Katz, E.B.5    Milner, T.A.6
  • 113
    • 0036267472 scopus 로고    scopus 로고
    • Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats
    • Obici S, Feng Z, Karkanias G, Baskin DG, Rossetti L. Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats. Nat Neurosci (2002) 5(6):566-72. doi:10.1038/nn0602-861
    • (2002) Nat Neurosci , vol.5 , Issue.6 , pp. 566-572
    • Obici, S.1    Feng, Z.2    Karkanias, G.3    Baskin, D.G.4    Rossetti, L.5
  • 114
    • 0036913187 scopus 로고    scopus 로고
    • Hypothalamic insulin signaling is required for inhibition of glucose production
    • Obici S, Zhang BB, Karkanias G, Rossetti L. Hypothalamic insulin signaling is required for inhibition of glucose production. Nat Med (2002) 8(12):1376-82. doi:10.1038/nm1202-798
    • (2002) Nat Med , vol.8 , Issue.12 , pp. 1376-1382
    • Obici, S.1    Zhang, B.B.2    Karkanias, G.3    Rossetti, L.4
  • 115
    • 33745225617 scopus 로고    scopus 로고
    • Central nervous system and control of endogenous glucose production
    • Demuro G, Obici S. Central nervous system and control of endogenous glucose production. Curr Diab Rep (2006) 6(3):188-93. doi:10.1007/s11892-006-0033-8
    • (2006) Curr Diab Rep , vol.6 , Issue.3 , pp. 188-193
    • Demuro, G.1    Obici, S.2
  • 116
    • 33845515150 scopus 로고    scopus 로고
    • The inhibitory effects of insulin on hepatic glucose production are both direct and indirect
    • Girard J. The inhibitory effects of insulin on hepatic glucose production are both direct and indirect. Diabetes (2006) 55(Suppl 2):S65-9. doi:10.2337/db06-S009
    • (2006) Diabetes , vol.55 , pp. S65-S69
    • Girard, J.1
  • 117
    • 17844379717 scopus 로고    scopus 로고
    • Hypothalamic K(ATP) channels control hepatic glucose production
    • Pocai A, Lam TK, Gutierrez-Juarez R, Obici S, Schwartz GJ, Bryan J, et al. Hypothalamic K(ATP) channels control hepatic glucose production. Nature (2005) 434(7036):1026-31. doi:10.1038/nature03439
    • (2005) Nature , vol.434 , Issue.7036 , pp. 1026-1031
    • Pocai, A.1    Lam, T.K.2    Gutierrez-Juarez, R.3    Obici, S.4    Schwartz, G.J.5    Bryan, J.6
  • 118
    • 66149083518 scopus 로고    scopus 로고
    • Insulin blunts the response of glucose-excited neurons in the ventrolateral-ventromedial hypothalamic nucleus to decreased glucose
    • Cotero VE, Routh VH. Insulin blunts the response of glucose-excited neurons in the ventrolateral-ventromedial hypothalamic nucleus to decreased glucose. Am J Physiol Endocrinol Metab (2009) 296(5):E1101-9. doi:10.1152/ajpendo.90932.2008
    • (2009) Am J Physiol Endocrinol Metab , vol.296 , Issue.5 , pp. E1101-E1109
    • Cotero, V.E.1    Routh, V.H.2
  • 120
    • 0037315042 scopus 로고    scopus 로고
    • Insulin activation of phosphatidylinositol 3-kinase in the hypothalamic arcuate nucleus: a key mediator of insulin-induced anorexia
    • Niswender KD, Morrison CD, Clegg DJ, Olson R, Baskin DG, Myers MG Jr, et al. Insulin activation of phosphatidylinositol 3-kinase in the hypothalamic arcuate nucleus: a key mediator of insulin-induced anorexia. Diabetes (2003) 52(2):227-31. doi:10.2337/diabetes.52.2.227
    • (2003) Diabetes , vol.52 , Issue.2 , pp. 227-231
    • Niswender, K.D.1    Morrison, C.D.2    Clegg, D.J.3    Olson, R.4    Baskin, D.G.5    Myers M.G, Jr.6
  • 121
    • 0034457883 scopus 로고    scopus 로고
    • In vivo administration of leptin activates signal transduction directly in insulin-sensitive tissues: overlapping but distinct pathways from insulin
    • Kim YB, Uotani S, Pierroz DD, Flier JS, Kahn BB. In vivo administration of leptin activates signal transduction directly in insulin-sensitive tissues: overlapping but distinct pathways from insulin. Endocrinology (2000) 141(7):2328-39. doi:10.1210/endo.141.7.7536
    • (2000) Endocrinology , vol.141 , Issue.7 , pp. 2328-2339
    • Kim, Y.B.1    Uotani, S.2    Pierroz, D.D.3    Flier, J.S.4    Kahn, B.B.5
  • 122
    • 57849148921 scopus 로고    scopus 로고
    • KiSS-1/kisspeptins and the metabolic control of reproduction: physiologic roles and putative physiopathological implications
    • Castellano JM, Roa J, Luque RM, Dieguez C, Aguilar E, Pinilla L, et al. KiSS-1/kisspeptins and the metabolic control of reproduction: physiologic roles and putative physiopathological implications. Peptides (2009) 30(1):139-45. doi:10.1016/j.peptides.2008.06.007
    • (2009) Peptides , vol.30 , Issue.1 , pp. 139-145
    • Castellano, J.M.1    Roa, J.2    Luque, R.M.3    Dieguez, C.4    Aguilar, E.5    Pinilla, L.6
  • 125
    • 0028826266 scopus 로고
    • The role of intracerebral insulin in the effect of nutrition on gonadotrophin secretion in mature male sheep
    • Miller DW, Blache D, Martin GB. The role of intracerebral insulin in the effect of nutrition on gonadotrophin secretion in mature male sheep. J Endocrinol (1995) 147(2):321-9. doi:10.1677/joe.0.1470321
    • (1995) J Endocrinol , vol.147 , Issue.2 , pp. 321-329
    • Miller, D.W.1    Blache, D.2    Martin, G.B.3
  • 126
    • 0025936927 scopus 로고
    • Pulsatile LH secretion in streptozotocin-induced diabetes in the rat
    • Dong Q, Lazarus RM, Wong LS, Vellios M, Handelsman DJ. Pulsatile LH secretion in streptozotocin-induced diabetes in the rat. J Endocrinol (1991) 131(1):49-55. doi:10.1677/joe.0.1310049
    • (1991) J Endocrinol , vol.131 , Issue.1 , pp. 49-55
    • Dong, Q.1    Lazarus, R.M.2    Wong, L.S.3    Vellios, M.4    Handelsman, D.J.5
  • 127
    • 0034007333 scopus 로고    scopus 로고
    • Central action of insulin regulates pulsatile luteinizing hormone secretion in the diabetic sheep model
    • Tanaka T, Nagatani S, Bucholtz DC, Ohkura S, Tsukamura H, Maeda K, et al. Central action of insulin regulates pulsatile luteinizing hormone secretion in the diabetic sheep model. Biol Reprod (2000) 62(5):1256-61. doi:10.1095/biolreprod62.5.1256
    • (2000) Biol Reprod , vol.62 , Issue.5 , pp. 1256-1261
    • Tanaka, T.1    Nagatani, S.2    Bucholtz, D.C.3    Ohkura, S.4    Tsukamura, H.5    Maeda, K.6
  • 128
    • 0034007026 scopus 로고    scopus 로고
    • Regulation of pulsatile luteinizing hormone secretion by insulin in the diabetic male lamb
    • Bucholtz DC, Chiesa A, Pappano WN, Nagatani S, Tsukamura H, Maeda KI, et al. Regulation of pulsatile luteinizing hormone secretion by insulin in the diabetic male lamb. Biol Reprod (2000) 62(5):1248-55. doi:10.1095/biolreprod62.5.1248
    • (2000) Biol Reprod , vol.62 , Issue.5 , pp. 1248-1255
    • Bucholtz, D.C.1    Chiesa, A.2    Pappano, W.N.3    Nagatani, S.4    Tsukamura, H.5    Maeda, K.I.6
  • 129
    • 0018862650 scopus 로고
    • Stimulation of ornithine decarboxylase activity by insulin in developing rat brain
    • Roger LJ, Fellows RE. Stimulation of ornithine decarboxylase activity by insulin in developing rat brain. Endocrinology (1980) 106(2):619-25. doi:10.1210/endo-106-2-619
    • (1980) Endocrinology , vol.106 , Issue.2 , pp. 619-625
    • Roger, L.J.1    Fellows, R.E.2
  • 130
    • 0027391758 scopus 로고
    • The cellular and physiological actions of insulin in the central nervous system
    • Wozniak M, Rydzewski B, Baker SP, Raizada MK. The cellular and physiological actions of insulin in the central nervous system. Neurochem Int (1993) 22(1):1-10. doi:10.1016/0197-0186(93)90062-A
    • (1993) Neurochem Int , vol.22 , Issue.1 , pp. 1-10
    • Wozniak, M.1    Rydzewski, B.2    Baker, S.P.3    Raizada, M.K.4
  • 131
    • 0043127453 scopus 로고    scopus 로고
    • Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation
    • Schubert M, Brazil DP, Burks DJ, Kushner JA, Ye J, Flint CL, et al. Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. J Neurosci (2003) 23(18):7084-92.
    • (2003) J Neurosci , vol.23 , Issue.18 , pp. 7084-7092
    • Schubert, M.1    Brazil, D.P.2    Burks, D.J.3    Kushner, J.A.4    Ye, J.5    Flint, C.L.6
  • 133
    • 0018909905 scopus 로고
    • Binding of [125I]insulin to specific receptors and stimulation of nucleotide incorporation in cells cultured from rat brain
    • Raizada MK, Yang JW, Fellows RE. Binding of [125I]insulin to specific receptors and stimulation of nucleotide incorporation in cells cultured from rat brain. Brain Res (1980) 200(2):389-400. doi:10.1016/0006-8993(80)90929-4
    • (1980) Brain Res , vol.200 , Issue.2 , pp. 389-400
    • Raizada, M.K.1    Yang, J.W.2    Fellows, R.E.3
  • 134
    • 0023772541 scopus 로고
    • Functional properties of the subtype of insulin receptor found on neurons
    • Heidenreich KA, de Vellis G, Gilmore PR. Functional properties of the subtype of insulin receptor found on neurons. J Neurochem (1988) 51(3):878-87. doi:10.1111/j.1471-4159.1988.tb01824.x
    • (1988) J Neurochem , vol.51 , Issue.3 , pp. 878-887
    • Heidenreich, K.A.1    de Vellis, G.2    Gilmore, P.R.3
  • 135
    • 0022152964 scopus 로고
    • Insulin stimulates macromolecular synthesis in cultured glial cells from rat brain
    • Clarke DW, Boyd FT Jr, Kappy MS, Raizada MK. Insulin stimulates macromolecular synthesis in cultured glial cells from rat brain. Am J Physiol (1985) 249(5 Pt 1):C484-9.
    • (1985) Am J Physiol , vol.249 , Issue.5 , pp. C484-C489
    • Clarke, D.W.1    Boyd F.T, Jr.2    Kappy, M.S.3    Raizada, M.K.4
  • 136
    • 0024426766 scopus 로고
    • Insulin receptors mediate growth effects in cultured fetal neurons. II. Activation of a protein kinase that phosphorylates ribosomal protein S6
    • Heidenreich KA, Toledo SP. Insulin receptors mediate growth effects in cultured fetal neurons. II. Activation of a protein kinase that phosphorylates ribosomal protein S6. Endocrinology (1989) 125(3):1458-63. doi:10.1210/endo-125-3-1451
    • (1989) Endocrinology , vol.125 , Issue.3 , pp. 1458-1463
    • Heidenreich, K.A.1    Toledo, S.P.2
  • 137
    • 0021416923 scopus 로고
    • Insulin and insulin-like growth factor II permit nerve growth factor binding and the neurite formation response in cultured human neuroblastoma cells
    • Recio-Pinto E, Lang FF, Ishii DN. Insulin and insulin-like growth factor II permit nerve growth factor binding and the neurite formation response in cultured human neuroblastoma cells. Proc Natl Acad Sci U S A (1984) 81(8):2562-6. doi:10.1073/pnas.81.8.2562
    • (1984) Proc Natl Acad Sci U S A , vol.81 , Issue.8 , pp. 2562-2566
    • Recio-Pinto, E.1    Lang, F.F.2    Ishii, D.N.3
  • 138
    • 0027328932 scopus 로고
    • Neurite promoting activity of insulin, insulin-like growth factor I and nerve growth factor on spinal motoneurons is astrocyte dependent
    • Ang LC, Bhaumick B, Juurlink BH. Neurite promoting activity of insulin, insulin-like growth factor I and nerve growth factor on spinal motoneurons is astrocyte dependent. Brain Res Dev Brain Res (1993) 74(1):83-8. doi:10.1016/0165-3806(93)90086-P
    • (1993) Brain Res Dev Brain Res , vol.74 , Issue.1 , pp. 83-88
    • Ang, L.C.1    Bhaumick, B.2    Juurlink, B.H.3
  • 139
    • 73049085414 scopus 로고    scopus 로고
    • Synergistic effect of glucagon-like peptide 2 (GLP-2) and of key growth factors on the proliferation of cultured rat astrocytes. Evidence for reciprocal upregulation of the mRNAs for GLP-2 and IGF-I receptors
    • Velazquez E, Blazquez E, Ruiz-Albusac JM. Synergistic effect of glucagon-like peptide 2 (GLP-2) and of key growth factors on the proliferation of cultured rat astrocytes. Evidence for reciprocal upregulation of the mRNAs for GLP-2 and IGF-I receptors. Mol Neurobiol (2009) 40(2):183-93. doi:10.1007/s12035-009-8080-1
    • (2009) Mol Neurobiol , vol.40 , Issue.2 , pp. 183-193
    • Velazquez, E.1    Blazquez, E.2    Ruiz-Albusac, J.M.3
  • 140
    • 79959576516 scopus 로고    scopus 로고
    • Insulin promotes glycogen storage and cell proliferation in primary human astrocytes
    • Heni M, Hennige AM, Peter A, Siegel-Axel D, Ordelheide AM, Krebs N, et al. Insulin promotes glycogen storage and cell proliferation in primary human astrocytes. PLoS One (2011) 6(6):e21594. doi:10.1371/journal.pone.0021594
    • (2011) PLoS One , vol.6 , Issue.6 , pp. e21594
    • Heni, M.1    Hennige, A.M.2    Peter, A.3    Siegel-Axel, D.4    Ordelheide, A.M.5    Krebs, N.6
  • 141
    • 0025143907 scopus 로고
    • Insulin stimulates the activity of a novel protein kinase C, PKC-epsilon, in cultured fetal chick neurons
    • Heidenreich KA, Toledo SP, Brunton LL, Watson MJ, Daniel-Issakani S, Strulovici B. Insulin stimulates the activity of a novel protein kinase C, PKC-epsilon, in cultured fetal chick neurons. J Biol Chem (1990) 265(25):15076-82.
    • (1990) J Biol Chem , vol.265 , Issue.25 , pp. 15076-15082
    • Heidenreich, K.A.1    Toledo, S.P.2    Brunton, L.L.3    Watson, M.J.4    Daniel-Issakani, S.5    Strulovici, B.6
  • 142
    • 0025143525 scopus 로고
    • Insulin and neuroparsin promote neurite outgrowth in cultured locust CNS
    • Vanhems E, Delbos M, Girardie J. Insulin and neuroparsin promote neurite outgrowth in cultured locust CNS. Eur J Neurosci (1990) 2(9):776-82. doi:10.1111/j.1460-9568.1990.tb00468.x
    • (1990) Eur J Neurosci , vol.2 , Issue.9 , pp. 776-782
    • Vanhems, E.1    Delbos, M.2    Girardie, J.3
  • 143
    • 0026068142 scopus 로고
    • Regulation of protein phosphorylation by insulin and insulin-like growth factors in cultured fetal neurons
    • Heidenreich KA, Toledo SP, Kenner KA. Regulation of protein phosphorylation by insulin and insulin-like growth factors in cultured fetal neurons. Adv Exp Med Biol (1991) 293:379-84. doi:10.1007/978-1-4684-5949-4_33
    • (1991) Adv Exp Med Biol , vol.293 , pp. 379-384
    • Heidenreich, K.A.1    Toledo, S.P.2    Kenner, K.A.3
  • 144
    • 0027286297 scopus 로고
    • Insulin stimulates phosphatidylinositol 3-kinase activity in rat neuronal primary cultures
    • Patel RA, Kurian P, Raizada MK, Crews FT. Insulin stimulates phosphatidylinositol 3-kinase activity in rat neuronal primary cultures. J Neurochem (1993) 61(1):360-3. doi:10.1111/j.1471-4159.1993.tb03578.x
    • (1993) J Neurochem , vol.61 , Issue.1 , pp. 360-363
    • Patel, R.A.1    Kurian, P.2    Raizada, M.K.3    Crews, F.T.4
  • 145
    • 24044474689 scopus 로고    scopus 로고
    • Insulin stimulates postsynaptic density-95 protein translation via the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin signaling pathway
    • Lee CC, Huang CC, Wu MY, Hsu KS. Insulin stimulates postsynaptic density-95 protein translation via the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin signaling pathway. J Biol Chem (2005) 280(18):18543-50. doi:10.1074/jbc.M414112200
    • (2005) J Biol Chem , vol.280 , Issue.18 , pp. 18543-18550
    • Lee, C.C.1    Huang, C.C.2    Wu, M.Y.3    Hsu, K.S.4
  • 146
    • 79960016949 scopus 로고    scopus 로고
    • Insulin promotes dendritic spine and synapse formation by the PI3K/Akt/mTOR and Rac1 signaling pathways
    • Lee CC, Huang CC, Hsu KS. Insulin promotes dendritic spine and synapse formation by the PI3K/Akt/mTOR and Rac1 signaling pathways. Neuropharmacology (2011) 61(4):867-79. doi:10.1016/j.neuropharm.2011.06.003
    • (2011) Neuropharmacology , vol.61 , Issue.4 , pp. 867-879
    • Lee, C.C.1    Huang, C.C.2    Hsu, K.S.3
  • 147
    • 80053362920 scopus 로고    scopus 로고
    • Insulin-induced neurite-like process outgrowth: acceleration of tau protein synthesis via a phosphoinositide 3-kinase mammalian target of rapamycin pathway
    • Nemoto T, Yanagita T, Satoh S, Maruta T, Kanai T, Murakami M, et al. Insulin-induced neurite-like process outgrowth: acceleration of tau protein synthesis via a phosphoinositide 3-kinase mammalian target of rapamycin pathway. Neurochem Int (2011) 59(6):880-8. doi:10.1016/j.neuint.2011.08.002
    • (2011) Neurochem Int , vol.59 , Issue.6 , pp. 880-888
    • Nemoto, T.1    Yanagita, T.2    Satoh, S.3    Maruta, T.4    Kanai, T.5    Murakami, M.6
  • 148
    • 0032547885 scopus 로고    scopus 로고
    • Effects of brain endogenous insulin on neurofilament and MAPK in fetal rat neuron cell cultures
    • Schechter R, Yanovitch T, Abboud M, Johnson G III, Gaskins J. Effects of brain endogenous insulin on neurofilament and MAPK in fetal rat neuron cell cultures. Brain Res (1998) 808(2):270-8. doi:10.1016/S0006-8993(98)00842-7
    • (1998) Brain Res , vol.808 , Issue.2 , pp. 270-278
    • Schechter, R.1    Yanovitch, T.2    Abboud, M.3    Johnson, G.4    Gaskins, J.5
  • 149
    • 84882591485 scopus 로고    scopus 로고
    • Insulin concentration is critical in culturing human neural stem cells and neurons
    • Rhee YH, Choi M, Lee HS, Park CH, Kim SM, Yi SH, et al. Insulin concentration is critical in culturing human neural stem cells and neurons. Cell Death Dis (2013) 4:e766. doi:10.1038/cddis.2013.295
    • (2013) Cell Death Dis , vol.4 , pp. e766
    • Rhee, Y.H.1    Choi, M.2    Lee, H.S.3    Park, C.H.4    Kim, S.M.5    Yi, S.H.6
  • 150
    • 5344243134 scopus 로고    scopus 로고
    • Neuronal differentiation: TOR and insulin receptor pathways set the tempo
    • Leopold P. Neuronal differentiation: TOR and insulin receptor pathways set the tempo. Cell (2004) 119(1):4-5. doi:10.1016/j.cell.2004.09.024
    • (2004) Cell , vol.119 , Issue.1 , pp. 4-5
    • Leopold, P.1
  • 151
    • 0006460824 scopus 로고    scopus 로고
    • Phosphatidylinositol 3-kinase-mediated regulation of neuronal apoptosis and necrosis by insulin and IGF-I
    • Ryu BR, Ko HW, Jou I, Noh JS, Gwag BJ. Phosphatidylinositol 3-kinase-mediated regulation of neuronal apoptosis and necrosis by insulin and IGF-I. J Neurobiol (1999) 39(4):536-46. doi:10.1002/(SICI)1097-4695(19990615)39:4<536::AID-NEU7>3.3.CO;2-A
    • (1999) J Neurobiol , vol.39 , Issue.4 , pp. 536-546
    • Ryu, B.R.1    Ko, H.W.2    Jou, I.3    Noh, J.S.4    Gwag, B.J.5
  • 153
    • 33644638104 scopus 로고    scopus 로고
    • Hyperglycemia, insulin, and acute ischemic stroke: a mechanistic justification for a trial of insulin infusion therapy
    • Garg R, Chaudhuri A, Munschauer F, Dandona P. Hyperglycemia, insulin, and acute ischemic stroke: a mechanistic justification for a trial of insulin infusion therapy. Stroke (2006) 37(1):267-73. doi:10.1161/01.STR.0000195175.29487.30
    • (2006) Stroke , vol.37 , Issue.1 , pp. 267-273
    • Garg, R.1    Chaudhuri, A.2    Munschauer, F.3    Dandona, P.4
  • 154
    • 33750882488 scopus 로고    scopus 로고
    • Insulin restores metabolic function in cultured cortical neurons subjected to oxidative stress
    • Duarte AI, Proenca T, Oliveira CR, Santos MS, Rego AC. Insulin restores metabolic function in cultured cortical neurons subjected to oxidative stress. Diabetes (2006) 55(10):2863-70. doi:10.2337/db06-0030
    • (2006) Diabetes , vol.55 , Issue.10 , pp. 2863-2870
    • Duarte, A.I.1    Proenca, T.2    Oliveira, C.R.3    Santos, M.S.4    Rego, A.C.5
  • 155
    • 0037684715 scopus 로고    scopus 로고
    • Insulin affects synaptosomal GABA and glutamate transport under oxidative stress conditions
    • Duarte AI, Santos MS, Seica R, de Oliveira CR. Insulin affects synaptosomal GABA and glutamate transport under oxidative stress conditions. Brain Res (2003) 977(1):23-30. doi:10.1016/S0006-8993(03)02679-9
    • (2003) Brain Res , vol.977 , Issue.1 , pp. 23-30
    • Duarte, A.I.1    Santos, M.S.2    Seica, R.3    de Oliveira, C.R.4
  • 156
    • 0025720431 scopus 로고
    • Serum urate as an antioxidant for ascorbic acid
    • Sevanian A, Davies KJ, Hochstein P. Serum urate as an antioxidant for ascorbic acid. Am J Clin Nutr (1991) 54(6 Suppl):1129S-34S.
    • (1991) Am J Clin Nutr , vol.54 , Issue.6 SUPPL , pp. 1129S-1134S
    • Sevanian, A.1    Davies, K.J.2    Hochstein, P.3
  • 157
    • 0031709432 scopus 로고    scopus 로고
    • Insulin, blood glucose levels, and ischemic brain damage
    • Auer RN. Insulin, blood glucose levels, and ischemic brain damage. Neurology (1998) 51(3 Suppl 3):S39-43. doi:10.1212/WNL.51.3_Suppl_3.S39
    • (1998) Neurology , vol.51 , Issue.3 , pp. S39-43
    • Auer, R.N.1
  • 158
    • 0029058085 scopus 로고
    • Insulin elevates hippocampal GABA levels during ischemia. This is independent of its hypoglycemic effect
    • Shuaib A, Ijaz MS, Waqar T, Voll C, Kanthan R, Miyashita H, et al. Insulin elevates hippocampal GABA levels during ischemia. This is independent of its hypoglycemic effect. Neuroscience (1995) 67(4):809-14. doi:10.1016/0306-4522(95)00093-X
    • (1995) Neuroscience , vol.67 , Issue.4 , pp. 809-814
    • Shuaib, A.1    Ijaz, M.S.2    Waqar, T.3    Voll, C.4    Kanthan, R.5    Miyashita, H.6
  • 159
    • 0021944111 scopus 로고
    • Hyperinsulinemia suppresses glucose utilization in specific brain regions: in vivo studies using the euglycemic clamp in the rat
    • Grunstein HS, James DE, Storlien LH, Smythe GA, Kraegen EW. Hyperinsulinemia suppresses glucose utilization in specific brain regions: in vivo studies using the euglycemic clamp in the rat. Endocrinology (1985) 116(2):604-10. doi:10.1210/endo-116-2-604
    • (1985) Endocrinology , vol.116 , Issue.2 , pp. 604-610
    • Grunstein, H.S.1    James, D.E.2    Storlien, L.H.3    Smythe, G.A.4    Kraegen, E.W.5
  • 160
    • 0026041696 scopus 로고
    • Insulin attenuates ischemic brain damage independent of its hypoglycemic effect
    • Voll CL, Auer RN. Insulin attenuates ischemic brain damage independent of its hypoglycemic effect. J Cereb Blood Flow Metab (1991) 11(6):1006-14. doi:10.1038/jcbfm.1991.168
    • (1991) J Cereb Blood Flow Metab , vol.11 , Issue.6 , pp. 1006-1014
    • Voll, C.L.1    Auer, R.N.2
  • 161
    • 67650986109 scopus 로고    scopus 로고
    • In vivo electrophysiological effects of insulin in the rat brain
    • Kovacs P, Hajnal A. In vivo electrophysiological effects of insulin in the rat brain. Neuropeptides (2009) 43(4):283-93. doi:10.1016/j.npep.2009.05.006
    • (2009) Neuropeptides , vol.43 , Issue.4 , pp. 283-293
    • Kovacs, P.1    Hajnal, A.2
  • 162
    • 0037523369 scopus 로고    scopus 로고
    • Control of synaptic strength, a novel function of Akt
    • Wang Q, Liu L, Pei L, Ju W, Ahmadian G, Lu J, et al. Control of synaptic strength, a novel function of Akt. Neuron (2003) 38(6):915-28. doi:10.1016/S0896-6273(03)00356-8
    • (2003) Neuron , vol.38 , Issue.6 , pp. 915-928
    • Wang, Q.1    Liu, L.2    Pei, L.3    Ju, W.4    Ahmadian, G.5    Lu, J.6
  • 163
    • 13944272576 scopus 로고    scopus 로고
    • The role of insulin receptor signaling in the brain
    • Plum L, Schubert M, Bruning JC. The role of insulin receptor signaling in the brain. Trends Endocrinol Metab (2005) 16(2):59-65. doi:10.1016/j.tem.2005.01.008
    • (2005) Trends Endocrinol Metab , vol.16 , Issue.2 , pp. 59-65
    • Plum, L.1    Schubert, M.2    Bruning, J.C.3
  • 164
    • 0031029133 scopus 로고    scopus 로고
    • Regulation by insulin of a unique neuronal Ca2+ pool and of neuropeptide secretion
    • Jonas EA, Knox RJ, Smith TC, Wayne NL, Connor JA, Kaczmarek LK. Regulation by insulin of a unique neuronal Ca2+ pool and of neuropeptide secretion. Nature (1997) 385(6614):343-6. doi:10.1038/385343a0
    • (1997) Nature , vol.385 , Issue.6614 , pp. 343-346
    • Jonas, E.A.1    Knox, R.J.2    Smith, T.C.3    Wayne, N.L.4    Connor, J.A.5    Kaczmarek, L.K.6
  • 165
    • 0022371436 scopus 로고
    • Insulin receptors and insulin modulation of norepinephrine uptake in neuronal cultures from rat brain
    • Boyd FT Jr, Clarke DW, Muther TF, Raizada MK. Insulin receptors and insulin modulation of norepinephrine uptake in neuronal cultures from rat brain. J Biol Chem (1985) 260(29):15880-4.
    • (1985) J Biol Chem , vol.260 , Issue.29 , pp. 15880-15884
    • Boyd F.T, Jr.1    Clarke, D.W.2    Muther, T.F.3    Raizada, M.K.4
  • 167
    • 0019865177 scopus 로고
    • Dopamine receptor binding is increased in diabetic rats
    • Lozovsky D, Saller CF, Kopin IJ. Dopamine receptor binding is increased in diabetic rats. Science (1981) 214(4524):1031-3. doi:10.1126/science.6458088
    • (1981) Science , vol.214 , Issue.4524 , pp. 1031-1033
    • Lozovsky, D.1    Saller, C.F.2    Kopin, I.J.3
  • 168
    • 0022408010 scopus 로고
    • Modulation of dopamine receptor supersensitivity by chronic insulin: implication in schizophrenia
    • Lozovsky DB, Kopin IJ, Saller CF. Modulation of dopamine receptor supersensitivity by chronic insulin: implication in schizophrenia. Brain Res (1985) 343(1):190-3. doi:10.1016/0006-8993(85)91178-3
    • (1985) Brain Res , vol.343 , Issue.1 , pp. 190-193
    • Lozovsky, D.B.1    Kopin, I.J.2    Saller, C.F.3
  • 169
    • 0031964158 scopus 로고    scopus 로고
    • Insulin selectively downregulates alpha2-adrenoceptors in the arcuate and dorsomedial nucleus
    • Levin BE, Israel P, Lattemann DP. Insulin selectively downregulates alpha2-adrenoceptors in the arcuate and dorsomedial nucleus. Brain Res Bull (1998) 45(2):179-81. doi:10.1016/S0361-9230(97)00336-5
    • (1998) Brain Res Bull , vol.45 , Issue.2 , pp. 179-181
    • Levin, B.E.1    Israel, P.2    Lattemann, D.P.3
  • 170
    • 0021230425 scopus 로고
    • Stimulation of synaptosomal uptake of neurotransmitter amino acids by insulin: possible role of insulin as a neuromodulator
    • Rhoads DE, DiRocco RJ, Osburn LD, Peterson NA, Raghupathy E. Stimulation of synaptosomal uptake of neurotransmitter amino acids by insulin: possible role of insulin as a neuromodulator. Biochem Biophys Res Commun (1984) 119(3):1198-204. doi:10.1016/0006-291X(84)90903-3
    • (1984) Biochem Biophys Res Commun , vol.119 , Issue.3 , pp. 1198-1204
    • Rhoads, D.E.1    DiRocco, R.J.2    Osburn, L.D.3    Peterson, N.A.4    Raghupathy, E.5
  • 171
    • 0034012731 scopus 로고    scopus 로고
    • Intracerebroventricular insulin enhances memory in a passive-avoidance task
    • Park CR, Seeley RJ, Craft S, Woods SC. Intracerebroventricular insulin enhances memory in a passive-avoidance task. Physiol Behav (2000) 68(4):509-14. doi:10.1016/S0031-9384(99)00220-6
    • (2000) Physiol Behav , vol.68 , Issue.4 , pp. 509-514
    • Park, C.R.1    Seeley, R.J.2    Craft, S.3    Woods, S.C.4
  • 172
    • 0033521130 scopus 로고    scopus 로고
    • Brain insulin receptors and spatial memory. Correlated changes in gene expression, tyrosine phosphorylation, and signaling molecules in the hippocampus of water maze trained rats
    • Zhao W, Chen H, Xu H, Moore E, Meiri N, Quon MJ, et al. Brain insulin receptors and spatial memory. Correlated changes in gene expression, tyrosine phosphorylation, and signaling molecules in the hippocampus of water maze trained rats. J Biol Chem (1999) 274(49):34893-902. doi:10.1074/jbc.274.49.34893
    • (1999) J Biol Chem , vol.274 , Issue.49 , pp. 34893-34902
    • Zhao, W.1    Chen, H.2    Xu, H.3    Moore, E.4    Meiri, N.5    Quon, M.J.6
  • 173
    • 0024337626 scopus 로고
    • Postischemic insulin reduces spatial learning deficit following transient forebrain ischemia in rats
    • Voll CL, Whishaw IQ, Auer RN. Postischemic insulin reduces spatial learning deficit following transient forebrain ischemia in rats. Stroke (1989) 20(5):646-51. doi:10.1161/01.STR.20.5.646
    • (1989) Stroke , vol.20 , Issue.5 , pp. 646-651
    • Voll, C.L.1    Whishaw, I.Q.2    Auer, R.N.3
  • 174
    • 0031797937 scopus 로고    scopus 로고
    • Intracerebroventricular administration of streptozotocin causes long-term diminutions in learning and memory abilities and in cerebral energy metabolism in adult rats
    • Lannert H, Hoyer S. Intracerebroventricular administration of streptozotocin causes long-term diminutions in learning and memory abilities and in cerebral energy metabolism in adult rats. Behav Neurosci (1998) 112(5):1199-208. doi:10.1037/0735-7044.112.5.1199
    • (1998) Behav Neurosci , vol.112 , Issue.5 , pp. 1199-1208
    • Lannert, H.1    Hoyer, S.2
  • 176
    • 67651030409 scopus 로고    scopus 로고
    • Synaptic mechanisms for plasticity in neocortex
    • Feldman DE. Synaptic mechanisms for plasticity in neocortex. Annu Rev Neurosci (2009) 32:33-55. doi:10.1146/annurev.neuro.051508.135516
    • (2009) Annu Rev Neurosci , vol.32 , pp. 33-55
    • Feldman, D.E.1
  • 177
    • 0036829584 scopus 로고    scopus 로고
    • Regulation of AMPA receptors during synaptic plasticity
    • Song I, Huganir RL. Regulation of AMPA receptors during synaptic plasticity. Trends Neurosci (2002) 25(11):578-88. doi:10.1016/S0166-2236(02)02270-1
    • (2002) Trends Neurosci , vol.25 , Issue.11 , pp. 578-588
    • Song, I.1    Huganir, R.L.2
  • 178
    • 1842562359 scopus 로고    scopus 로고
    • An investigation into signal transduction mechanisms involved in insulin-induced long-term depression in the CA1 region of the hippocampus
    • Huang CC, Lee CC, Hsu KS. An investigation into signal transduction mechanisms involved in insulin-induced long-term depression in the CA1 region of the hippocampus. J Neurochem (2004) 89(1):217-31. doi:10.1111/j.1471-4159.2003.02307.x
    • (2004) J Neurochem , vol.89 , Issue.1 , pp. 217-231
    • Huang, C.C.1    Lee, C.C.2    Hsu, K.S.3
  • 179
    • 12144288368 scopus 로고    scopus 로고
    • Tyrosine phosphorylation of GluR2 is required for insulin-stimulated AMPA receptor endocytosis and LTD
    • Ahmadian G, Ju W, Liu L, Wyszynski M, Lee SH, Dunah AW, et al. Tyrosine phosphorylation of GluR2 is required for insulin-stimulated AMPA receptor endocytosis and LTD. EMBO J (2004) 23(5):1040-50. doi:10.1038/sj.emboj.7600126
    • (2004) EMBO J , vol.23 , Issue.5 , pp. 1040-1050
    • Ahmadian, G.1    Ju, W.2    Liu, L.3    Wyszynski, M.4    Lee, S.H.5    Dunah, A.W.6
  • 180
    • 43049093341 scopus 로고    scopus 로고
    • Involvement of nitric oxide in insulin induced memory improvement
    • Choopani S, Moosavi M, Naghdi N. Involvement of nitric oxide in insulin induced memory improvement. Peptides (2008) 29(6):898-903. doi:10.1016/j.peptides.2008.01.005
    • (2008) Peptides , vol.29 , Issue.6 , pp. 898-903
    • Choopani, S.1    Moosavi, M.2    Naghdi, N.3
  • 181
    • 21544452752 scopus 로고    scopus 로고
    • Functional characterization of des-IGF-1 action at excitatory synapses in the CA1 region of rat hippocampus
    • Ramsey MM, Adams MM, Ariwodola OJ, Sonntag WE, Weiner JL. Functional characterization of des-IGF-1 action at excitatory synapses in the CA1 region of rat hippocampus. J Neurophysiol (2005) 94(1):247-54. doi:10.1152/jn.00768.2004
    • (2005) J Neurophysiol , vol.94 , Issue.1 , pp. 247-254
    • Ramsey, M.M.1    Adams, M.M.2    Ariwodola, O.J.3    Sonntag, W.E.4    Weiner, J.L.5
  • 182
    • 33747588120 scopus 로고    scopus 로고
    • Growth hormone replacement in hypophysectomized rats affects spatial performance and hippocampal levels of NMDA receptor subunit and PSD-95 gene transcript levels
    • Le Grevès M, Zhou Q, Berg M, Le Grevès P, Fhölenhag K, Meyerson B, et al. Growth hormone replacement in hypophysectomized rats affects spatial performance and hippocampal levels of NMDA receptor subunit and PSD-95 gene transcript levels. Exp Brain Res (2006) 173(2):267-73. doi:10.1007/s00221-006-0438-2
    • (2006) Exp Brain Res , vol.173 , Issue.2 , pp. 267-273
    • Le Grevès, M.1    Zhou, Q.2    Berg, M.3    Le Grevès, P.4    Fhölenhag, K.5    Meyerson, B.6
  • 184
    • 0034745018 scopus 로고    scopus 로고
    • Cerebrospinal fluid abeta42, tau, and f2-isoprostane concentrations in patients with Alzheimer disease, other dementias, and in age-matched controls
    • Montine TJ, Kaye JA, Montine KS, McFarland L, Morrow JD, Quinn JF. Cerebrospinal fluid abeta42, tau, and f2-isoprostane concentrations in patients with Alzheimer disease, other dementias, and in age-matched controls. Arch Pathol Lab Med (2001) 125(4):510-2. doi:10.1043/0003-9985(2001)125<0510:CFATAF>2.0.CO;2
    • (2001) Arch Pathol Lab Med , vol.125 , Issue.4 , pp. 510-512
    • Montine, T.J.1    Kaye, J.A.2    Montine, K.S.3    McFarland, L.4    Morrow, J.D.5    Quinn, J.F.6
  • 185
    • 0042835437 scopus 로고    scopus 로고
    • Lipopolysaccharide-induced-neuroinflammation increases intracellular accumulation of amyloid precursor protein and amyloid beta peptide in APPswe transgenic mice
    • Sheng JG, Bora SH, Xu G, Borchelt DR, Price DL, Koliatsos VE. Lipopolysaccharide-induced-neuroinflammation increases intracellular accumulation of amyloid precursor protein and amyloid beta peptide in APPswe transgenic mice. Neurobiol Dis (2003) 14(1):133-45. doi:10.1016/S0969-9961(03)00069-X
    • (2003) Neurobiol Dis , vol.14 , Issue.1 , pp. 133-145
    • Sheng, J.G.1    Bora, S.H.2    Xu, G.3    Borchelt, D.R.4    Price, D.L.5    Koliatsos, V.E.6
  • 186
    • 0036706725 scopus 로고    scopus 로고
    • Endothelium, inflammation, and diabetes
    • Dandona P. Endothelium, inflammation, and diabetes. Curr Diab Rep (2002) 2(4):311-5. doi:10.1007/s11892-002-0019-0
    • (2002) Curr Diab Rep , vol.2 , Issue.4 , pp. 311-315
    • Dandona, P.1
  • 187
    • 0036086942 scopus 로고    scopus 로고
    • Euglycemic hyperinsulinemia augments the cytokine and endocrine responses to endotoxin in humans
    • Soop M, Duxbury H, Agwunobi AO, Gibson JM, Hopkins SJ, Childs C, et al. Euglycemic hyperinsulinemia augments the cytokine and endocrine responses to endotoxin in humans. Am J Physiol Endocrinol Metab (2002) 282(6):E1276-85. doi:10.1152/ajpendo.00535.2001
    • (2002) Am J Physiol Endocrinol Metab , vol.282 , Issue.6 , pp. E1276-E1285
    • Soop, M.1    Duxbury, H.2    Agwunobi, A.O.3    Gibson, J.M.4    Hopkins, S.J.5    Childs, C.6
  • 188
    • 26444553356 scopus 로고    scopus 로고
    • Hyperinsulinemia provokes synchronous increases in central inflammation and beta-amyloid in normal adults
    • Fishel MA, Watson GS, Montine TJ, Wang Q, Green PS, Kulstad JJ, et al. Hyperinsulinemia provokes synchronous increases in central inflammation and beta-amyloid in normal adults. Arch Neurol (2005) 62(10):1539-44. doi:10.1001/archneur.62.10.noc50112
    • (2005) Arch Neurol , vol.62 , Issue.10 , pp. 1539-1544
    • Fishel, M.A.1    Watson, G.S.2    Montine, T.J.3    Wang, Q.4    Green, P.S.5    Kulstad, J.J.6
  • 189
    • 79960880118 scopus 로고    scopus 로고
    • Assessing the contribution of inflammation in models of Alzheimer's disease
    • Johnston H, Boutin H, Allan SM. Assessing the contribution of inflammation in models of Alzheimer's disease. Biochem Soc Trans (2011) 39(4):886-90. doi:10.1042/BST0390886
    • (2011) Biochem Soc Trans , vol.39 , Issue.4 , pp. 886-890
    • Johnston, H.1    Boutin, H.2    Allan, S.M.3
  • 190
    • 34547380204 scopus 로고    scopus 로고
    • Neuroinflammation in Alzheimer's disease and Parkinson's disease: are microglia pathogenic in either disorder?
    • Rogers J, Mastroeni D, Leonard B, Joyce J, Grover A. Neuroinflammation in Alzheimer's disease and Parkinson's disease: are microglia pathogenic in either disorder? Int Rev Neurobiol (2007) 82:235-46. doi:10.1016/S0074-7742(07)82012-5
    • (2007) Int Rev Neurobiol , vol.82 , pp. 235-246
    • Rogers, J.1    Mastroeni, D.2    Leonard, B.3    Joyce, J.4    Grover, A.5
  • 191
    • 77249153620 scopus 로고    scopus 로고
    • Cognitive impairment following high fat diet consumption is associated with brain inflammation
    • Pistell PJ, Morrison CD, Gupta S, Knight AG, Keller JN, Ingram DK, et al. Cognitive impairment following high fat diet consumption is associated with brain inflammation. J Neuroimmunol (2010) 219(1-2):25-32. doi:10.1016/j.jneuroim.2009.11.010
    • (2010) J Neuroimmunol , vol.219 , Issue.1-2 , pp. 25-32
    • Pistell, P.J.1    Morrison, C.D.2    Gupta, S.3    Knight, A.G.4    Keller, J.N.5    Ingram, D.K.6
  • 192
    • 53549097355 scopus 로고    scopus 로고
    • Insulin resistance and Alzheimer's disease: molecular links & clinical implications
    • Neumann KF, Rojo L, Navarrete LP, Farias G, Reyes P, Maccioni RB. Insulin resistance and Alzheimer's disease: molecular links & clinical implications. Curr Alzheimer Res (2008) 5(5):438-47. doi:10.2174/156720508785908919
    • (2008) Curr Alzheimer Res , vol.5 , Issue.5 , pp. 438-447
    • Neumann, K.F.1    Rojo, L.2    Navarrete, L.P.3    Farias, G.4    Reyes, P.5    Maccioni, R.B.6
  • 193
    • 0037289757 scopus 로고    scopus 로고
    • The induction of the TNFalpha death domain signaling pathway in Alzheimer's disease brain
    • Zhao M, Cribbs DH, Anderson AJ, Cummings BJ, Su JH, Wasserman AJ, et al. The induction of the TNFalpha death domain signaling pathway in Alzheimer's disease brain. Neurochem Res (2003) 28(2):307-18. doi:10.1023/A:1022337519035
    • (2003) Neurochem Res , vol.28 , Issue.2 , pp. 307-318
    • Zhao, M.1    Cribbs, D.H.2    Anderson, A.J.3    Cummings, B.J.4    Su, J.H.5    Wasserman, A.J.6
  • 194
    • 0037836085 scopus 로고    scopus 로고
    • Soluble tumor necrosis factor-alpha receptors in young obese subjects with normal and impaired glucose tolerance
    • Dzienis-Straczkowska S, Straczkowski M, Szelachowska M, Stepien A, Kowalska I, Kinalska I. Soluble tumor necrosis factor-alpha receptors in young obese subjects with normal and impaired glucose tolerance. Diabetes Care (2003) 26(3):875-80. doi:10.2337/diacare.26.3.875
    • (2003) Diabetes Care , vol.26 , Issue.3 , pp. 875-880
    • Dzienis-Straczkowska, S.1    Straczkowski, M.2    Szelachowska, M.3    Stepien, A.4    Kowalska, I.5    Kinalska, I.6
  • 195
    • 0034524936 scopus 로고    scopus 로고
    • Variations in plasma soluble tumour necrosis factor receptors after diet-induced weight loss in obesity
    • Bastard JP, Jardel C, Bruckert E, Vidal H, Hainque B. Variations in plasma soluble tumour necrosis factor receptors after diet-induced weight loss in obesity. Diabetes Obes Metab (2000) 2(5):323-5. doi:10.1046/j.1463-1326.2000.00090.x
    • (2000) Diabetes Obes Metab , vol.2 , Issue.5 , pp. 323-325
    • Bastard, J.P.1    Jardel, C.2    Bruckert, E.3    Vidal, H.4    Hainque, B.5
  • 196
    • 64249121164 scopus 로고    scopus 로고
    • A look inside the diabetic brain: contributors to diabetes-induced brain aging
    • Wrighten SA, Piroli GG, Grillo CA, Reagan LP. A look inside the diabetic brain: contributors to diabetes-induced brain aging. Biochim Biophys Acta (2009) 1792(5):444-53. doi:10.1016/j.bbadis.2008.10.013
    • (2009) Biochim Biophys Acta , vol.1792 , Issue.5 , pp. 444-453
    • Wrighten, S.A.1    Piroli, G.G.2    Grillo, C.A.3    Reagan, L.P.4
  • 197
    • 84892780571 scopus 로고    scopus 로고
    • Brain insulin dysregulation: implication for neurological and neuropsychiatric disorders
    • Ghasemi R, Dargahi L, Haeri A, Moosavi M, Mohamed Z, Ahmadiani A. Brain insulin dysregulation: implication for neurological and neuropsychiatric disorders. Mol Neurobiol (2013) 47(3):1045-65. doi:10.1007/s12035-013-8404-z
    • (2013) Mol Neurobiol , vol.47 , Issue.3 , pp. 1045-1065
    • Ghasemi, R.1    Dargahi, L.2    Haeri, A.3    Moosavi, M.4    Mohamed, Z.5    Ahmadiani, A.6
  • 198
    • 77951875902 scopus 로고    scopus 로고
    • Role of glycogen synthase kinase-3 in Alzheimer's disease pathogenesis and glycogen synthase kinase-3 inhibitors
    • Avila J, Wandosell F, Hernandez F. Role of glycogen synthase kinase-3 in Alzheimer's disease pathogenesis and glycogen synthase kinase-3 inhibitors. Expert Rev Neurother (2010) 10(5):703-10. doi:10.1586/ern.10.40
    • (2010) Expert Rev Neurother , vol.10 , Issue.5 , pp. 703-710
    • Avila, J.1    Wandosell, F.2    Hernandez, F.3
  • 199
    • 0029587224 scopus 로고
    • Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B
    • Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature (1995) 378(6559):785-9. doi:10.1038/378785a0
    • (1995) Nature , vol.378 , Issue.6559 , pp. 785-789
    • Cross, D.A.1    Alessi, D.R.2    Cohen, P.3    Andjelkovich, M.4    Hemmings, B.A.5
  • 200
    • 34547691071 scopus 로고    scopus 로고
    • The role of GSK3 in glucose homeostasis and the development of insulin resistance
    • Lee J, Kim MS. The role of GSK3 in glucose homeostasis and the development of insulin resistance. Diabetes Res Clin Pract (2007) 77(Suppl 1):S49-57. doi:10.1016/j.diabres.2007.01.033
    • (2007) Diabetes Res Clin Pract , vol.77 , pp. S49-57
    • Lee, J.1    Kim, M.S.2
  • 201
    • 52049094596 scopus 로고    scopus 로고
    • Differential regulation of STAT family members by glycogen synthase kinase-3
    • Beurel E, Jope RS. Differential regulation of STAT family members by glycogen synthase kinase-3. J Biol Chem (2008) 283(32):21934-44. doi:10.1074/jbc.M802481200
    • (2008) J Biol Chem , vol.283 , Issue.32 , pp. 21934-21944
    • Beurel, E.1    Jope, R.S.2
  • 202
    • 23944486750 scopus 로고    scopus 로고
    • Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3
    • Martin M, Rehani K, Jope RS, Michalek SM. Toll-like receptor-mediated cytokine production is differentially regulated by glycogen synthase kinase 3. Nat Immunol (2005) 6(8):777-84. doi:10.1038/ni1221
    • (2005) Nat Immunol , vol.6 , Issue.8 , pp. 777-784
    • Martin, M.1    Rehani, K.2    Jope, R.S.3    Michalek, S.M.4
  • 203
    • 77957970155 scopus 로고    scopus 로고
    • Neuroinflammation, oxidative stress and the pathogenesis of Alzheimer's disease
    • Agostinho P, Cunha RA, Oliveira C. Neuroinflammation, oxidative stress and the pathogenesis of Alzheimer's disease. Curr Pharm Des (2010) 16(25):2766-78. doi:10.2174/138161210793176572
    • (2010) Curr Pharm Des , vol.16 , Issue.25 , pp. 2766-2778
    • Agostinho, P.1    Cunha, R.A.2    Oliveira, C.3
  • 204
    • 84907167305 scopus 로고    scopus 로고
    • Insulin resistance, neuroinflammation, and Alzheimer's disease
    • Najem D, Bamji-Mirza M, Chang N, Liu QY, Zhang W. Insulin resistance, neuroinflammation, and Alzheimer's disease. Rev Neurosci (2014) 25(4):509-25. doi:10.1515/revneuro-2013-0050
    • (2014) Rev Neurosci , vol.25 , Issue.4 , pp. 509-525
    • Najem, D.1    Bamji-Mirza, M.2    Chang, N.3    Liu, Q.Y.4    Zhang, W.5
  • 205
    • 77951918926 scopus 로고    scopus 로고
    • Macrophages, inflammation, and insulin resistance
    • Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol (2010) 72:219-46. doi:10.1146/annurev-physiol-021909-135846
    • (2010) Annu Rev Physiol , vol.72 , pp. 219-246
    • Olefsky, J.M.1    Glass, C.K.2
  • 206
    • 80053406579 scopus 로고    scopus 로고
    • Inflammation is necessary for long-term but not short-term high-fat diet-induced insulin resistance
    • Lee YS, Li P, Huh JY, Hwang IJ, Lu M, Kim JI, et al. Inflammation is necessary for long-term but not short-term high-fat diet-induced insulin resistance. Diabetes (2011) 60(10):2474-83. doi:10.2337/db11-0194
    • (2011) Diabetes , vol.60 , Issue.10 , pp. 2474-2483
    • Lee, Y.S.1    Li, P.2    Huh, J.Y.3    Hwang, I.J.4    Lu, M.5    Kim, J.I.6
  • 207
    • 0028031569 scopus 로고
    • Reduced tyrosine kinase activity of the insulin receptor in obesity-diabetes. Central role of tumor necrosis factor-alpha
    • Hotamisligil GS, Budavari A, Murray D, Spiegelman BM. Reduced tyrosine kinase activity of the insulin receptor in obesity-diabetes. Central role of tumor necrosis factor-alpha. J Clin Invest (1994) 94(4):1543-9.
    • (1994) J Clin Invest , vol.94 , Issue.4 , pp. 1543-1549
    • Hotamisligil, G.S.1    Budavari, A.2    Murray, D.3    Spiegelman, B.M.4
  • 208
    • 84855457188 scopus 로고    scopus 로고
    • The renin-angiotensin system: a link between obesity, inflammation and insulin resistance
    • Kalupahana NS, Moustaid-Moussa N. The renin-angiotensin system: a link between obesity, inflammation and insulin resistance. Obes Rev (2012) 13(2):136-49. doi:10.1111/j.1467-789X.2011.00942.x
    • (2012) Obes Rev , vol.13 , Issue.2 , pp. 136-149
    • Kalupahana, N.S.1    Moustaid-Moussa, N.2
  • 209
    • 18244370762 scopus 로고    scopus 로고
    • Inflammation, stress, and diabetes
    • Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest (2005) 115(5):1111-9. doi:10.1172/JCI25102
    • (2005) J Clin Invest , vol.115 , Issue.5 , pp. 1111-1119
    • Wellen, K.E.1    Hotamisligil, G.S.2
  • 210
    • 79955008199 scopus 로고    scopus 로고
    • Overweight worsens apoptosis, neuroinflammation and blood-brain barrier damage after hypoxic ischemia in neonatal brain through JNK hyperactivation
    • Tu YF, Tsai YS, Wang LW, Wu HC, Huang CC, Ho CJ. Overweight worsens apoptosis, neuroinflammation and blood-brain barrier damage after hypoxic ischemia in neonatal brain through JNK hyperactivation. J Neuroinflammation (2011) 8:40. doi:10.1186/1742-2094-8-40
    • (2011) J Neuroinflammation , vol.8 , pp. 40
    • Tu, Y.F.1    Tsai, Y.S.2    Wang, L.W.3    Wu, H.C.4    Huang, C.C.5    Ho, C.J.6
  • 211
    • 84860903172 scopus 로고    scopus 로고
    • Toll-like receptors 2 and 4 impair insulin-mediated brain activity by interleukin-6 and osteopontin and alter sleep architecture
    • Sartorius T, Lutz SZ, Hoene M, Waak J, Peter A, Weigert C, et al. Toll-like receptors 2 and 4 impair insulin-mediated brain activity by interleukin-6 and osteopontin and alter sleep architecture. FASEB J (2012) 26(5):1799-809. doi:10.1096/fj.11-191023
    • (2012) FASEB J , vol.26 , Issue.5 , pp. 1799-1809
    • Sartorius, T.1    Lutz, S.Z.2    Hoene, M.3    Waak, J.4    Peter, A.5    Weigert, C.6
  • 212
    • 79952316899 scopus 로고    scopus 로고
    • Low-grade hypothalamic inflammation leads to defective thermogenesis, insulin resistance, and impaired insulin secretion
    • Arruda AP, Milanski M, Coope A, Torsoni AS, Ropelle E, Carvalho DP, et al. Low-grade hypothalamic inflammation leads to defective thermogenesis, insulin resistance, and impaired insulin secretion. Endocrinology (2011) 152(4):1314-26. doi:10.1210/en.2010-0659
    • (2011) Endocrinology , vol.152 , Issue.4 , pp. 1314-1326
    • Arruda, A.P.1    Milanski, M.2    Coope, A.3    Torsoni, A.S.4    Ropelle, E.5    Carvalho, D.P.6
  • 213
    • 78650472774 scopus 로고    scopus 로고
    • Toll-like receptors: linking inflammation to metabolism
    • Konner AC, Bruning JC. Toll-like receptors: linking inflammation to metabolism. Trends Endocrinol Metab (2011) 22(1):16-23. doi:10.1016/j.tem.2010.08.007
    • (2011) Trends Endocrinol Metab , vol.22 , Issue.1 , pp. 16-23
    • Konner, A.C.1    Bruning, J.C.2
  • 214
    • 0033378682 scopus 로고    scopus 로고
    • A disturbance in the neuronal insulin receptor signal transduction in sporadic Alzheimer's disease
    • Frolich L, Blum-Degen D, Riederer P, Hoyer S. A disturbance in the neuronal insulin receptor signal transduction in sporadic Alzheimer's disease. Ann N Y Acad Sci (1999) 893:290-3. doi:10.1111/j.1749-6632.1999.tb07839.x
    • (1999) Ann N Y Acad Sci , vol.893 , pp. 290-293
    • Frolich, L.1    Blum-Degen, D.2    Riederer, P.3    Hoyer, S.4
  • 215
    • 67650544970 scopus 로고    scopus 로고
    • Insulin receptor dysfunction impairs cellular clearance of neurotoxic oligomeric a{beta}
    • Zhao WQ, Lacor PN, Chen H, Lambert MP, Quon MJ, Krafft GA, et al. Insulin receptor dysfunction impairs cellular clearance of neurotoxic oligomeric a{beta}. J Biol Chem (2009) 284(28):18742-53. doi:10.1074/jbc.M109.011015
    • (2009) J Biol Chem , vol.284 , Issue.28 , pp. 18742-18753
    • Zhao, W.Q.1    Lacor, P.N.2    Chen, H.3    Lambert, M.P.4    Quon, M.J.5    Krafft, G.A.6
  • 216
    • 18744438248 scopus 로고    scopus 로고
    • Alzheimer's beta-amyloid peptides compete for insulin binding to the insulin receptor
    • Xie L, Helmerhorst E, Taddei K, Plewright B, Van Bronswijk W, Martins R. Alzheimer's beta-amyloid peptides compete for insulin binding to the insulin receptor. J Neurosci (2002) 22(10):RC221.
    • (2002) J Neurosci , vol.22 , Issue.10 , pp. RC221
    • Xie, L.1    Helmerhorst, E.2    Taddei, K.3    Plewright, B.4    Van Bronswijk, W.5    Martins, R.6
  • 217
    • 65249113325 scopus 로고    scopus 로고
    • The insulin/Akt signaling pathway is targeted by intracellular beta-amyloid
    • Lee HK, Kumar P, Fu Q, Rosen KM, Querfurth HW. The insulin/Akt signaling pathway is targeted by intracellular beta-amyloid. Mol Biol Cell (2009) 20(5):1533-44. doi:10.1091/mbc.E08-07-0777
    • (2009) Mol Biol Cell , vol.20 , Issue.5 , pp. 1533-1544
    • Lee, H.K.1    Kumar, P.2    Fu, Q.3    Rosen, K.M.4    Querfurth, H.W.5
  • 218
    • 79960881826 scopus 로고    scopus 로고
    • Diabetes as a risk factor for Alzheimer's disease: insulin signalling impairment in the brain as an alternative model of Alzheimer's disease
    • Holscher C. Diabetes as a risk factor for Alzheimer's disease: insulin signalling impairment in the brain as an alternative model of Alzheimer's disease. Biochem Soc Trans (2011) 39(4):891-7. doi:10.1042/BST0390891
    • (2011) Biochem Soc Trans , vol.39 , Issue.4 , pp. 891-897
    • Holscher, C.1
  • 219
    • 79960555700 scopus 로고    scopus 로고
    • Possible implications of insulin resistance and glucose metabolism in Alzheimer's disease pathogenesis
    • Bosco D, Fava A, Plastino M, Montalcini T, Pujia A. Possible implications of insulin resistance and glucose metabolism in Alzheimer's disease pathogenesis. J Cell Mol Med (2011) 15(9):1807-21. doi:10.1111/j.1582-4934.2011.01318.x
    • (2011) J Cell Mol Med , vol.15 , Issue.9 , pp. 1807-1821
    • Bosco, D.1    Fava, A.2    Plastino, M.3    Montalcini, T.4    Pujia, A.5
  • 220
    • 0036828769 scopus 로고    scopus 로고
    • Alzheimer's disease: treatments in discovery and development
    • Citron M. Alzheimer's disease: treatments in discovery and development. Nat Neurosci (2002) 5(Suppl):1055-7. doi:10.1038/nn940
    • (2002) Nat Neurosci , vol.5 , pp. 1055-1057
    • Citron, M.1
  • 221
    • 84857788978 scopus 로고    scopus 로고
    • Insulin signaling, glucose metabolism and mitochondria: major players in Alzheimer's disease and diabetes interrelation
    • Correia SC, Santos RX, Carvalho C, Cardoso S, Candeias E, Santos MS, et al. Insulin signaling, glucose metabolism and mitochondria: major players in Alzheimer's disease and diabetes interrelation. Brain Res (2012) 1441:64-78. doi:10.1016/j.brainres.2011.12.063
    • (2012) Brain Res , vol.1441 , pp. 64-78
    • Correia, S.C.1    Santos, R.X.2    Carvalho, C.3    Cardoso, S.4    Candeias, E.5    Santos, M.S.6
  • 222
    • 0021440736 scopus 로고
    • Aspects of the pathogenesis of type 2 diabetes
    • Efendic S, Luft R, Wajngot A. Aspects of the pathogenesis of type 2 diabetes. Endocr Rev (1984) 5(3):395-410. doi:10.1210/edrv-5-3-395
    • (1984) Endocr Rev , vol.5 , Issue.3 , pp. 395-410
    • Efendic, S.1    Luft, R.2    Wajngot, A.3
  • 223
    • 84883053304 scopus 로고    scopus 로고
    • Decoding Alzheimer's disease from perturbed cerebral glucose metabolism: implications for diagnostic and therapeutic strategies
    • Chen Z, Zhong C. Decoding Alzheimer's disease from perturbed cerebral glucose metabolism: implications for diagnostic and therapeutic strategies. Prog Neurobiol (2013) 108:21-43. doi:10.1016/j.pneurobio.2013.06.004
    • (2013) Prog Neurobiol , vol.108 , pp. 21-43
    • Chen, Z.1    Zhong, C.2
  • 224
    • 0033048453 scopus 로고    scopus 로고
    • The mechanism of islet amyloid polypeptide toxicity is membrane disruption by intermediate-sized toxic amyloid particles
    • Janson J, Ashley RH, Harrison D, McIntyre S, Butler PC. The mechanism of islet amyloid polypeptide toxicity is membrane disruption by intermediate-sized toxic amyloid particles. Diabetes (1999) 48(3):491-8. doi:10.2337/diabetes.48.3.491
    • (1999) Diabetes , vol.48 , Issue.3 , pp. 491-498
    • Janson, J.1    Ashley, R.H.2    Harrison, D.3    McIntyre, S.4    Butler, P.C.5
  • 225
    • 5344256250 scopus 로고    scopus 로고
    • Hyperinsulinemia and risk of Alzheimer disease
    • Luchsinger JA, Tang MX, Shea S, Mayeux R. Hyperinsulinemia and risk of Alzheimer disease. Neurology (2004) 63(7):1187-92. doi:10.1212/01.WNL.0000140292.04932.87
    • (2004) Neurology , vol.63 , Issue.7 , pp. 1187-1192
    • Luchsinger, J.A.1    Tang, M.X.2    Shea, S.3    Mayeux, R.4
  • 227
    • 84906987763 scopus 로고    scopus 로고
    • Altered expression of diabetes-related genes in Alzheimer's disease brains: the Hisayama study
    • Hokama M, Oka S, Leon J, Ninomiya T, Honda H, Sasaki K, et al. Altered expression of diabetes-related genes in Alzheimer's disease brains: the Hisayama study. Cereb Cortex (2013) 24(9):2476-88. doi:10.1093/cercor/bht101
    • (2013) Cereb Cortex , vol.24 , Issue.9 , pp. 2476-2488
    • Hokama, M.1    Oka, S.2    Leon, J.3    Ninomiya, T.4    Honda, H.5    Sasaki, K.6
  • 228
    • 68949089312 scopus 로고    scopus 로고
    • Alzheimer's disease is type 3 diabetes-evidence reviewed
    • de la Monte SM, Wands JR. Alzheimer's disease is type 3 diabetes-evidence reviewed. J Diabetes Sci Technol (2008) 2(6):1101-13. doi:10.1177/193229680800200619
    • (2008) J Diabetes Sci Technol , vol.2 , Issue.6 , pp. 1101-1113
    • de la Monte, S.M.1    Wands, J.R.2
  • 229
    • 79952449386 scopus 로고    scopus 로고
    • Insulin-resistant brain state: the culprit in sporadic Alzheimer's disease?
    • Correia SC, Santos RX, Perry G, Zhu X, Moreira PI, Smith MA. Insulin-resistant brain state: the culprit in sporadic Alzheimer's disease? Ageing Res Rev (2011) 10(2):264-73. doi:10.1016/j.arr.2011.01.001
    • (2011) Ageing Res Rev , vol.10 , Issue.2 , pp. 264-273
    • Correia, S.C.1    Santos, R.X.2    Perry, G.3    Zhu, X.4    Moreira, P.I.5    Smith, M.A.6
  • 230
    • 78651274000 scopus 로고    scopus 로고
    • Insulin resistance and Alzheimer-like reductions in regional cerebral glucose metabolism for cognitively normal adults with prediabetes or early type 2 diabetes
    • Baker LD, Cross DJ, Minoshima S, Belongia D, Watson GS, Craft S. Insulin resistance and Alzheimer-like reductions in regional cerebral glucose metabolism for cognitively normal adults with prediabetes or early type 2 diabetes. Arch Neurol (2011) 68(1):51-7. doi:10.1001/archneurol.2010.225
    • (2011) Arch Neurol , vol.68 , Issue.1 , pp. 51-57
    • Baker, L.D.1    Cross, D.J.2    Minoshima, S.3    Belongia, D.4    Watson, G.S.5    Craft, S.6
  • 231
  • 232
    • 9844229370 scopus 로고    scopus 로고
    • Association between features of the insulin resistance syndrome and Alzheimer's disease independently of apolipoprotein E4 phenotype: cross sectional population based study
    • Kuusisto J, Koivisto K, Mykkanen L, Helkala EL, Vanhanen M, Hanninen T, et al. Association between features of the insulin resistance syndrome and Alzheimer's disease independently of apolipoprotein E4 phenotype: cross sectional population based study. BMJ (1997) 315(7115):1045-9. doi:10.1136/bmj.315.7115.1045
    • (1997) BMJ , vol.315 , Issue.7115 , pp. 1045-1049
    • Kuusisto, J.1    Koivisto, K.2    Mykkanen, L.3    Helkala, E.L.4    Vanhanen, M.5    Hanninen, T.6
  • 233
    • 77956364857 scopus 로고    scopus 로고
    • Insulin resistance is associated with the pathology of Alzheimer disease: the Hisayama study
    • Matsuzaki T, Sasaki K, Tanizaki Y, Hata J, Fujimi K, Matsui Y, et al. Insulin resistance is associated with the pathology of Alzheimer disease: the Hisayama study. Neurology (2010) 75(9):764-70. doi:10.1212/WNL.0b013e3181eee25f
    • (2010) Neurology , vol.75 , Issue.9 , pp. 764-770
    • Matsuzaki, T.1    Sasaki, K.2    Tanizaki, Y.3    Hata, J.4    Fujimi, K.5    Matsui, Y.6
  • 234
    • 84885980196 scopus 로고    scopus 로고
    • History of medically treated diabetes and risk of Alzheimer disease in a nationwide case-control study
    • Tolppanen AM, Lavikainen P, Solomon A, Kivipelto M, Uusitupa M, Soininen H, et al. History of medically treated diabetes and risk of Alzheimer disease in a nationwide case-control study. Diabetes Care (2013) 36(7):2015-9. doi:10.2337/dc12-1287
    • (2013) Diabetes Care , vol.36 , Issue.7 , pp. 2015-2019
    • Tolppanen, A.M.1    Lavikainen, P.2    Solomon, A.3    Kivipelto, M.4    Uusitupa, M.5    Soininen, H.6
  • 235
    • 71949112504 scopus 로고    scopus 로고
    • Increased tau phosphorylation and cleavage in mouse models of type 1 and type 2 diabetes
    • Kim B, Backus C, Oh S, Hayes JM, Feldman EL. Increased tau phosphorylation and cleavage in mouse models of type 1 and type 2 diabetes. Endocrinology (2009) 150(12):5294-301. doi:10.1210/en.2009-0695
    • (2009) Endocrinology , vol.150 , Issue.12 , pp. 5294-5301
    • Kim, B.1    Backus, C.2    Oh, S.3    Hayes, J.M.4    Feldman, E.L.5
  • 236
    • 4644286910 scopus 로고    scopus 로고
    • Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer's disease
    • Ho L, Qin W, Pompl PN, Xiang Z, Wang J, Zhao Z, et al. Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer's disease. FASEB J (2004) 18(7):902-4. doi:10.1096/fj.03-0978fje
    • (2004) FASEB J , vol.18 , Issue.7 , pp. 902-904
    • Ho, L.1    Qin, W.2    Pompl, P.N.3    Xiang, Z.4    Wang, J.5    Zhao, Z.6
  • 237
    • 84873035509 scopus 로고    scopus 로고
    • Deregulation of protein phosphatase 2A and hyperphosphorylation of tau protein following onset of diabetes in NOD mice
    • Papon MA, El Khoury NB, Marcouiller F, Julien C, Morin F, Bretteville A, et al. Deregulation of protein phosphatase 2A and hyperphosphorylation of tau protein following onset of diabetes in NOD mice. Diabetes (2013) 62(2):609-17. doi:10.2337/db12-0187
    • (2013) Diabetes , vol.62 , Issue.2 , pp. 609-617
    • Papon, M.A.1    El Khoury, N.B.2    Marcouiller, F.3    Julien, C.4    Morin, F.5    Bretteville, A.6
  • 238
    • 37249040432 scopus 로고    scopus 로고
    • Insulin dysfunction induces in vivo tau hyperphosphorylation through distinct mechanisms
    • Planel E, Tatebayashi Y, Miyasaka T, Liu L, Wang L, Herman M, et al. Insulin dysfunction induces in vivo tau hyperphosphorylation through distinct mechanisms. J Neurosci (2007) 27(50):13635-48. doi:10.1523/JNEUROSCI.3949-07.2007
    • (2007) J Neurosci , vol.27 , Issue.50 , pp. 13635-13648
    • Planel, E.1    Tatebayashi, Y.2    Miyasaka, T.3    Liu, L.4    Wang, L.5    Herman, M.6
  • 239
    • 84871831927 scopus 로고    scopus 로고
    • Pathological interaction between diabetes mellitus and Alzheimer's disease
    • Takeda S. Pathological interaction between diabetes mellitus and Alzheimer's disease. Nihon Shinkei Seishin Yakurigaku Zasshi (2012) 32(5-6):239-44.
    • (2012) Nihon Shinkei Seishin Yakurigaku Zasshi , vol.32 , Issue.5-6 , pp. 239-244
    • Takeda, S.1
  • 240
    • 84911895310 scopus 로고    scopus 로고
    • Insulin reverses the high-fat diet-induced increase in brain abeta and improves memory in an animal model of Alzheimer disease
    • Vandal M, White PJ, Tremblay C, St-Amour I, Chevrier G, Emond V, et al. Insulin reverses the high-fat diet-induced increase in brain abeta and improves memory in an animal model of Alzheimer disease. Diabetes (2014). doi:10.2337/db14-0375
    • (2014) Diabetes
    • Vandal, M.1    White, P.J.2    Tremblay, C.3    St-Amour, I.4    Chevrier, G.5    Emond, V.6
  • 241
    • 0032764016 scopus 로고    scopus 로고
    • Diabetes mellitus and the risk of dementia: the Rotterdam study
    • Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A, Breteler MM. Diabetes mellitus and the risk of dementia: the Rotterdam study. Neurology (1999) 53(9):1937-42. doi:10.1212/WNL.53.9.1937
    • (1999) Neurology , vol.53 , Issue.9 , pp. 1937-1942
    • Ott, A.1    Stolk, R.P.2    van Harskamp, F.3    Pols, H.A.4    Hofman, A.5    Breteler, M.M.6
  • 242
    • 28444491100 scopus 로고    scopus 로고
    • Cognitive decline and dementia in diabetes - systematic overview of prospective observational studies
    • Cukierman T, Gerstein HC, Williamson JD. Cognitive decline and dementia in diabetes - systematic overview of prospective observational studies. Diabetologia (2005) 48(12):2460-9. doi:10.1007/s00125-005-0023-4
    • (2005) Diabetologia , vol.48 , Issue.12 , pp. 2460-2469
    • Cukierman, T.1    Gerstein, H.C.2    Williamson, J.D.3
  • 243
    • 0036160510 scopus 로고    scopus 로고
    • Impaired intellectual development in children with type I diabetes: association with HbA(1c), age at diagnosis and sex
    • Schoenle EJ, Schoenle D, Molinari L, Largo RH. Impaired intellectual development in children with type I diabetes: association with HbA(1c), age at diagnosis and sex. Diabetologia (2002) 45(1):108-14. doi:10.1007/s125-002-8250-6
    • (2002) Diabetologia , vol.45 , Issue.1 , pp. 108-114
    • Schoenle, E.J.1    Schoenle, D.2    Molinari, L.3    Largo, R.H.4
  • 244
    • 34147130778 scopus 로고    scopus 로고
    • School performance in children with type 1 diabetes - a population-based register study
    • Dahlquist G, Kallen B. School performance in children with type 1 diabetes - a population-based register study. Diabetologia (2007) 50(5):957-64. doi:10.1007/s00125-007-0615-2
    • (2007) Diabetologia , vol.50 , Issue.5 , pp. 957-964
    • Dahlquist, G.1    Kallen, B.2
  • 245
    • 33644764957 scopus 로고    scopus 로고
    • Effects of type 1 diabetes on gray matter density as measured by voxel-based morphometry
    • Musen G, Lyoo IK, Sparks CR, Weinger K, Hwang J, Ryan CM, et al. Effects of type 1 diabetes on gray matter density as measured by voxel-based morphometry. Diabetes (2006) 55(2):326-33. doi:10.2337/diabetes.55.02.06.db05-0520
    • (2006) Diabetes , vol.55 , Issue.2 , pp. 326-333
    • Musen, G.1    Lyoo, I.K.2    Sparks, C.R.3    Weinger, K.4    Hwang, J.5    Ryan, C.M.6
  • 246
    • 15244351255 scopus 로고    scopus 로고
    • Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer's disease - is this type 3 diabetes?
    • Steen E, Terry BM, Rivera EJ, Cannon JL, Neely TR, Tavares R, et al. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer's disease - is this type 3 diabetes? J Alzheimers Dis (2005) 7(1):63-80.
    • (2005) J Alzheimers Dis , vol.7 , Issue.1 , pp. 63-80
    • Steen, E.1    Terry, B.M.2    Rivera, E.J.3    Cannon, J.L.4    Neely, T.R.5    Tavares, R.6
  • 247
    • 84889238217 scopus 로고    scopus 로고
    • Insulin and insulin-like growth factor prevent brain atrophy and cognitive impairment in diabetic rats
    • Serbedzija P, Ishii DN. Insulin and insulin-like growth factor prevent brain atrophy and cognitive impairment in diabetic rats. Indian J Endocrinol Metab (2012) 16(Suppl 3):S601-10. doi:10.4103/2230-8210.105578
    • (2012) Indian J Endocrinol Metab , vol.16 , pp. S601-S610
    • Serbedzija, P.1    Ishii, D.N.2
  • 248
    • 0036973393 scopus 로고    scopus 로고
    • Diabetes mellitus and the risk of dementia, Alzheimer's disease and vascular cognitive impairment in the Canadian study of health and aging
    • MacKnight C, Rockwood K, Awalt E, McDowell I. Diabetes mellitus and the risk of dementia, Alzheimer's disease and vascular cognitive impairment in the Canadian study of health and aging. Dement Geriatr Cogn Disord (2002) 14(2):77-83. doi:10.1159/000064928
    • (2002) Dement Geriatr Cogn Disord , vol.14 , Issue.2 , pp. 77-83
    • MacKnight, C.1    Rockwood, K.2    Awalt, E.3    McDowell, I.4
  • 249
    • 0036759508 scopus 로고    scopus 로고
    • Diabetes mellitus is a risk factor for vascular dementia, but not for Alzheimer's disease: a population-based study of the oldest old
    • Hassing LB, Johansson B, Nilsson SE, Berg S, Pedersen NL, Gatz M, et al. Diabetes mellitus is a risk factor for vascular dementia, but not for Alzheimer's disease: a population-based study of the oldest old. Int Psychogeriatr (2002) 14(3):239-48. doi:10.1017/S104161020200844X
    • (2002) Int Psychogeriatr , vol.14 , Issue.3 , pp. 239-248
    • Hassing, L.B.1    Johansson, B.2    Nilsson, S.E.3    Berg, S.4    Pedersen, N.L.5    Gatz, M.6
  • 250
    • 33746238205 scopus 로고    scopus 로고
    • Molecular indices of oxidative stress and mitochondrial dysfunction occur early and often progress with severity of Alzheimer's disease
    • de la Monte SM, Wands JR. Molecular indices of oxidative stress and mitochondrial dysfunction occur early and often progress with severity of Alzheimer's disease. J Alzheimers Dis (2006) 9(2):167-81.
    • (2006) J Alzheimers Dis , vol.9 , Issue.2 , pp. 167-181
    • de la Monte, S.M.1    Wands, J.R.2
  • 251
    • 17844364259 scopus 로고    scopus 로고
    • Brain glucose metabolism in the early and specific diagnosis of Alzheimer's disease. FDG-PET studies in MCI and AD
    • Mosconi L. Brain glucose metabolism in the early and specific diagnosis of Alzheimer's disease. FDG-PET studies in MCI and AD. Eur J Nucl Med Mol Imaging (2005) 32(4):486-510. doi:10.1007/s00259-005-1762-7
    • (2005) Eur J Nucl Med Mol Imaging , vol.32 , Issue.4 , pp. 486-510
    • Mosconi, L.1
  • 252
    • 0030874251 scopus 로고    scopus 로고
    • Metabolic reduction in the posterior cingulate cortex in very early Alzheimer's disease
    • Minoshima S, Giordani B, Berent S, Frey KA, Foster NL, Kuhl DE. Metabolic reduction in the posterior cingulate cortex in very early Alzheimer's disease. Ann Neurol (1997) 42(1):85-94. doi:10.1002/ana.410420114
    • (1997) Ann Neurol , vol.42 , Issue.1 , pp. 85-94
    • Minoshima, S.1    Giordani, B.2    Berent, S.3    Frey, K.A.4    Foster, N.L.5    Kuhl, D.E.6
  • 253
    • 0028241915 scopus 로고
    • Decreased concentrations of GLUT1 and GLUT3 glucose transporters in the brains of patients with Alzheimer's disease
    • Simpson IA, Chundu KR, Davies-Hill T, Honer WG, Davies P. Decreased concentrations of GLUT1 and GLUT3 glucose transporters in the brains of patients with Alzheimer's disease. Ann Neurol (1994) 35(5):546-51. doi:10.1002/ana.410350507
    • (1994) Ann Neurol , vol.35 , Issue.5 , pp. 546-551
    • Simpson, I.A.1    Chundu, K.R.2    Davies-Hill, T.3    Honer, W.G.4    Davies, P.5
  • 254
    • 67650072530 scopus 로고    scopus 로고
    • Reduced O-GlcNAcylation links lower brain glucose metabolism and tau pathology in Alzheimer's disease
    • Liu F, Shi J, Tanimukai H, Gu J, Grundke-Iqbal I, Iqbal K, et al. Reduced O-GlcNAcylation links lower brain glucose metabolism and tau pathology in Alzheimer's disease. Brain (2009) 132(Pt 7):1820-32. doi:10.1093/brain/awp099
    • (2009) Brain , vol.132 , pp. 1820-1832
    • Liu, F.1    Shi, J.2    Tanimukai, H.3    Gu, J.4    Grundke-Iqbal, I.5    Iqbal, K.6
  • 255
    • 0030009819 scopus 로고    scopus 로고
    • Brain thiamine, its phosphate esters, and its metabolizing enzymes in Alzheimer's disease
    • Mastrogiacoma F, Bettendorff L, Grisar T, Kish SJ. Brain thiamine, its phosphate esters, and its metabolizing enzymes in Alzheimer's disease. Ann Neurol (1996) 39(5):585-91. doi:10.1002/ana.410390507
    • (1996) Ann Neurol , vol.39 , Issue.5 , pp. 585-591
    • Mastrogiacoma, F.1    Bettendorff, L.2    Grisar, T.3    Kish, S.J.4
  • 256
    • 38849201796 scopus 로고    scopus 로고
    • Cholesterol in islet dysfunction and type 2 diabetes
    • Brunham LR, Kruit JK, Verchere CB, Hayden MR. Cholesterol in islet dysfunction and type 2 diabetes. J Clin Invest (2008) 118(2):403-8. doi:10.1172/JCI33296
    • (2008) J Clin Invest , vol.118 , Issue.2 , pp. 403-408
    • Brunham, L.R.1    Kruit, J.K.2    Verchere, C.B.3    Hayden, M.R.4
  • 257
    • 2442456925 scopus 로고    scopus 로고
    • Identification of insulin-responsive regions in the HMG-CoA reductase promoter
    • Osborne AR, Pollock VV, Lagor WR, Ness GC. Identification of insulin-responsive regions in the HMG-CoA reductase promoter. Biochem Biophys Res Commun (2004) 318(4):814-8. doi:10.1016/j.bbrc.2004.04.105
    • (2004) Biochem Biophys Res Commun , vol.318 , Issue.4 , pp. 814-818
    • Osborne, A.R.1    Pollock, V.V.2    Lagor, W.R.3    Ness, G.C.4
  • 258
    • 27844458139 scopus 로고    scopus 로고
    • Insulin and cholesterol pathways in neuronal function, memory and neurodegeneration
    • Nelson TJ, Alkon DL. Insulin and cholesterol pathways in neuronal function, memory and neurodegeneration. Biochem Soc Trans (2005) 33(Pt 5):1033-6. doi:10.1042/BST20051033
    • (2005) Biochem Soc Trans , vol.33 , pp. 1033-1036
    • Nelson, T.J.1    Alkon, D.L.2
  • 259
    • 14844304305 scopus 로고    scopus 로고
    • Oxidation of cholesterol by amyloid precursor protein and beta-amyloid peptide
    • Nelson TJ, Alkon DL. Oxidation of cholesterol by amyloid precursor protein and beta-amyloid peptide. J Biol Chem (2005) 280(8):7377-87. doi:10.1074/jbc.M409071200
    • (2005) J Biol Chem , vol.280 , Issue.8 , pp. 7377-7387
    • Nelson, T.J.1    Alkon, D.L.2
  • 260
    • 3242739968 scopus 로고    scopus 로고
    • O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer's disease
    • Liu F, Iqbal K, Grundke-Iqbal I, Hart GW, Gong CX. O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer's disease. Proc Natl Acad Sci U S A (2004) 101(29):10804-9. doi:10.1073/pnas.0400348101
    • (2004) Proc Natl Acad Sci U S A , vol.101 , Issue.29 , pp. 10804-10809
    • Liu, F.1    Iqbal, K.2    Grundke-Iqbal, I.3    Hart, G.W.4    Gong, C.X.5
  • 261
    • 35348866703 scopus 로고    scopus 로고
    • O-GlcNAc modification in diabetes and Alzheimer's disease
    • Dias WB, Hart GW. O-GlcNAc modification in diabetes and Alzheimer's disease. Mol Biosyst (2007) 3(11):766-72. doi:10.1039/b704905f
    • (2007) Mol Biosyst , vol.3 , Issue.11 , pp. 766-772
    • Dias, W.B.1    Hart, G.W.2
  • 262
    • 70349216418 scopus 로고    scopus 로고
    • Brain glucose transporters, O-GlcNAcylation and phosphorylation of tau in diabetes and Alzheimer's disease
    • Liu Y, Liu F, Grundke-Iqbal I, Iqbal K, Gong CX. Brain glucose transporters, O-GlcNAcylation and phosphorylation of tau in diabetes and Alzheimer's disease. J Neurochem (2009) 111(1):242-9. doi:10.1111/j.1471-4159.2009.06320.x
    • (2009) J Neurochem , vol.111 , Issue.1 , pp. 242-249
    • Liu, Y.1    Liu, F.2    Grundke-Iqbal, I.3    Iqbal, K.4    Gong, C.X.5
  • 263
    • 34247117730 scopus 로고    scopus 로고
    • Amyloid-beta aggregation
    • Finder VH, Glockshuber R. Amyloid-beta aggregation. Neurodegener Dis (2007) 4(1):13-27. doi:10.1159/000100355
    • (2007) Neurodegener Dis , vol.4 , Issue.1 , pp. 13-27
    • Finder, V.H.1    Glockshuber, R.2
  • 264
    • 34347377319 scopus 로고    scopus 로고
    • Alzheimer-like changes in rat models of spontaneous diabetes
    • Li ZG, Zhang W, Sima AA. Alzheimer-like changes in rat models of spontaneous diabetes. Diabetes (2007) 56(7):1817-24. doi:10.2337/db07-0171
    • (2007) Diabetes , vol.56 , Issue.7 , pp. 1817-1824
    • Li, Z.G.1    Zhang, W.2    Sima, A.A.3
  • 265
    • 10944228564 scopus 로고    scopus 로고
    • Insulin-degrading enzyme as a downstream target of insulin receptor signaling cascade: implications for Alzheimer's disease intervention
    • Zhao L, Teter B, Morihara T, Lim GP, Ambegaokar SS, Ubeda OJ, et al. Insulin-degrading enzyme as a downstream target of insulin receptor signaling cascade: implications for Alzheimer's disease intervention. J Neurosci (2004) 24(49):11120-6. doi:10.1523/JNEUROSCI.2860-04.2004
    • (2004) J Neurosci , vol.24 , Issue.49 , pp. 11120-11126
    • Zhao, L.1    Teter, B.2    Morihara, T.3    Lim, G.P.4    Ambegaokar, S.S.5    Ubeda, O.J.6
  • 266
    • 0037390039 scopus 로고    scopus 로고
    • Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, and the beta-amyloid precursor protein intracellular domain in vivo
    • Farris W, Mansourian S, Chang Y, Lindsley L, Eckman EA, Frosch MP, et al. Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, and the beta-amyloid precursor protein intracellular domain in vivo. Proc Natl Acad Sci U S A (2003) 100(7):4162-7. doi:10.1073/pnas.0230450100
    • (2003) Proc Natl Acad Sci U S A , vol.100 , Issue.7 , pp. 4162-4167
    • Farris, W.1    Mansourian, S.2    Chang, Y.3    Lindsley, L.4    Eckman, E.A.5    Frosch, M.P.6
  • 267
    • 33747401659 scopus 로고    scopus 로고
    • Tau phosphorylation and proteolysis: insights and perspectives
    • Johnson GV. Tau phosphorylation and proteolysis: insights and perspectives. J Alzheimers Dis (2006) 9(3 Suppl):243-50.
    • (2006) J Alzheimers Dis , vol.9 , Issue.3 , pp. 243-250
    • Johnson, G.V.1
  • 268
    • 3242749074 scopus 로고    scopus 로고
    • Caspase-cleavage of tau is an early event in Alzheimer disease tangle pathology
    • Rissman RA, Poon WW, Blurton-Jones M, Oddo S, Torp R, Vitek MP, et al. Caspase-cleavage of tau is an early event in Alzheimer disease tangle pathology. J Clin Invest (2004) 114(1):121-30. doi:10.1172/JCI20640
    • (2004) J Clin Invest , vol.114 , Issue.1 , pp. 121-130
    • Rissman, R.A.1    Poon, W.W.2    Blurton-Jones, M.3    Oddo, S.4    Torp, R.5    Vitek, M.P.6
  • 269
    • 71449097274 scopus 로고    scopus 로고
    • Role of infection in the pathogenesis of Alzheimer's disease: implications for treatment
    • Holmes C, Cotterell D. Role of infection in the pathogenesis of Alzheimer's disease: implications for treatment. CNS Drugs (2009) 23(12):993-1002. doi:10.2165/11310910-000000000-00000
    • (2009) CNS Drugs , vol.23 , Issue.12 , pp. 993-1002
    • Holmes, C.1    Cotterell, D.2
  • 272
    • 33644878338 scopus 로고    scopus 로고
    • The novel beta-secretase inhibitor KMI-429 reduces amyloid beta peptide production in amyloid precursor protein transgenic and wild-type mice
    • Asai M, Hattori C, Iwata N, Saido TC, Sasagawa N, Szabo B, et al. The novel beta-secretase inhibitor KMI-429 reduces amyloid beta peptide production in amyloid precursor protein transgenic and wild-type mice. J Neurochem (2006) 96(2):533-40. doi:10.1111/j.1471-4159.2005.03576.x
    • (2006) J Neurochem , vol.96 , Issue.2 , pp. 533-540
    • Asai, M.1    Hattori, C.2    Iwata, N.3    Saido, T.C.4    Sasagawa, N.5    Szabo, B.6
  • 273
    • 21044449225 scopus 로고    scopus 로고
    • Inhibition of glycogen synthase kinase-3 by lithium correlates with reduced tauopathy and degeneration in vivo
    • Noble W, Planel E, Zehr C, Olm V, Meyerson J, Suleman F, et al. Inhibition of glycogen synthase kinase-3 by lithium correlates with reduced tauopathy and degeneration in vivo. Proc Natl Acad Sci U S A (2005) 102(19):6990-5. doi:10.1073/pnas.0500466102
    • (2005) Proc Natl Acad Sci U S A , vol.102 , Issue.19 , pp. 6990-6995
    • Noble, W.1    Planel, E.2    Zehr, C.3    Olm, V.4    Meyerson, J.5    Suleman, F.6
  • 275
  • 276
    • 45749135146 scopus 로고    scopus 로고
    • Optimal drug cocktail design: methods for targeting molecular ensembles and insights from theoretical model systems
    • Radhakrishnan ML, Tidor B. Optimal drug cocktail design: methods for targeting molecular ensembles and insights from theoretical model systems. J Chem Inf Model (2008) 48(5):1055-73. doi:10.1021/ci700452r
    • (2008) J Chem Inf Model , vol.48 , Issue.5 , pp. 1055-1073
    • Radhakrishnan, M.L.1    Tidor, B.2
  • 277
    • 78650746798 scopus 로고    scopus 로고
    • Biguanide metformin acts on tau phosphorylation via mTOR/protein phosphatase 2A (PP2A) signaling
    • Kickstein E, Krauss S, Thornhill P, Rutschow D, Zeller R, Sharkey J, et al. Biguanide metformin acts on tau phosphorylation via mTOR/protein phosphatase 2A (PP2A) signaling. Proc Natl Acad Sci U S A (2010) 107(50):21830-5. doi:10.1073/pnas.0912793107
    • (2010) Proc Natl Acad Sci U S A , vol.107 , Issue.50 , pp. 21830-21835
    • Kickstein, E.1    Krauss, S.2    Thornhill, P.3    Rutschow, D.4    Zeller, R.5    Sharkey, J.6
  • 278
    • 79956277464 scopus 로고    scopus 로고
    • Incidence of dementia is increased in type 2 diabetes and reduced by the use of sulfonylureas and metformin
    • Hsu CC, Wahlqvist ML, Lee MS, Tsai HN. Incidence of dementia is increased in type 2 diabetes and reduced by the use of sulfonylureas and metformin. J Alzheimers Dis (2011) 24(3):485-93. doi:10.3233/JAD-2011-101524
    • (2011) J Alzheimers Dis , vol.24 , Issue.3 , pp. 485-493
    • Hsu, C.C.1    Wahlqvist, M.L.2    Lee, M.S.3    Tsai, H.N.4
  • 279
    • 13744249406 scopus 로고    scopus 로고
    • PPARgamma-mediated insulin sensitization: the importance of fat versus muscle
    • Kintscher U, Law RE. PPARgamma-mediated insulin sensitization: the importance of fat versus muscle. Am J Physiol Endocrinol Metab (2005) 288(2):E287-91. doi:10.1152/ajpendo.00440.2004
    • (2005) Am J Physiol Endocrinol Metab , vol.288 , Issue.2 , pp. E287-E291
    • Kintscher, U.1    Law, R.E.2
  • 280
    • 0034651101 scopus 로고    scopus 로고
    • Inflammatory mechanisms in Alzheimer's disease: inhibition of beta-amyloid-stimulated proinflammatory responses and neurotoxicity by PPARgamma agonists
    • Combs CK, Johnson DE, Karlo JC, Cannady SB, Landreth GE. Inflammatory mechanisms in Alzheimer's disease: inhibition of beta-amyloid-stimulated proinflammatory responses and neurotoxicity by PPARgamma agonists. J Neurosci (2000) 20(2):558-67.
    • (2000) J Neurosci , vol.20 , Issue.2 , pp. 558-567
    • Combs, C.K.1    Johnson, D.E.2    Karlo, J.C.3    Cannady, S.B.4    Landreth, G.E.5
  • 281
    • 0034965596 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptors in inflammation control
    • Delerive P, Fruchart JC, Staels B. Peroxisome proliferator-activated receptors in inflammation control. J Endocrinol (2001) 169(3):453-9. doi:10.1677/joe.0.1690453
    • (2001) J Endocrinol , vol.169 , Issue.3 , pp. 453-459
    • Delerive, P.1    Fruchart, J.C.2    Staels, B.3
  • 282
    • 33644592598 scopus 로고    scopus 로고
    • Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study
    • Watson GS, Cholerton BA, Reger MA, Baker LD, Plymate SR, Asthana S, et al. Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study. Am J Geriatr Psychiatry (2005) 13(11):950-8. doi:10.1176/appi.ajgp.13.11.950
    • (2005) Am J Geriatr Psychiatry , vol.13 , Issue.11 , pp. 950-958
    • Watson, G.S.1    Cholerton, B.A.2    Reger, M.A.3    Baker, L.D.4    Plymate, S.R.5    Asthana, S.6
  • 283
    • 79955749452 scopus 로고    scopus 로고
    • The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer's disease
    • McClean PL, Parthsarathy V, Faivre E, Holscher C. The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer's disease. J Neurosci (2011) 31(17):6587-94. doi:10.1523/JNEUROSCI.0529-11.2011
    • (2011) J Neurosci , vol.31 , Issue.17 , pp. 6587-6594
    • McClean, P.L.1    Parthsarathy, V.2    Faivre, E.3    Holscher, C.4
  • 284
    • 78649385516 scopus 로고    scopus 로고
    • The role of GLP-1 in neuronal activity and neurodegeneration
    • Holscher C. The role of GLP-1 in neuronal activity and neurodegeneration. Vitam Horm (2010) 84:331-54. doi:10.1016/B978-0-12-381517-0.00013-8
    • (2010) Vitam Horm , vol.84 , pp. 331-354
    • Holscher, C.1
  • 285
    • 77953880083 scopus 로고    scopus 로고
    • Incretin analogues that have been developed to treat type 2 diabetes hold promise as a novel treatment strategy for Alzheimer's disease
    • Holscher C. Incretin analogues that have been developed to treat type 2 diabetes hold promise as a novel treatment strategy for Alzheimer's disease. Recent Pat CNS Drug Discov (2010) 5(2):109-17. doi:10.2174/157488910791213130
    • (2010) Recent Pat CNS Drug Discov , vol.5 , Issue.2 , pp. 109-117
    • Holscher, C.1
  • 286
    • 77950352189 scopus 로고    scopus 로고
    • GLP-1 receptor stimulation reduces amyloid-beta peptide accumulation and cytotoxicity in cellular and animal models of Alzheimer's disease
    • Li Y, Duffy KB, Ottinger MA, Ray B, Bailey JA, Holloway HW, et al. GLP-1 receptor stimulation reduces amyloid-beta peptide accumulation and cytotoxicity in cellular and animal models of Alzheimer's disease. J Alzheimers Dis (2010) 19(4):1205-19. doi:10.3233/JAD-2010-1314
    • (2010) J Alzheimers Dis , vol.19 , Issue.4 , pp. 1205-1219
    • Li, Y.1    Duffy, K.B.2    Ottinger, M.A.3    Ray, B.4    Bailey, J.A.5    Holloway, H.W.6
  • 287
    • 79751479118 scopus 로고    scopus 로고
    • Novel GLP-1 mimetics developed to treat type 2 diabetes promote progenitor cell proliferation in the brain
    • Hamilton A, Patterson S, Porter D, Gault VA, Holscher C. Novel GLP-1 mimetics developed to treat type 2 diabetes promote progenitor cell proliferation in the brain. J Neurosci Res (2011) 89(4):481-9. doi:10.1002/jnr.22565
    • (2011) J Neurosci Res , vol.89 , Issue.4 , pp. 481-489
    • Hamilton, A.1    Patterson, S.2    Porter, D.3    Gault, V.A.4    Holscher, C.5
  • 288
    • 28444459842 scopus 로고    scopus 로고
    • Insulin resistance syndrome and Alzheimer's disease: age- and obesity-related effects on memory, amyloid, and inflammation
    • Craft S. Insulin resistance syndrome and Alzheimer's disease: age- and obesity-related effects on memory, amyloid, and inflammation. Neurobiol Aging (2005) 26(Suppl 1):65-9. doi:10.1016/j.neurobiolaging.2005.08.021
    • (2005) Neurobiol Aging , vol.26 , pp. 65-69
    • Craft, S.1
  • 289
    • 84855613853 scopus 로고    scopus 로고
    • Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial
    • Craft S, Baker LD, Montine TJ, Minoshima S, Watson GS, Claxton A, et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch Neurol (2012) 69(1):29-38. doi:10.1001/archneurol.2011.233
    • (2012) Arch Neurol , vol.69 , Issue.1 , pp. 29-38
    • Craft, S.1    Baker, L.D.2    Montine, T.J.3    Minoshima, S.4    Watson, G.S.5    Claxton, A.6
  • 290
    • 84857034930 scopus 로고    scopus 로고
    • Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism
    • Ott V, Benedict C, Schultes B, Born J, Hallschmid M. Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism. Diabetes Obes Metab (2012) 14(3):214-21. doi:10.1111/j.1463-1326.2011.01490.x
    • (2012) Diabetes Obes Metab , vol.14 , Issue.3 , pp. 214-221
    • Ott, V.1    Benedict, C.2    Schultes, B.3    Born, J.4    Hallschmid, M.5
  • 291
    • 79960034311 scopus 로고    scopus 로고
    • Development of the novel delivery system of GLP-1 administration for the treatment of diabetes mellitus
    • Nakazato M. Development of the novel delivery system of GLP-1 administration for the treatment of diabetes mellitus. Nihon Rinsho (2011) 69(5):918-22.
    • (2011) Nihon Rinsho , vol.69 , Issue.5 , pp. 918-922
    • Nakazato, M.1
  • 292
    • 84866122780 scopus 로고    scopus 로고
    • Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2
    • Copps KD, White MF. Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2. Diabetologia (2012) 55(10):2565-82. doi:10.1007/s00125-012-2644-8
    • (2012) Diabetologia , vol.55 , Issue.10 , pp. 2565-2582
    • Copps, K.D.1    White, M.F.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.