The Strategies for Treating “Alzheimer’s Disease”: Insulin Signaling May Be a Feasible Target
Abstract
:1. Introduction
2. Restricting the Overload of Aβ
3. Rescuing Insulin Signaling
4. Preventing Tau Pathology
5. Microglia Depletion and Repopulation
6. Conclusions and Perspectives
Author Contributions
Funding
Conflicts of Interest
References
- Busche, M.A.; Hyman, B.T. Synergy between amyloid-beta and tau in Alzheimer’s disease. Nat. Neurosci. 2020, 23, 1183–1193. [Google Scholar] [CrossRef] [PubMed]
- Jack, C.R.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef] [PubMed]
- Hardy, J.A.; Higgins, G.A. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef] [PubMed]
- Priller, C.; Bauer, T.; Mitteregger, G.; Krebs, B.; Kretzschmar, H.A.; Herms, J. Synapse formation and function is modulated by the amyloid precursor protein. J. Neurosci. 2006, 26, 7212–7221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duce, J.A.; Tsatsanis, A.; Cater, M.A.; James, S.A.; Robb, E.; Wikhe, K.; Leong, S.L.; Perez, K.; Johanssen, T.; Greenough, M.A.; et al. Iron-Export Ferroxidase Activity of beta-Amyloid Precursor Protein Is Inhibited by Zinc in Alzheimer’s Disease. Cell 2010, 142, 857–867. [Google Scholar] [CrossRef] [Green Version]
- Muller, U.C.; Deller, T.; Korte, M. Not just amyloid: Physiological functions of the amyloid precursor protein family. Nat. Rev. Neurosci. 2017, 18, 281–298. [Google Scholar] [CrossRef]
- Kim, M.; Suh, J.; Romano, D.; Truong, M.H.; Mullin, K.; Hooli, B.; Norton, D.; Tesco, G.; Elliott, K.; Wagner, S.L.; et al. Potential late-onset Alzheimer’s disease-associated mutations in the ADAM10 gene attenuate alpha-secretase activity. Hum. Mol. Genet. 2009, 18, 3987–3996. [Google Scholar] [CrossRef] [Green Version]
- Aguero, P.; Sainz, M.J.; Garcia-Ayllon, M.S.; Saez-Valero, J.; Tellez, R.; Guerrero-Lopez, R.; Perez-Perez, J.; Jimenez-Escrig, A.; Gomez-Tortosa, E. alpha-Secretase nonsense mutation (ADAM10 Tyr167*) in familial Alzheimer’s disease. Alzheimer’s Res. Ther. 2020, 12, 139. [Google Scholar] [CrossRef]
- Chang, Y.J.; Chen, Y.R. The coexistence of an equal amount of Alzheimer’s amyloid-beta 40 and 42 forms structurally stable and toxic oligomers through a distinct pathway. FEBS J. 2014, 281, 2674–2687. [Google Scholar] [CrossRef]
- Jang, H.; Connelly, L.; Arce, F.T.; Ramachandran, S.; Kagan, B.L.; Lal, R.; Nussinov, R. Mechanisms for the Insertion of Toxic, Fibril-like beta-Amyloid Oligomers into the Membrane. J. Chem. Theory Comput. 2013, 9, 822–833. [Google Scholar] [CrossRef]
- Morkuniene, R.; Cizas, P.; Jankeviciute, S.; Petrolis, R.; Arandarcikaite, O.; Krisciukaitis, A.; Borutaite, V. Small A beta(1-42) Oligomer-Induced Membrane Depolarization of Neuronal and Microglial Cells: Role of N-Methyl-D-Aspartate Receptors. J. Neurosci. Res. 2015, 93, 475–486. [Google Scholar] [CrossRef]
- Li, S.M.; Jin, M.; Koeglsperger, T.; Shepardson, N.E.; Shankar, G.M.; Selkoe, D.J. Soluble A beta Oligomers Inhibit Long-Term Potentiation through a Mechanism Involving Excessive Activation of Extrasynaptic NR2B-Containing NMDA Receptors. J. Neurosci. 2011, 31, 6627–6638. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.L.; Su, B.; Siedlak, S.L.; Moreira, P.I.; Fujioka, H.; Wang, Y.; Casadesus, G.; Zhu, X.W. Amyloid-beta overproduction causes abnormal mitochondrial dynamics via differential modulation of mitochondrial fission/fusion proteins. Proc. Natl. Acad. Sci. USA 2008, 105, 19318–19323. [Google Scholar] [CrossRef] [Green Version]
- Du, H.; Guo, L.; Yan, S.Q.; Sosunov, A.A.; McKhann, G.M.; Yan, S.S. Early deficits in synaptic mitochondria in an Alzheimer’s disease mouse model. Proc. Natl. Acad. Sci. USA 2010, 107, 18670–18675. [Google Scholar] [CrossRef] [Green Version]
- He, Y.; Zheng, M.M.; Ma, Y.; Han, X.J.; Ma, X.Q.; Qu, C.Q.; Du, Y.F. Soluble oligomers and fibrillar species of amyloid beta-peptide differentially affect cognitive functions and hippocampal inflammatory response. Biochem. Biophys. Res. Commun. 2012, 429, 125–130. [Google Scholar] [CrossRef]
- Zhang, Y.L.; Wang, J.; Zhang, Z.N.; Su, Q.; Guo, J.H. The relationship between amyloid-beta and brain capillary endothelial cells in Alzheimer’s disease. Neural Regen. Res. 2022, 17, 2355–2363. [Google Scholar] [CrossRef]
- Endres, K.; Fahrenholz, F.; Lotz, J.; Hiemke, C.; Teipel, S.; Lieb, K.; Tuscher, O.; Fellgiebel, A. Increased CSF APPs-alpha levels in patients with Alzheimer disease treated with acitretin. Neurology 2014, 83, 1930–1935. [Google Scholar] [CrossRef]
- Musardo, S.; Therin, S.; Pelucchi, S.; D’Andrea, L.; Stringhi, R.; Ribeiro, A.; Manca, A.; Balducci, C.; Pagano, J.; Sala, C.; et al. The development of ADAM10 endocytosis inhibitors for the treatment of Alzheimer’s disease. Mol. Ther. J. Am. Soc. Gene Ther. 2022, 30, 2474–2490. [Google Scholar] [CrossRef]
- Egan, M.F.; Kost, J.; Tariot, P.N.; Aisen, P.S.; Cummings, J.L.; Vellas, B.; Sur, C.; Mukai, Y.; Voss, T.; Furtek, C.; et al. Randomized Trial of Verubecestat for Mild-to-Moderate Alzheimer’s Disease. N. Engl. J. Med. 2018, 378, 1691–1703. [Google Scholar] [CrossRef]
- Wessels, A.M.; Tariot, P.N.; Zimmer, J.A.; Selzler, K.J.; Bragg, S.M.; Andersen, S.W.; Landry, J.; Krull, J.H.; Downing, A.M.; Willis, B.A.; et al. Efficacy and Safety of Lanabecestat for Treatment of Early and Mild Alzheimer Disease: The AMARANTH and DAYBREAK-ALZ Randomized Clinical Trials. JAMA Neurol. 2020, 77, 199–209. [Google Scholar] [CrossRef] [Green Version]
- Hur, J.Y. Gamma-Secretase in Alzheimer’s disease. Exp. Mol. Med. 2022, 54, 433–446. [Google Scholar] [CrossRef] [PubMed]
- Endres, K.; Deller, T. Regulation of Alpha-Secretase ADAM10 In vitro and In vivo: Genetic, Epigenetic, and Protein-Based Mechanisms. Front. Mol. Neurosci. 2017, 10, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taylor, H.A.; Przemylska, L.; Clavane, E.M.; Meakin, P.J. BACE1: More than just a beta-secretase. Obes. Rev. 2022, 23, e13430. [Google Scholar] [CrossRef] [PubMed]
- Blanchard, J.W.; Bula, M.; Davila-Velderrain, J.; Akay, L.A.; Zhu, L.N.; Frank, A.; Victor, M.B.; Bonner, J.M.; Mathys, H.; Lin, Y.T.; et al. Reconstruction of the human blood-brain barrier in vitro reveals a pathogenic mechanism of APOE4 in pericytes. Nat. Med. 2020, 26, 952–963. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.M.; Cella, M.; Mallinson, K.; Ulrich, J.D.; Young, K.L.; Robinette, M.L.; Gilfillan, S.; Krishnan, G.M.; Sudhakar, S.; Zinselmeyer, B.H.; et al. TREM2 Lipid Sensing Sustains the Microglial Response in an Alzheimer’s Disease Model. Cell 2015, 160, 1061–1071. [Google Scholar] [CrossRef] [Green Version]
- Parikh, I.; Fardo, D.W.; Estus, S. Genetics of PICALM expression and Alzheimer’s disease. PLoS ONE 2014, 9, e91242. [Google Scholar] [CrossRef]
- Narayan, P.; Sienski, G.; Bonner, J.M.; Lin, Y.T.; Seo, J.; Baru, V.; Haque, A.; Milo, B.; Akay, L.A.; Graziosi, A.; et al. PICALM Rescues Endocytic Defects Caused by the Alzheimer’s Disease Risk Factor APOE4. Cell Rep. 2020, 33, 108224. [Google Scholar] [CrossRef]
- Miyagawa, T.; Ebinuma, I.; Morohashi, Y.; Hori, Y.; Chang, M.Y.; Hattori, H.; Maehara, T.; Yokoshima, S.; Fukuyama, T.; Tsuji, S.; et al. BIN1 regulates BACE1 intracellular trafficking and amyloid-beta production. Hum. Mol. Genet. 2016, 25, 2948–2958. [Google Scholar] [CrossRef] [Green Version]
- Schneider, L. A resurrection of aducanumab for Alzheimer’s disease. Lancet Neurol. 2020, 19, 111–112. [Google Scholar] [CrossRef] [Green Version]
- Behl, T.; Kaur, I.; Sehgal, A.; Singh, S.; Sharma, N.; Makeen, H.A.; Albratty, M.; Alhazmi, H.A.; Felemban, S.G.; Alsubayiel, A.M.; et al. “Aducanumab” making a comeback in Alzheimer’s disease: An old wine in a new bottle. Biomed. Pharmacother. 2022, 148, 112746. [Google Scholar] [CrossRef]
- Maitra, S.; Vincent, B. Cdk5-p25 as a key element linking amyloid and tau pathologies in Alzheimer’s disease: Mechanisms and possible therapeutic interventions. Life Sci. 2022, 308, 120986. [Google Scholar] [CrossRef]
- Atagi, Y.; Liu, C.C.; Painter, M.M.; Chen, X.F.; Verbeeck, C.; Zheng, H.H.; Li, X.; Rademakers, R.; Kang, S.S.; Xu, H.X.; et al. Apolipoprotein E Is a Ligand for Triggering Receptor Expressed on Myeloid Cells 2 (TREM2). J. Biol. Chem. 2015, 290, 26043–26050. [Google Scholar] [CrossRef] [Green Version]
- Yeh, F.L.; Wang, Y.Y.; Tom, I.; Gonzalez, L.C.; Sheng, M. TREM2 Binds to Apolipoproteins, Including APOE and CLU/APOJ, and Thereby Facilitates Uptake of Amyloid-Beta by Microglia. Neuron 2016, 91, 328–340. [Google Scholar] [CrossRef]
- Steen, E.; Terry, B.M.; Rivera, E.J.; Cannon, J.L.; Neely, T.R.; Tavares, R.; Xu, X.J.; Wands, J.R.; de la Monte, S.M. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer’s disease—Is this type 3 diabetes? J. Alzheimer’s Dis. 2005, 7, 63–80. [Google Scholar] [CrossRef] [Green Version]
- Marks, J.L.; Porte, D., Jr.; Stahl, W.L.; Baskin, D.G. Localization of insulin receptor mRNA in rat brain by in situ hybridization. Endocrinology 1990, 127, 3234–3236. [Google Scholar] [CrossRef]
- Adamo, M.; Raizada, M.K.; LeRoith, D. Insulin and insulin-like growth factor receptors in the nervous system. Mol. Neurobiol. 1989, 3, 71–100. [Google Scholar] [CrossRef]
- Zhang, W.D.; Liu, Q.Y.; Haqqani, A.S.; Leclerc, S.; Liu, Z.Y.; Fauteux, F.; Baumann, E.; Delaney, C.E.; Ly, D.; Star, A.T.; et al. Differential expression of receptors mediating receptor-mediated transcytosis (RMT) in brain microvessels, brain parenchyma and peripheral tissues of the mouse and the human. Fluids Barriers CNS 2020, 17, 47. [Google Scholar] [CrossRef]
- Leclerc, M.; Bourassa, P.; Tremblay, C.; Caron, V.; Sugere, C.; Emond, V.; Bennett, D.A.; Calon, F. Cerebrovascular insulin receptors are defective in Alzheimer’s disease. Brain 2022, awac309. [Google Scholar] [CrossRef]
- Gali, C.C.; Fanaee-Danesh, E.; Zandl-Lang, M.; Albrecher, N.M.; Tam-Amersdorfer, C.; Stracke, A.; Sachdev, V.; Reichmann, F.; Sun, Y.D.; Avdili, A.; et al. Amyloid-beta impairs insulin signaling by accelerating autophagy-lysosomal degradation of LRP-1 and IR-beta in blood-brain barrier endothelial cells in vitro and in 3XTg-AD mice. Mol. Cell. Neurosci. 2019, 99, 103390. [Google Scholar] [CrossRef]
- De la Monte, S.M.; Tong, M.; Daiello, L.A.; Ott, B.R. Early-Stage Alzheimer’s Disease Is Associated with Simultaneous Systemic and Central Nervous System Dysregulation of Insulin-Linked Metabolic Pathways. J. Alzheimer’s Dis. 2019, 68, 657–668. [Google Scholar] [CrossRef]
- Confettura, A.D.; Cuboni, E.; Ammar, M.R.; Jia, S.; Gomes, G.M.; Yuanxiang, P.; Raman, R.; Li, T.T.; Grochowska, K.M.; Ahrends, R.; et al. Neddylation-dependent protein degradation is a nexus between synaptic insulin resistance, neuroinflammation and Alzheimer’s disease. Transl. Neurodegener. 2022, 11, 2. [Google Scholar] [CrossRef] [PubMed]
- Ma, Q.L.; Yang, F.; Rosario, E.R.; Ubeda, O.J.; Beech, W.; Gant, D.J.; Chen, P.P.; Hudspeth, B.; Chen, C.; Zhao, Y.; et al. Beta-amyloid oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling: Suppression by omega-3 fatty acids and curcumin. J. Neurosci. 2009, 29, 9078–9089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duarte, A.I.; Moreira, P.I.; Oliveira, C.R. Insulin in central nervous system: More than just a peripheral hormone. J. Aging Res. 2012, 2012, 384017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Banks, W.A. The source of cerebral insulin. Eur. J. Pharmacol. 2004, 490, 5–12. [Google Scholar] [CrossRef] [PubMed]
- Banks, W.A.; Owen, J.B.; Erickson, M.A. Insulin in the brain: There and back again. Pharmacol. Ther. 2012, 136, 82–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grillo, C.A.; Piroli, G.G.; Hendry, R.M.; Reagan, L.P. Insulin-stimulated translocation of GLUT4 to the plasma membrane in rat hippocampus is PI3-kinase dependent. Brain Res. 2009, 1296, 35–45. [Google Scholar] [CrossRef] [Green Version]
- Pearson-Leary, J.; Mcnay, E.C. Novel Roles for the Insulin-Regulated Glucose Transporter-4 in Hippocampally Dependent Memory. J. Neurosci. 2016, 36, 11851–11864. [Google Scholar] [CrossRef] [Green Version]
- Radhakrishnan, Y.; Maile, L.A.; Ling, Y.; Graves, L.M.; Clemmons, D.R. Insulin-like growth factor-I stimulates Shc-dependent phosphatidylinositol 3-kinase activation via Grb2-associated p85 in vascular smooth muscle cells. J. Biol. Chem. 2008, 283, 16320–16331. [Google Scholar] [CrossRef] [Green Version]
- Belaidi, A.A.; Masaldan, S.; Southon, A.; Kalinowski, P.; Acevedo, K.; Appukuttan, A.T.; Portbury, S.; Lei, P.; Agarwal, P.; Leurgans, S.E.; et al. Apolipoprotein E potently inhibits ferroptosis by blocking ferritinophagy. Mol. Psychiatry 2022, 1–10. [Google Scholar] [CrossRef]
- Shen, L.; Wang, D.Q.H.; Tso, P.; Jandacek, R.J.; Woods, S.C.; Liu, M. Apolipoprotein E reduces food intake via PI3K/Akt signaling pathway in the hypothalamus. Physiol. Behav. 2011, 105, 124–128. [Google Scholar] [CrossRef]
- Ong, Q.R.; Chan, E.S.; Lim, M.L.; Wong, B.S. Expression of human apolipoprotein E4 reduces insulin-receptor substrate 1 expression and Akt phosphorylation in the ageing liver. FEBS Open Bio 2014, 4, 260–265. [Google Scholar] [CrossRef] [Green Version]
- Zhao, N.; Liu, C.C.; Van Ingelgom, A.J.; Martens, Y.A.; Linares, C.; Knight, J.A.; Painter, M.M.; Sullivan, P.M.; Bu, G.J. Apolipoprotein E4 Impairs Neuronal Insulin Signaling by Trapping Insulin Receptor in the Endosomes. Neuron 2017, 96, 115–129.e5. [Google Scholar] [CrossRef] [Green Version]
- Han, X.; Cheng, X.L.; Xu, J.Y.; Liu, Y.J.; Zhou, J.W.; Jiang, L.H.; Gu, X.P.; Xia, T.J. Activation of TREM2 attenuates neuroinflammation via PI3K/Akt signaling pathway to improve postoperative cognitive dysfunction in mice. Neuropharmacology 2022, 219, 109231. [Google Scholar] [CrossRef]
- Fu, C.; Xu, Q.C.; Liu, J.C.; Tang, S.X.; Liu, C.; Cao, Y.H. Triggering receptor expressed on myeloid cells-2 promotes survival of cardiomyocytes after myocardial ischemic injury through PI3K/AKT pathway. Cardiovasc. Diagn. Ther. 2022, 12, 24. [Google Scholar] [CrossRef]
- Wang, S.; Sudan, R.; Peng, V.; Zhou, Y.; Du, S.; Yuede, C.M.; Lei, T.; Hou, J.; Cai, Z.; Cella, M.; et al. TREM2 drives microglia response to amyloid-beta via SYK-dependent and -independent pathways. Cell 2022, 185, 4153–4169.e19. [Google Scholar] [CrossRef]
- Jiang, T.; Zhang, Y.D.; Gao, Q.; Ou, Z.; Gong, P.Y.; Shi, J.Q.; Wu, L.; Zhou, J.S. TREM2 Ameliorates Neuronal Tau Pathology through Suppression of Microglial Inflammatory Response. Inflammation 2018, 41, 811–823. [Google Scholar] [CrossRef]
- Frolich, L.; Blum-Degen, D.; Bernstein, H.G.; Engelsberger, S.; Humrich, J.; Laufer, S.; Muschner, D.; Thalheimer, A.; Turk, A.; Hoyer, S.; et al. Brain insulin and insulin receptors in aging and sporadic Alzheimer’s disease. J. Neural Transm. 1998, 105, 423–438. [Google Scholar] [CrossRef]
- Morelli, L.; Llovera, R.E.; Mathov, I.; Lue, L.F.; Frangione, B.; Ghiso, J.; Castano, E.M. Insulin-degrading enzyme in brain microvessels: Proteolysis of amyloid β vasculotropic variants and reduced activity in cerebral amyloid angiopathy. J. Biol. Chem. 2004, 279, 56004–56013. [Google Scholar] [CrossRef] [Green Version]
- Farris, W.; Mansourian, S.; Chang, Y.; Lindsley, L.; Eckman, E.A.; Frosch, M.P.; Eckman, C.B.; Tanzi, R.E.; Selkoe, D.J.; Guenette, S. 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. USA 2003, 100, 4162–4167. [Google Scholar] [CrossRef] [Green Version]
- Cook, D.G.; Leverenz, J.B.; McMillan, P.J.; Kulstad, J.J.; Ericksen, S.; Roth, R.A.; Schellenberg, G.D.; Jin, L.W.; Kovacina, K.S.; Craft, S. Reduced hippocampal insulin-degrading enzyme in late-onset Alzheimer’s disease is associated with the apolipoprotein E-epsilon 4 allele. Am. J. Pathol. 2003, 162, 313–319. [Google Scholar] [CrossRef]
- Schechter, R.; Beju, D.; Miller, K.E. The effect of insulin deficiency on tau and neurofilament in the insulin knockout mouse. Biochem. Biophys. Res. Commun. 2005, 334, 979–986. [Google Scholar] [CrossRef] [PubMed]
- Clodfelder-Miller, B.J.; Zmijewska, A.A.; Johnson, G.V.W.; Jope, R.S. Tau is hyperphosphorylated at multiple sites in mouse brain in vivo after streptozotocin-induced insulin deficiency. Diabetes 2006, 55, 3320–3325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schubert, M.; Brazil, D.P.; Burks, D.J.; Kushner, J.A.; Ye, J.; Flint, C.L.; Farhang-Fallah, J.; Dikkes, P.; Warot, X.M.; Rio, C.; et al. Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. J. Neurosci. 2003, 23, 7084–7092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adzovic, L.; Lynn, A.E.; D’Angelo, H.M.; Crockett, A.M.; Kaercher, R.M.; Royer, S.E.; Hopp, S.C.; Wenk, G.L. Insulin improves memory and reduces chronic neuroinflammation in the hippocampus of young but not aged brains. J. Neuroinflamm. 2015, 12, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reger, M.A.; Watson, G.S.; Frey, W.H., II; Baker, L.D.; Cholerton, B.; Keeling, M.L.; Belongia, D.A.; Fishel, M.A.; Plymate, S.R.; Schellenberg, G.D.; et al. Effects of intranasal insulin on cognition in memory-impaired older adults: Modulation by APOE genotype. Neurobiol. Aging 2006, 27, 451–458. [Google Scholar] [CrossRef]
- Reger, M.A.; Watson, G.S.; Green, P.S.; Wilkinson, C.W.; Baker, L.D.; Cholerton, B.; Fishel, M.A.; Plymate, S.R.; Breitner, J.C.; DeGroodt, W.; et al. Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology 2008, 70, 440–448. [Google Scholar] [CrossRef]
- Claxton, A.; Baker, L.D.; Hanson, A.; Trittschuh, E.H.; Cholerton, B.; Morgan, A.; Callaghan, M.; Arbuckle, M.; Behl, C.; Craft, S. Long Acting Intranasal Insulin Detemir Improves Cognition for Adults with Mild Cognitive Impairment or Early-Stage Alzheimer’s Disease Dementia. J. Alzheimer’s Dis. 2015, 45, 1269–1270. [Google Scholar] [CrossRef] [Green Version]
- Craft, S.; Claxton, A.; Baker, L.D.; Hanson, A.J.; Cholerton, B.; Trittschuh, E.H.; Dahl, D.; Caulder, E.; Neth, B.; Montine, T.J.; et al. Effects of Regular and Long-Acting Insulin on Cognition and Alzheimer’s Disease Biomarkers: A Pilot Clinical Trial. J. Alzheimer’s Dis. 2017, 57, 1325–1334. [Google Scholar] [CrossRef] [Green Version]
- Born, J.; Lange, T.; Kern, W.; McGregor, G.P.; Bickel, U.; Fehm, H.L. Sniffing neuropeptides: A transnasal approach to the human brain. Nat. Neurosci. 2002, 5, 514–516. [Google Scholar] [CrossRef]
- Craft, S.; Raman, R.; Chow, T.W.; Rafii, M.S.; Sun, C.K.; Rissman, R.A.; Donohue, M.C.; Brewer, J.B.; Jenkins, C.; Harless, K.; et al. Safety, Efficacy, and Feasibility of Intranasal Insulin for the Treatment of Mild Cognitive Impairment and Alzheimer Disease Dementia: A Randomized Clinical Trial. JAMA Neurol. 2020, 77, 1099–1109. [Google Scholar] [CrossRef]
- Sato, T.; Hanyu, H.; Hirao, K.; Kanetaka, H.; Sakurai, H.; Iwamoto, T. Efficacy of PPAR-gamma agonist pioglitazone in mild Alzheimer disease. Neurobiol. Aging 2011, 32, 1626–1633. [Google Scholar] [CrossRef]
- Watson, G.S.; Cholerton, B.A.; Reger, M.A.; Baker, L.D.; Plymate, S.R.; Asthana, S.; Fishel, M.A.; Kulstad, J.J.; Green, P.S.; Cook, D.G.; 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, 950–958. [Google Scholar] [CrossRef]
- Liu, J.; Wang, L.N.; Jia, J.P. Peroxisome Proliferator-Activated Receptor-Gamma Agonists for Alzheimer’s Disease and Amnestic Mild Cognitive Impairment: A Systematic Review and Meta-Analysis. Drugs Aging 2015, 32, 57–65. [Google Scholar] [CrossRef]
- Risner, M.E.; Saunders, A.M.; Altman, J.F.B.; Ormandy, G.C.; Craft, S.; Foley, I.M.; Zvartau-Hind, M.E.; Hosford, D.A.; Roses, A.D. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer’s disease. Pharm. J. 2006, 6, 246–254. [Google Scholar] [CrossRef] [Green Version]
- Harrington, C.; Sawchak, S.; Chiang, C.; Davies, J.; Donovan, C.; Saunders, A.M.; Irizarry, M.; Jeter, B.; Zvartau-Hind, M.; van Dyck, C.H.; et al. Rosiglitazone Does Not Improve Cognition or Global Function when Used as Adjunctive Therapy to AChE Inhibitors in Mild-to-Moderate Alzheimer’s Disease: Two Phase 3 Studies. Curr. Alzheimer Res. 2011, 8, 592–606. [Google Scholar] [CrossRef]
- Lu, X.Y.; Huang, S.; Chen, Q.B.; Zhang, D.P.; Li, W.Y.; Ao, R.; Leung, F.C.Y.; Zhang, Z.M.; Huang, J.S.; Tang, Y.; et al. Metformin Ameliorates A beta Pathology by Insulin-Degrading Enzyme in a Transgenic Mouse Model of Alzheimer’s Disease. Oxidative Med. Cell. Longev. 2020, 2020, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Ou, Z.R.; Kong, X.J.; Sun, X.D.; He, X.S.; Zhang, L.; Gong, Z.; Huang, J.Y.; Xu, B.A.; Long, D.H.; Li, J.H.; et al. Metformin treatment prevents amyloid plaque deposition and memory impairment in APP/PS1 mice. Brain Behav. Immun. 2018, 69, 351–363. [Google Scholar] [CrossRef]
- Kickstein, E.; Krauss, S.; Thornhill, P.; Rutschow, D.; Zeller, R.; Sharkey, J.; Williamson, R.; Fuchs, M.; Kohler, A.; Glossmann, H.; et al. Biguanide metformin acts on tau phosphorylation via mTOR/protein phosphatase 2A (PP2A) signaling. Proc. Natl. Acad. Sci. USA 2010, 107, 21830–21835. [Google Scholar] [CrossRef] [Green Version]
- Nabizadeh, F.; Kankam, S.B.; Balabandian, M.; Hashemi, S.M.; Sharifkazemi, H.; Rostami, M.R.; Alzheimer’s Disease Neuroimaging Initiative (ADNI). Metformin use and brain atrophy in nondemented elderly individuals with diabetes. Exp. Gerontol. 2022, 166, 111890. [Google Scholar] [CrossRef]
- Koenig, A.M.; Mechanic-Hamilton, D.; Xie, S.X.; Combs, M.F.; Cappola, A.R.; Xie, L.; Detre, J.A.; Wolk, D.A.; Arnold, S.E. Effects of the Insulin Sensitizer Metformin in Alzheimer Disease: Pilot Data from a Randomized Placebo-controlled Crossover Study. Alzheimer Dis. Assoc. Disord. 2017, 31, 107–113. [Google Scholar] [CrossRef]
- He, Z.J.; Wang, M.H.; Zhao, Q.H.; Li, X.Q.; Liu, P.G.; Ren, B.Y.; Wu, C.; Du, X.B.; Li, N.; Liu, Q. Bis(ethylmaltolato)oxidovanadium (IV) mitigates neuronal apoptosis resulted from amyloid-beta induced endoplasmic reticulum stress through activating peroxisome proliferator-activated receptor gamma. J. Inorg. Biochem. 2020, 208, 111073. [Google Scholar] [CrossRef]
- He, Z.J.; Song, J.X.; Li, X.X.; Li, X.Q.; Zhu, H.Z.; Wu, C.; Xiao, W.; Du, X.B.; Ni, J.Z.; Li, N.; et al. Bis(ethylmaltolato)oxidovanadium (IV) alleviates neuronal apoptosis through regulating peroxisome proliferator-activated receptor gamma in a triple transgenic animal model of Alzheimer’s disease. J. Biol. Inorg. Chem. 2021, 26, 551–568. [Google Scholar] [CrossRef] [PubMed]
- He, Z.J.; Han, S.X.; Wu, C.; Liu, L.N.; Zhu, H.Z.; Liu, A.; Lu, Q.Y.; Huang, J.Q.; Du, X.B.; Li, N.; et al. Bis(ethylmaltolato)oxidovanadium(iv) inhibited the pathogenesis of Alzheimer’s disease in triple transgenic model mice. Metallomics 2020, 12, 631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, Z.H.; Guo, J.L.; McBride, J.D.; Narasimhan, S.; Kim, H.; Changolkar, L.; Zhang, B.; Gathagan, R.J.; Yue, C.Y.; Dengler, C.; et al. Amyloid-beta plaques enhance Alzheimer’s brain tau-seeded pathologies by facilitating neuritic plaque tau aggregation. Nat. Med. 2018, 24, 29–38. [Google Scholar] [CrossRef] [PubMed]
- Pallo, S.P.; Johnson, G.V.W. Tau facilitates A beta-induced loss of mitochondrial membrane potential independent of cytosolic calcium fluxes in mouse cortical neurons. Neurosci. Lett. 2015, 597, 32–37. [Google Scholar] [CrossRef] [Green Version]
- Roberson, E.D.; Scearce-Levie, K.; Palop, J.J.; Yan, F.R.; Cheng, I.H.; Wu, T.; Gerstein, H.; Yu, G.Q.; Mucke, L. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse model. Science 2007, 316, 750–754. [Google Scholar] [CrossRef] [Green Version]
- Vossel, K.A.; Zhang, K.; Brodbeck, J.; Daub, A.C.; Sharma, P.; Finkbeiner, S.; Cui, B.; Mucke, L. Tau reduction prevents Abeta-induced defects in axonal transport. Science 2010, 330, 198. [Google Scholar] [CrossRef] [Green Version]
- Rapoport, M.; Dawson, H.N.; Binder, L.I.; Vitek, M.P.; Ferreira, A. Tau is essential to beta-amyloid-induced neurotoxicity. Proc. Natl. Acad. Sci. USA 2002, 99, 6364–6369. [Google Scholar] [CrossRef] [Green Version]
- Wijesekara, N.; Goncalves, R.A.; Ahrens, R.; De Felice, F.G.; Fraser, P.E. Tau ablation in mice leads to pancreatic beta cell dysfunction and glucose intolerance. FASEB J. 2018, 32, 3166–3173. [Google Scholar] [CrossRef] [Green Version]
- Marciniak, E.; Leboucher, A.; Caron, E.; Ahmed, T.; Tailleux, A.; Dumont, J.; Issad, T.; Gerhardt, E.; Pagesy, P.; Vileno, M.; et al. Tau deletion promotes brain insulin resistance. J. Exp. Med. 2017, 214, 2257–2269. [Google Scholar] [CrossRef]
- Lei, P.; Ayton, S.; Finkelstein, D.I.; Spoerri, L.; Ciccotosto, G.D.; Wright, D.K.; Wong, B.X.; Adlard, P.A.; Cherny, R.A.; Lam, L.Q.; et al. Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export. Nat. Med. 2012, 18, 291–295. [Google Scholar] [CrossRef]
- Abbondante, S.; Baglietto-Vargas, D.; Rodriguez-Ortiz, C.J.; Estrada-Hernandez, T.; Medeiros, R.; LaFerla, F.M. Genetic Ablation of Tau Mitigates Cognitive Impairment Induced by Type 1 Diabetes. Am. J. Pathol. 2014, 184, 819–826. [Google Scholar] [CrossRef] [Green Version]
- Trujillo-Estrada, L.; Nguyen, C.; da Cunha, C.; Cai, L.N.; Forner, S.; Martini, A.C.; Ager, R.R.; Prieto, G.A.; Cotman, C.W.; Baglietto-Vargas, D.; et al. Tau underlies synaptic and cognitive deficits for type 1, but not type 2 diabetes mouse models. Aging Cell 2019, 18, e12919. [Google Scholar] [CrossRef] [Green Version]
- Spires-Jones, T.L.; Stoothoff, W.H.; de Calignon, A.; Jones, P.B.; Hyman, B.T. Tau pathophysiology in neurodegeneration: A tangled issue. Trends Neurosci. 2009, 32, 150–159. [Google Scholar] [CrossRef]
- Hull, C.; Dekeryte, R.; Koss, D.J.; Crouch, B.; Buchanan, H.; Delibegovic, M.; Platt, B. Knock-in of Mutated hTAU Causes Insulin Resistance, Inflammation and Proteostasis Disturbance in a Mouse Model of Frontotemporal Dementia. Mol. Neurobiol. 2020, 57, 539–550. [Google Scholar] [CrossRef] [Green Version]
- Leboucher, A.; Ahmed, T.; Caron, E.; Tailleux, A.; Raison, S.; Joly-Amado, A.; Marciniak, E.; Carvalho, K.; Hamdane, M.; Bantubungi, K.; et al. Brain insulin response and peripheral metabolic changes in a Tau transgenic mouse model. Neurobiol. Dis. 2019, 125, 14–22. [Google Scholar] [CrossRef]
- El Idrissi, A.; Alonso, A.D.C. Pathological Human Tau Induces Alterations in the Brain Insulin Signaling Cascade. Front. Neurosci. 2022, 16, 805046. [Google Scholar] [CrossRef]
- Rodriguez-Rodriguez, P.; Sandebring-Matton, A.; Merino-Serrais, P.; Parrado-Fernandez, C.; Rabano, A.; Winblad, B.; Avila, J.; Ferrer, I.; Cedazo-Minguez, A. Tau hyperphosphorylation induces oligomeric insulin accumulation and insulin resistance in neurons. Brain 2017, 140, 3269–3285. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Balaji, V.; Kaniyappan, S.; Kruger, L.; Irsen, S.; Tepper, K.; Chandupatla, R.; Maetzler, W.; Schneider, A.; Mandelkow, E.; et al. The release and trans-synaptic transmission of Tau via exosomes. Mol. Neurodegener. 2017, 12, 5. [Google Scholar] [CrossRef] [Green Version]
- Sanders, D.W.; Kaufman, S.K.; DeVos, S.L.; Sharma, A.M.; Mirbaha, H.; Li, A.M.; Barker, S.J.; Foley, A.C.; Thorpe, J.R.; Serpell, L.C.; et al. Distinct Tau Prion Strains Propagate in Cells and Mice and Define Different Tauopathies. Neuron 2014, 82, 1271–1288. [Google Scholar] [CrossRef]
- Wegmann, S.; Maury, E.A.; Kirk, M.J.; Saqran, L.; Roe, A.; DeVos, S.L.; Nicholls, S.; Fan, Z.; Takeda, S.; Cagsal-Getkin, O.; et al. Removing endogenous tau does not prevent tau propagation yet reduces its neurotoxicity. EMBO J. 2015, 34, 3028–3041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prokopovich, D.V.; Whittaker, J.W.; Muthee, M.M.; Ahmed, A.; Larini, L. Impact of Phosphorylation and Pseudophosphorylation on the Early Stages of Aggregation of the Microtubule-Associated Protein Tau. J. Phys. Chem. B 2017, 121, 2095–2103. [Google Scholar] [CrossRef] [PubMed]
- Chang, E.; Kim, S.; Schafer, K.N.; Kuret, J. Pseudophosphorylation of tau protein directly modulates its aggregation kinetics. BBA Proteins Proteom. 2011, 1814, 388–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Briner, A.; Gotz, J.; Polanco, J.C. Fyn Kinase Controls Tau Aggregation In Vivo. Cell Rep. 2020, 32, 108045. [Google Scholar] [CrossRef] [PubMed]
- Haj-Yahya, M.; Gopinath, P.; Rajasekhar, K.; Mirbaha, H.; Diamond, M.I.; Lashuel, H.A. Site-Specific Hyperphosphorylation Inhibits, Rather than Promotes, Tau Fibrillization, Seeding Capacity, and Its Microtubule Binding. Angew. Chem. Int. Ed. 2020, 59, 4059–4067. [Google Scholar] [CrossRef] [Green Version]
- Ait-Bouziad, N.; Chiki, A.; Limorenko, G.; Xiao, S.F.; Eliezer, D.; Lashuel, H.A. Phosphorylation of the overlooked tyrosine 310 regulates the structure, aggregation, and microtubule- and lipid-binding properties of Tau. J. Biol. Chem. 2020, 295, 7905–7922. [Google Scholar] [CrossRef]
- Liu, K.F.; Liu, Y.T.; Li, L.Y.; Qin, P.B.; Iqbal, J.; Deng, Y.L.; Qing, H. Glycation alter the process of Tau phosphorylation to change Tau isoforms aggregation property. BBA Mol. Basis Dis. 2016, 1862, 192–201. [Google Scholar] [CrossRef]
- SantaCruz, K.; Lewis, J.; Spires, T.; Paulson, J.; Kotilinek, L.; Ingelsson, M.; Guimaraes, A.; DeTure, M.; Ramsden, M.; McGowan, E.; et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science 2005, 309, 476–481. [Google Scholar] [CrossRef] [Green Version]
- David, D.C.; Hauptmann, S.; Scherping, I.; Schuessel, K.; Keil, U.; Rizzu, P.; Ravid, R.; Drose, S.; Brandt, U.; Muller, W.E.; et al. Proteomic and functional analyses reveal a mitochondrial dysfunction in P301L Tau transgenic mice. J. Biol. Chem. 2005, 280, 23802–23814. [Google Scholar] [CrossRef] [Green Version]
- Esteras, N.; Rohrer, J.D.; Hardy, J.; Wray, S.; Abramov, A.Y. Mitochondrial hyperpolarization in iPSC-derived neurons from patients of FTDP-17 with 10+16 MAPT mutation leads to oxidative stress and neurodegeneration. Redox Biol. 2017, 12, 410–422. [Google Scholar] [CrossRef]
- Manczak, M.; Reddy, P.H. Abnormal interaction between the mitochondrial fission protein Drp1 and hyperphosphorylated tau in Alzheimer’s disease neurons: Implications for mitochondrial dysfunction and neuronal damage. Hum. Mol. Genet. 2012, 21, 2538–2547. [Google Scholar] [CrossRef] [Green Version]
- De Calignon, A.; Fox, L.M.; Pitstick, R.; Carlson, G.A.; Bacskai, B.J.; Spires-Jones, T.L.; Hyman, B.T. Caspase activation precedes and leads to tangles. Nature 2010, 464, 1201–1204. [Google Scholar] [CrossRef] [Green Version]
- Quintanilla, R.A.; Matthews-Roberson, T.A.; Dolan, P.J.; Johnson, G.V.W. Caspase-cleaved Tau Expression Induces Mitochondrial Dysfunction in Immortalized Cortical Neurons IMPLICATIONS FOR THE PATHOGENESIS OF ALZHEIMER DISEASE. J. Biol. Chem. 2009, 284, 18754–18766. [Google Scholar] [CrossRef] [Green Version]
- Perez, M.J.; Vergara-Pulgar, K.; Jara, C.; Cabezas-Opazo, F.; Quintanilla, R.A. Caspase-Cleaved Tau Impairs Mitochondrial Dynamics in Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 1004–1018. [Google Scholar] [CrossRef]
- Sengupta, A.; Novak, M.; Grundke-Iqbal, I.; Iqbal, K. Regulation of phosphorylation of tau by cyclin-dependent kinase 5 and glycogen synthase kinase-3 at substrate level. FEBS Lett. 2006, 580, 5925–5933. [Google Scholar] [CrossRef] [Green Version]
- Vingtdeux, V.; Davies, P.; Dickson, D.W.; Marambaud, P. AMPK is abnormally activated in tangle- and pre-tangle-bearing neurons in Alzheimer’s disease and other tauopathies. Acta Neuropathol. 2011, 121, 337–349. [Google Scholar] [CrossRef] [Green Version]
- Hanger, D.P.; Anderton, B.H.; Noble, W. Tau phosphorylation: The therapeutic challenge for neurodegenerative disease. Trends Mol. Med. 2009, 15, 112–119. [Google Scholar] [CrossRef]
- Caccamo, A.; Oddo, S.; Tran, L.X.; LaFerla, F.M. Lithium reduces tau phosphorylation but not A beta or working memory deficits in a transgenic model with both plaques and tangles. Am. J. Pathol. 2007, 170, 1669–1675. [Google Scholar] [CrossRef] [Green Version]
- Sereno, L.; Coma, M.; Rodriguez, M.; Sanchez-Ferrer, P.; Sanchez, M.B.; Gich, I.; Agullo, J.M.; Perez, M.; Avila, J.; Guardia-Laguarta, C.; et al. A novel GSK-3beta inhibitor reduces Alzheimer’s pathology and rescues neuronal loss in vivo. Neurobiol. Dis. 2009, 35, 359–367. [Google Scholar] [CrossRef]
- Hu, S.; Begum, A.N.; Jones, M.R.; Oh, M.S.; Beech, W.K.; Beech, B.H.; Yang, F.; Chen, P.; Ubeda, O.J.; Kim, P.C.; et al. GSK3 inhibitors show benefits in an Alzheimer’s disease (AD) model of neurodegeneration but adverse effects in control animals. Neurobiol. Dis. 2009, 33, 193–206. [Google Scholar] [CrossRef]
- Saraswati, A.P.; Ali Hussaini, S.M.; Krishna, N.H.; Babu, B.N.; Kamal, A. Glycogen synthase kinase-3 and its inhibitors: Potential target for various therapeutic conditions. Eur. J. Med. Chem. 2018, 144, 843–858. [Google Scholar] [CrossRef] [PubMed]
- Sontag, E.; Luangpirom, A.; Hladik, C.; Mudrak, I.; Ogris, E.; Speciale, S.; White, C.L., III. Altered expression levels of the protein phosphatase 2A ABalphaC enzyme are associated with Alzheimer disease pathology. J. Neuropathol. Exp. Neurol. 2004, 63, 287–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kins, S.; Crameri, A.; Evans, D.R.H.; Hemmings, B.A.; Nitsch, R.M.; Gotz, J. Reduced protein phosphatase 2A activity induces hyperphosphorylation and altered compartmentalization of tau in transgenic mice. J. Biol. Chem. 2001, 276, 38193–38200. [Google Scholar] [CrossRef] [PubMed]
- Pei, J.J.; Gong, C.X.; An, W.L.; Winblad, B.; Cowburn, R.F.; Grundke-Iqbal, I.; Iqbal, K. Okadaic-acid-induced inhibition of protein phosphatase 2A produces activation of mitogen-activated protein kinases ERK1/2, MEK-1/2, and p70 S6, similar to that in Alzheimer’s disease. Am. J. Pathol. 2003, 163, 845–858. [Google Scholar] [CrossRef] [PubMed]
- St-Cyr Giguere, F.; Attiori Essis, S.; Chagniel, L.; Germain, M.; Cyr, M.; Massicotte, G. The sphingosine-1-phosphate receptor 1 agonist SEW2871 reduces Tau-Ser262 phosphorylation in rat hippocampal slices. Brain Res. 2017, 1658, 51–59. [Google Scholar] [CrossRef]
- Xiong, Y.; Jing, X.P.; Zhou, X.W.; Wang, X.L.; Yang, Y.; Sun, X.Y.; Qiu, M.; Cao, F.Y.; Lu, Y.M.; Liu, R.; et al. Zinc induces protein phosphatase 2A inactivation and tau hyperphosphorylation through Src dependent PP2A (tyrosine 307) phosphorylation. Neurobiol. Aging 2013, 34, 745–756. [Google Scholar] [CrossRef]
- Ginhoux, F.; Lim, S.; Hoeffel, G.; Low, D.; Huber, T. Origin and differentiation of microglia. Front. Cell. Neurosci. 2013, 7, 45. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.; Xu, Z.; Xiong, S.; Sun, F.; Qin, G.; Hu, G.; Wang, J.; Zhao, L.; Liang, Y.X.; Wu, T.; et al. Repopulated microglia are solely derived from the proliferation of residual microglia after acute depletion. Nat. Neurosci. 2018, 21, 530–540. [Google Scholar] [CrossRef]
- Paolicelli, R.C.; Bolasco, G.; Pagani, F.; Maggi, L.; Scianni, M.; Panzanelli, P.; Giustetto, M.; Ferreira, T.A.; Guiducci, E.; Dumas, L.; et al. Synaptic pruning by microglia is necessary for normal brain development. Science 2011, 333, 1456–1458. [Google Scholar] [CrossRef] [Green Version]
- Condello, C.; Yuan, P.; Schain, A.; Grutzendler, J. Microglia constitute a barrier that prevents neurotoxic protofibrillar A beta 42 hotspots around plaques. Nat. Commun. 2015, 6, 6176. [Google Scholar] [CrossRef]
- Yuan, P.; Condello, C.; Keene, C.D.; Wang, Y.M.; Bird, T.D.; Paul, S.M.; Luo, W.J.; Colonna, M.; Baddeley, D.; Grutzendler, J. TREM2 Haplodeficiency in Mice and Humans Impairs the Microglia Barrier Function Leading to Decreased Amyloid Compaction and Severe Axonal Dystrophy. Neuron 2016, 92, 252–264. [Google Scholar] [CrossRef] [Green Version]
- Heneka, M.T.; Golenbock, D.T.; Latz, E. Innate immunity in Alzheimer’s disease. Nat. Immunol. 2015, 16, 229–236. [Google Scholar] [CrossRef]
- Heneka, M.T.; Kummer, M.P.; Stutz, A.; Delekate, A.; Schwartz, S.; Vieira-Saecker, A.; Griep, A.; Axt, D.; Remus, A.; Tzeng, T.C.; et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 2013, 493, 674–678. [Google Scholar] [CrossRef] [Green Version]
- Keren-Shaul, H.; Spinrad, A.; Weiner, A.; Matcovitch-Natan, O.; Dvir-Szternfeld, R.; Ulland, T.K.; David, E.; Baruch, K.; Lara-Astaiso, D.; Toth, B.; et al. A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease. Cell 2017, 169, 1276–1290.e17. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Colonna, M. Microglia in Alzheimer’s disease at single-cell level. Are there common in humans and mice? J. Exp. Med. 2021, 218, e20202717. [Google Scholar] [CrossRef]
- Bhaskar, K.; Konerth, M.; Kokiko-Cochran, O.N.; Cardona, A.; Ransohoff, R.M.; Lamb, B.T. Regulation of Tau Pathology by the Microglial Fractalkine Receptor. Neuron 2010, 68, 19–31. [Google Scholar] [CrossRef] [Green Version]
- Kitazawa, M.; Oddo, S.; Yamasaki, T.R.; Green, K.N.; LaFerla, F.M. Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer’s disease. J. Neurosci. 2005, 25, 8843–8853. [Google Scholar] [CrossRef] [Green Version]
- Shi, Y.; Yamada, K.; Liddelow, S.A.; Smith, S.T.; Zhao, L.Z.; Luo, W.J.; Tsai, R.M.; Spina, S.; Grinberg, L.T.; Rojas, J.C.; et al. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 2017, 549, 523–527. [Google Scholar] [CrossRef]
- Gratuze, M.; Chen, Y.; Parhizkar, S.; Jain, N.; Strickland, M.R.; Serrano, J.R.; Colonna, M.; Ulrich, J.D.; Holtzman, D.M. Activated microglia mitigate Abeta-associated tau seeding and spreading. J. Exp. Med. 2021, 218, e20210542. [Google Scholar] [CrossRef]
- Leyns, C.E.G.; Gratuze, M.; Narasimhan, S.; Jain, N.; Koscal, L.J.; Jiang, H.; Manis, M.; Colonna, M.; Lee, V.M.Y.; Ulrich, J.D.; et al. TREM2 function impedes tau seeding in neuritic plaques. Nat. Neurosci. 2019, 22, 1217–1222. [Google Scholar] [CrossRef]
- Lee, S.H.; Meilandt, W.J.; Xie, L.; Gandham, V.D.; Ngu, H.; Barck, K.H.; Rezzonico, M.G.; Imperio, J.; Lalehzadeh, G.; Huntley, M.A.; et al. Trem2 restrains the enhancement of tau accumulation and neurodegeneration by beta-amyloid pathology. Neuron 2021, 109, 1283–1301.e6. [Google Scholar] [CrossRef] [PubMed]
- Ulland, T.K.; Colonna, M. TREM2—A key player in microglial biology and Alzheimer disease. Nat. Rev. Neurol. 2018, 14, 667–675. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.H.; Chen, S.W. Role of TREM2 in the Development of Neurodegenerative Diseases after Traumatic Brain Injury. Mol. Neurobiol. 2022, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Elmore, M.R.; Najafi, A.R.; Koike, M.A.; Dagher, N.N.; Spangenberg, E.E.; Rice, R.A.; Kitazawa, M.; Matusow, B.; Nguyen, H.; West, B.L.; et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 2014, 82, 380–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, S.; Player, M.R. Colony-stimulating factor-1 receptor inhibitors for the treatment of cancer and inflammatory disease. Curr. Top. Med. Chem. 2009, 9, 599–610. [Google Scholar] [CrossRef] [PubMed]
- Dai, X.M.; Ryan, G.R.; Hapel, A.J.; Dominguez, M.G.; Russell, R.G.; Kapp, S.; Sylvestre, V.; Stanley, E.R. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects. Blood 2002, 99, 111–120. [Google Scholar] [CrossRef]
- Erblich, B.; Zhu, L.; Etgen, A.M.; Dobrenis, K.; Pollard, J.W. Absence of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain development and olfactory deficits. PLoS ONE 2011, 6, e26317. [Google Scholar] [CrossRef] [Green Version]
- Conway, J.G.; McDonald, B.; Parham, J.; Keith, B.; Rusnak, D.W.; Shaw, E.; Jansen, M.; Lin, P.; Payne, A.; Crosby, R.M.; et al. Inhibition of colony-stimulating-factor-1 signaling in vivo with the orally bioavailable cFMS kinase inhibitor GW2580. Proc. Natl. Acad. Sci. USA 2005, 102, 16078–16083. [Google Scholar] [CrossRef] [Green Version]
- Olmos-Alonso, A.; Schetters, S.T.; Sri, S.; Askew, K.; Mancuso, R.; Vargas-Caballero, M.; Holscher, C.; Perry, V.H.; Gomez-Nicola, D. Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology. Brain 2016, 139, 891–907. [Google Scholar] [CrossRef] [Green Version]
- Spangenberg, E.E.; Lee, R.J.; Najafi, A.R.; Rice, R.A.; Elmore, M.R.P.; Blurton-Jones, M.; West, B.L.; Green, K.N. Eliminating microglia in Alzheimer’s mice prevents neuronal loss without modulating amyloid-beta pathology. Brain 2016, 139, 1265–1281. [Google Scholar] [CrossRef]
- Spangenberg, E.; Severson, P.L.; Hohsfield, L.A.; Crapser, J.; Zhang, J.Z.; Burton, E.A.; Zhang, Y.; Spevak, W.; Line, J.; Phan, N.Y.; et al. Sustained microglial depletion with CSF1R inhibitor impairs parenchymal plaque development in an Alzheimer’s disease model. Nat. Commun. 2019, 10, 3758. [Google Scholar] [CrossRef] [Green Version]
- Lamb, Y.N. Pexidartinib: First Approval. Drugs 2019, 79, 1805–1812. [Google Scholar] [CrossRef]
- Bennett, R.E.; Bryant, A.; Hu, M.; Robbins, A.B.; Hopp, S.C.; Hyman, B.T. Partial reduction of microglia does not affect tau pathology in aged mice. J. Neuroinflamm. 2018, 15, 311. [Google Scholar] [CrossRef]
- Sosna, J.; Philipp, S.; Albay, R., III; Reyes-Ruiz, J.M.; Baglietto-Vargas, D.; LaFerla, F.M.; Glabe, C.G. Early long-term administration of the CSF1R inhibitor PLX3397 ablates microglia and reduces accumulation of intraneuronal amyloid, neuritic plaque deposition and pre-fibrillar oligomers in 5XFAD mouse model of Alzheimer’s disease. Mol. Neurodegener. 2018, 13, 11. [Google Scholar] [CrossRef]
- Asai, H.; Ikezu, S.; Tsunoda, S.; Medalla, M.; Luebke, J.; Haydar, T.; Wolozin, B.; Butovsky, O.; Kugler, S.; Ikezu, T. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci. 2015, 18, 1584–1593. [Google Scholar] [CrossRef] [Green Version]
- Clayton, K.; Delpech, J.C.; Herron, S.; Iwahara, N.; Ericsson, M.; Saito, T.; Saido, T.C.; Ikezu, S.; Ikezu, T. Plaque associated microglia hyper-secrete extracellular vesicles and accelerate tau propagation in a humanized APP mouse model. Mol. Neurodegener. 2021, 16, 18. [Google Scholar] [CrossRef]
- Casali, B.T.; MacPherson, K.P.; Reed-Geaghan, E.G.; Landreth, G.E. Microglia depletion rapidly and reversibly alters amyloid pathology by modification of plaque compaction and morphologies. Neurobiol. Dis. 2020, 142, 104956. [Google Scholar] [CrossRef]
- Karaahmet, B.; Le, L.; Mendes, M.S.; Majewska, A.K.; O’Banion, M.K. Repopulated microglia induce expression of Cxcl13 with differential changes in Tau phosphorylation but do not impact amyloid pathology. J. Neuroinflamm. 2022, 19, 173. [Google Scholar] [CrossRef]
- Selkoe, D.J.; Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 2016, 8, 595–608. [Google Scholar] [CrossRef]
- Bartus, R.T.; Dean, R.L., III; Beer, B.; Lippa, A.S. The cholinergic hypothesis of geriatric memory dysfunction. Science 1982, 217, 408–414. [Google Scholar] [CrossRef]
- McGeer, P.L.; Rogers, J.; McGeer, E.G. Neuroimmune mechanisms in Alzheimer disease pathogenesis. Alzheimer Dis. Assoc. Disord. 1994, 8, 149–158. [Google Scholar] [CrossRef] [PubMed]
- Swerdlow, R.H.; Burns, J.M.; Khan, S.M. The Alzheimer’s disease mitochondrial cascade hypothesis. J. Alzheimer’s Dis. 2010, 20 (Suppl. 2), S265–S279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, X.; Raina, A.K.; Perry, G.; Smith, M.A. Alzheimer’s disease: The two-hit hypothesis. Lancet Neurol. 2004, 3, 219–226. [Google Scholar] [CrossRef] [PubMed]
- De la Monte, S.M. Insulin resistance and Alzheimer’s disease. BMB Rep. 2009, 42, 475–481. [Google Scholar] [CrossRef] [PubMed]
- Alzheimer’s Association Calcium Hypothesis Workgroup; Khachaturian, Z.S. Calcium Hypothesis of Alzheimer’s disease and brain aging: A framework for integrating new evidence into a comprehensive theory of pathogenesis. Alzheimer’s Dement. 2017, 13, 178–182.e17. [Google Scholar] [CrossRef]
Strategies | Targets | Drugs or Methods | Anticipating Function | Side Effects |
---|---|---|---|---|
Lowering Aβ | ADAM 10 | Retinoid acitretin PEP3 | Activating ADAM10 Upregulating the postsynaptic localization and activity of ADAM10 | Perturbed unspecific substrates besides APP |
BACE1 | Verubecestat Lanabecestat | BACE1 inhibitor | Perturbed unspecific substrates besides APP | |
γ-secretase | Semagacestat Avagacestat | γ-secretase inhibitor | Perturbed unspecific substrates besides APP | |
Aβ-oligomer | Aducanumab | Aβ-oligomer antibody | Encephaledema | |
Increasing insulin signaling | Insulin deficiency | Intranasal insulin administration | Increasing insulin level in CNS | - |
Insulin insensitivity | Metformin | Increasing insulin insensitivity | - | |
PPARγ | Pioglitazone Rosiglitazone | Increasing insulin insensitivity | Edema and weight gain | |
Limiting tau phosphorylation | GSK3β | Lithium Tideglusib | GSK3β inhibitor | Perturbed unspecific substrates besides tau |
PP2A | SEW2871 Zinc chelator clioquinol | PP2A activator | Perturbed unspecific substrates besides tau | |
Restrict inflammations | Microglia | GW2580 PLX5622 PLX3397 | Depletion and repopulation of microglia | - |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
You, G.; Yao, J.; Liu, Q.; Li, N. The Strategies for Treating “Alzheimer’s Disease”: Insulin Signaling May Be a Feasible Target. Curr. Issues Mol. Biol. 2022, 44, 6172-6188. https://doi.org/10.3390/cimb44120421
You G, Yao J, Liu Q, Li N. The Strategies for Treating “Alzheimer’s Disease”: Insulin Signaling May Be a Feasible Target. Current Issues in Molecular Biology. 2022; 44(12):6172-6188. https://doi.org/10.3390/cimb44120421
Chicago/Turabian StyleYou, Guanying, Jinyi Yao, Qiong Liu, and Nan Li. 2022. "The Strategies for Treating “Alzheimer’s Disease”: Insulin Signaling May Be a Feasible Target" Current Issues in Molecular Biology 44, no. 12: 6172-6188. https://doi.org/10.3390/cimb44120421
APA StyleYou, G., Yao, J., Liu, Q., & Li, N. (2022). The Strategies for Treating “Alzheimer’s Disease”: Insulin Signaling May Be a Feasible Target. Current Issues in Molecular Biology, 44(12), 6172-6188. https://doi.org/10.3390/cimb44120421