Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s Disease (AD): From Risk Factors to Therapeutic Targeting
Abstract
:1. Introduction
2. The Pivotal Role of Neuroinflammation in AD Onset
3. Evidence of HMGB1 Implication in AD Pathogenesis
4. Implication of RAGE in AD
5. TLR4 Involvement in AD Pathogenesis
6. HMGB1, RAGE, and TLR4 as Potential Clinical Biomarkers of AD
7. HMGB1, RAGE, and TLR4 Inhibition/Blockade as a Potential Therapy against AD
7.1. Effects of HMGB1 Neutralization in AD
7.2. Effects of RAGE Inhibition in AD
7.3. Effects of TLR4 Blockade in AD
8. Discussion and Future Implications
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
References
- Tavana, J.P.; Rosene, M.; Jensen, N.O.; Ridge, P.G.; Kauwe, J.S.; Karch, C.M. RAB10: An Alzheimer’s disease resilience locus and potential drug target. Clin. Interv. Aging 2019, 14, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laurent, C.; Buée, L.; Blum, D. Tau and neuroinflammation: What impact for Alzheimer’s disease and tauopathies? Biomed. J. 2018, 41, 21–33. [Google Scholar] [CrossRef] [PubMed]
- Saido, T. Alzheimer’s disease as proteolytic disorders: Anabolism and catabolism of β-amyloid. Neurobiol. Aging 1998, 19, S69–S75. [Google Scholar] [CrossRef]
- Piaceri, I.; Nacmias, B.; Sorbi, S. Genetics of familial and sporadic Alzheimer’s disease. Front. Biosci. Elite Ed. 2013, 5, 167–177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bertram, L.; Lill, C.M.; Tanzi, R.E. The genetics of Alzheimer disease: Back to the future. Neuron 2010, 68, 270–281. [Google Scholar] [CrossRef] [Green Version]
- Salomone, S.; Caraci, F.; Leggio, G.M.; Fedotova, J.; Drago, F. New pharmacological strategies for treatment of Alzheimer’s disease: Focus on disease modifying drugs. Br. J. Clin. Pharmacol. 2012, 73, 504–517. [Google Scholar] [CrossRef] [Green Version]
- Castellani, R.J.; Perry, G. Pathogenesis and disease-modifying therapy in Alzheimer’s disease: The flat line of progress. Arch. Med. Res. 2012, 43, 694–698. [Google Scholar] [CrossRef]
- Van Eldik, L.J.; Carrillo, M.C.; Cole, P.E.; Feuerbach, D.; Greenberg, B.D.; Hendrix, J.A.; Kennedy, M.; Kozauer, N.; Margolin, R.A.; Molinuevo, J.L. The roles of inflammation and immune mechanisms in Alzheimer’s disease. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2016, 2, 99–109. [Google Scholar] [CrossRef] [Green Version]
- Štros, M. HMGB proteins: Interactions with DNA and chromatin. Biochim. Biophys. Acta BBA Gene Regul. Mech. 2010, 1799, 101–113. [Google Scholar] [CrossRef]
- Andersson, U.; Yang, H.; Harris, H. High-mobility group box 1 protein (HMGB1) operates as an alarmin outside as well as inside cells. In Seminars in Immunology; Academic Press: New York, NY, USA, 2018; pp. 40–48. [Google Scholar]
- Paudel, Y.N.; Angelopoulou, E.; Piperi, C.; Balasubramaniam, V.R.; Othman, I.; Shaikh, M.F. Enlightening the role of high mobility group box 1 (HMGB1) in inflammation: Updates on receptor signalling. Eur. J. Pharmacol. 2019, 858, 172487. [Google Scholar] [CrossRef]
- Andersson, U.; Yang, H.; Harris, H. Extracellular HMGB1 as a therapeutic target in inflammatory diseases. Expert Opin. Ther. Targets 2018, 22, 263–277. [Google Scholar] [CrossRef] [PubMed]
- Tang, Y.; Zhao, X.; Antoine, D.; Xiao, X.; Wang, H.; Andersson, U.; Billiar, T.R.; Tracey, K.J.; Lu, B. Regulation of posttranslational modifications of HMGB1 during immune responses. Antioxid. Redox Signal. 2016, 24, 620–634. [Google Scholar] [CrossRef] [PubMed]
- Andersson, U.; Antoine, D.; Tracey, K. Expression of Concern: The functions of HMGB 1 depend on molecular localization and post-Translational modifications. J. Intern. Med. 2014, 276, 420–424. [Google Scholar] [CrossRef] [Green Version]
- Bianchi, M.E. HMGB1 loves company. J. Leukoc. Biol. 2009, 86, 573–576. [Google Scholar] [CrossRef] [PubMed]
- Klune, J.R.; Dhupar, R.; Cardinal, J.; Billiar, T.R.; Tsung, A. HMGB1: Endogenous danger signaling. Mol. Med. 2008, 14, 476–484. [Google Scholar] [CrossRef] [PubMed]
- Bianchi, M.E. DAMPs, PAMPs and alarmins: All we need to know about danger. J. Leukoc. Biol. 2007, 81, 1–5. [Google Scholar] [CrossRef]
- Nan, K.; Han, Y.; Fang, Q.; Huang, C.; Yu, L.; Ge, W.; Xiang, F.; Tao, Y.-X.; Cao, H.; Li, J. HMGB1 gene silencing inhibits neuroinflammation via down-regulation of NF-κB signaling in primary hippocampal neurons induced by Aβ25–35. Int. Immunopharmacol. 2019, 67, 294–301. [Google Scholar] [CrossRef]
- Takata, K.; Kitamura, Y.; Kakimura, J.-I.; Shibagaki, K.; Tsuchiya, D.; Taniguchi, T.; Smith, M.A.; Perry, G.; Shimohama, S. Role of high mobility group protein-1 (HMG1) in amyloid-β homeostasis. Biochem. Biophys. Res. Commun. 2003, 301, 699–703. [Google Scholar] [CrossRef]
- Du Yan, S.; Chen, X.; Fu, J.; Chen, M.; Zhu, H.; Roher, A.; Slattery, T.; Zhao, L.; Nagashima, M.; Morser, J. RAGE and amyloid-β peptide neurotoxicity in Alzheimer’s disease. Nature 1996, 382, 685. [Google Scholar] [CrossRef]
- Miron, J.; Picard, C.; Frappier, J.; Dea, D.; Theroux, L.; Poirier, J. TLR4 gene expression and pro-inflammatory cytokines in Alzheimer’s disease and in response to hippocampal deafferentation in rodents. J. Alzheimer’s Dis. 2018, 63, 1547–1556. [Google Scholar] [CrossRef]
- Cai, Z.; Liu, N.; Wang, C.; Qin, B.; Zhou, Y.; Xiao, M.; Chang, L.; Yan, L.-J.; Zhao, B. Role of RAGE in Alzheimer’s disease. Cell. Mol. Neurobiol. 2016, 36, 483–495. [Google Scholar] [CrossRef] [PubMed]
- Walter, S.; Letiembre, M.; Liu, Y.; Heine, H.; Penke, B.; Hao, W.; Bode, B.; Manietta, N.; Walter, J.; Schulz-Schüffer, W. Role of the toll-like receptor 4 in neuroinflammation in Alzheimer’s disease. Cell. Physiol. Biochem. 2007, 20, 947–956. [Google Scholar] [CrossRef] [PubMed]
- Kinney, J.W.; Bemiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2018, 4, 575–590. [Google Scholar] [CrossRef] [PubMed]
- Heppner, F.L.; Ransohoff, R.M.; Becher, B. Immune attack: The role of inflammation in Alzheimer disease. Nat. Rev. Neurosci. 2015, 16, 358. [Google Scholar] [CrossRef] [PubMed]
- Chaney, A.; Williams, S.R.; Boutin, H. In vivo molecular imaging of neuroinflammation in Alzheimer’s disease. J. Neurochem. 2019, 149, 438–451. [Google Scholar] [CrossRef] [Green Version]
- Heneka, M.T.; O’Banion, M.K.; Terwel, D.; Kummer, M.P. Neuroinflammatory processes in Alzheimer’s disease. J. Neural Transm. 2010, 117, 919–947. [Google Scholar] [CrossRef]
- Hu, J.; Akama, K.T.; Krafft, G.A.; Chromy, B.A.; Van Eldik, L.J. Amyloid-β peptide activates cultured astrocytes: Morphological alterations, cytokine induction and nitric oxide release. Brain Res. 1998, 785, 195–206. [Google Scholar] [CrossRef]
- Zhu, M.; Wang, X.; Sun, L.; Schultzberg, M.; Hjorth, E. Can inflammation be resolved in Alzheimer’s disease? Ther. Adv. Neurol. Disord. 2018, 11, 1756286418791107. [Google Scholar] [CrossRef]
- Pasqualetti, G.; Brooks, D.J.; Edison, P. The role of neuroinflammation in dementias. Curr. Neurol. Neurosci. Rep. 2015, 15, 17. [Google Scholar] [CrossRef]
- Dionisio-Santos, D.A.; Olschowka, J.A.; O’Banion, M.K. Exploiting microglial and peripheral immune cell crosstalk to treat Alzheimer’s disease. J. Neuroinflamm. 2019, 16, 74. [Google Scholar] [CrossRef] [Green Version]
- Chi, W.; Chen, H.; Li, F.; Zhu, Y.; Yin, W.; Zhuo, Y. HMGB1 promotes the activation of NLRP3 and caspase-8 inflammasomes via NF-κB pathway in acute glaucoma. J. Neuroinflamm. 2015, 12, 137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fang, P.; Schachner, M.; Shen, Y.-Q. HMGB1 in development and diseases of the central nervous system. Mol. Neurobiol. 2012, 45, 499–506. [Google Scholar] [CrossRef] [PubMed]
- Harris, H.E.; Andersson, U.; Pisetsky, D.S. HMGB1: A multifunctional alarmin driving autoimmune and inflammatory disease. Nat. Rev. Rheumatol. 2012, 8, 195. [Google Scholar] [CrossRef] [PubMed]
- Magna, M.; Pisetsky, D.S. The role of HMGB1 in the pathogenesis of inflammatory and autoimmune diseases. Mol. Med. 2014, 20, 138–146. [Google Scholar] [CrossRef]
- Falcão, A.S.; Carvalho, L.A.; Lidónio, G.A.; Vaz, A.R.; Lucas, S.D.; Moreira, R.; Brites, D. Dipeptidyl vinyl sulfone as a novel chemical tool to inhibit HMGB1/NLRP3-inflammasome and inflamma-miRs in Aβ-mediated microglial inflammation. ACS Chem. Neurosci. 2016, 8, 89–99. [Google Scholar] [CrossRef]
- Fujita, K.; Motoki, K.; Tagawa, K.; Chen, X.; Hama, H.; Nakajima, K.; Homma, H.; Tamura, T.; Watanabe, H.; Katsuno, M. HMGB1, a pathogenic molecule that induces neurite degeneration via TLR4-MARCKS, is a potential therapeutic target for Alzheimer’s disease. Sci. Rep. 2016, 6, 31895. [Google Scholar] [CrossRef] [Green Version]
- Takata, K.; Kitamura, Y.; Tsuchiya, D.; Kawasaki, T.; Taniguchi, T.; Shimohama, S. High mobility group box protein-1 inhibits microglial Aβ clearance and enhances Aβ neurotoxicity. J. Neurosci. Res. 2004, 78, 880–891. [Google Scholar] [CrossRef]
- Takata, K.; Takada, T.; Ito, A.; Asai, M.; Tawa, M.; Saito, Y.; Ashihara, E.; Tomimoto, H.; Kitamura, Y.; Shimohama, S. Microglial Amyloid-β1-40 Phagocytosis Dysfunction Is Caused by High-Mobility Group Box Protein-1: Implications for the Pathological Progression of Alzheimer’s Disease. Int. J. Alzheimer’s Dis. 2012, 2012, 685739. [Google Scholar] [CrossRef] [Green Version]
- Lue, L.-F.; Walker, D.G.; Brachova, L.; Beach, T.G.; Rogers, J.; Schmidt, A.M.; Stern, D.M.; Du Yan, S. Involvement of microglial receptor for advanced glycation endproducts (RAGE) in Alzheimer’s disease: Identification of a cellular activation mechanism. Exp. Neurol. 2001, 171, 29–45. [Google Scholar] [CrossRef]
- Tahara, K.; Kim, H.-D.; Jin, J.-J.; Maxwell, J.A.; Li, L.; Fukuchi, K.-I. Role of toll-like receptor signalling in Aβ uptake and clearance. Brain 2006, 129, 3006–3019. [Google Scholar] [CrossRef] [Green Version]
- Jang, A.; Liew, H.; Kim, Y.-M.; Choi, H.; Kim, S.; Hyung Lee, S.; Ohshima, T.; Mikoshiba, K.; Suh, Y.-H. p35 deficiency accelerates HMGB-1-mediated neuronal death in the early stages of an Alzheimer’s disease mouse model. Curr. Alzheimer Res. 2013, 10, 829–843. [Google Scholar] [CrossRef]
- Tobin, M.K.; Musaraca, K.; Disouky, A.; Shetti, A.; Bheri, A.; Honer, W.G.; Kim, N.; Dawe, R.J.; Bennett, D.A.; Arfanakis, K. Human Hippocampal Neurogenesis Persists in Aged Adults and Alzheimer’s Disease Patients. Cell Stem Cell 2019, 24, 974–982. [Google Scholar] [CrossRef] [PubMed]
- Moreno-Jiménez, E.P.; Flor-García, M.; Terreros-Roncal, J.; Rábano, A.; Cafini, F.; Pallas-Bazarra, N.; Ávila, J.; Llorens-Martín, M. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat. Med. 2019, 25, 554. [Google Scholar] [CrossRef] [PubMed]
- Zeng, Q.; Zheng, M.; Zhang, T.; He, G. Hippocampal neurogenesis in the APP/PS1/nestin-GFP triple transgenic mouse model of Alzheimer’s disease. Neuroscience 2016, 314, 64–74. [Google Scholar] [CrossRef] [PubMed]
- Donovan, M.H.; Yazdani, U.; Norris, R.D.; Games, D.; German, D.C.; Eisch, A.J. Decreased adult hippocampal neurogenesis in the PDAPP mouse model of Alzheimer’s disease. J. Comp. Neurol. 2006, 495, 70–83. [Google Scholar] [CrossRef] [PubMed]
- Jin, K.; Galvan, V.; Xie, L.; Mao, X.O.; Gorostiza, O.F.; Bredesen, D.E.; Greenberg, D.A. Enhanced neurogenesis in Alzheimer’s disease transgenic (PDGF-APPSw, Ind) mice. Proc. Natl. Acad. Sci. USA 2004, 101, 13363–13367. [Google Scholar] [CrossRef] [Green Version]
- Choi, S.H.; Tanzi, R.E. Is Alzheimer’s Disease a Neurogenesis Disorder? Cell Stem Cell 2019, 25, 7–8. [Google Scholar] [CrossRef]
- Meneghini, V.; Bortolotto, V.; Francese, M.T.; Dellarole, A.; Carraro, L.; Terzieva, S.; Grilli, M. High-mobility group box-1 protein and β-amyloid oligomers promote neuronal differentiation of adult hippocampal neural progenitors via receptor for advanced glycation end products/nuclear factor-κB axis: Relevance for Alzheimer’s disease. J. Neurosci. 2013, 33, 6047–6059. [Google Scholar] [CrossRef] [Green Version]
- Mazarati, A.; Maroso, M.; Iori, V.; Vezzani, A.; Carli, M. High-mobility group box-1 impairs memory in mice through both toll-like receptor 4 and receptor for advanced glycation end products. Exp. Neurol. 2011, 232, 143–148. [Google Scholar] [CrossRef] [Green Version]
- MacLean, M.; Derk, J.; Ruiz, H.H.; Juranek, J.K.; Ramasamy, R.; Schmidt, A.M. The Receptor for Advanced Glycation End Products (RAGE) and DIAPH1: Implications for vascular and neuroinflammatory dysfunction in disorders of the central nervous system. Neurochem. Int. 2019, 126, 154–164. [Google Scholar] [CrossRef]
- Avalos, A.M.; Kiefer, K.; Tian, J.; Christensen, S.; Shlomchik, M.; Coyle, A.J.; Marshak-Rothstein, A. RAGE-independent autoreactive B cell activation in response to chromatin and HMGB1/DNA immune complexes. Autoimmunity 2010, 43, 103–110. [Google Scholar] [CrossRef] [PubMed]
- Daffu, G.; Shen, X.; Senatus, L.; Thiagarajan, D.; Abedini, A.; del Pozo, C.H.; Rosario, R.; Song, F.; Friedman, R.A.; Ramasamy, R. RAGE suppresses ABCG1-mediated macrophage cholesterol efflux in diabetes. Diabetes 2015, 64, 4046–4060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dumitriu, I.E.; Baruah, P.; Bianchi, M.E.; Manfredi, A.A.; Rovere-Querini, P. Requirement of HMGB1 and RAGE for the maturation of human plasmacytoid dendritic cells. Eur. J. Immunol. 2005, 35, 2184–2190. [Google Scholar] [CrossRef] [PubMed]
- Moser, B.; Szabolcs, M.; Ankersmit, H.; Lu, Y.; Qu, W.; Weinberg, A.; Herold, K.; Schmidt, A. Blockade of RAGE suppresses alloimmune reactions in vitro and delays allograft rejection in murine heart transplantation. Am. J. Transplant. 2007, 7, 293–302. [Google Scholar] [CrossRef]
- Chavakis, T.; Bierhaus, A.; Al-Fakhri, N.; Schneider, D.; Witte, S.; Linn, T.; Nagashima, M.; Morser, J.; Arnold, B.; Preissner, K.T. The pattern recognition receptor (RAGE) is a counterreceptor for leukocyte integrins: A novel pathway for inflammatory cell recruitment. J. Exp. Med. 2003, 198, 1507–1515. [Google Scholar] [CrossRef]
- Hudson, B.I.; Lippman, M.E. Targeting RAGE signaling in inflammatory disease. Annu. Rev. Med. 2018, 69, 349–364. [Google Scholar] [CrossRef]
- Juranek, J.; Ray, R.; Banach, M.; Rai, V. Receptor for advanced glycation end-products in neurodegenerative diseases. Rev. Neurosci. 2015, 26, 691–698. [Google Scholar] [CrossRef]
- Yan, S.D.; Bierhaus, A.; Nawroth, P.P.; Stern, D.M. RAGE and Alzheimer’s disease: A progression factor for amyloid-β-induced cellular perturbation? J. Alzheimer’s Dis. 2009, 16, 833–843. [Google Scholar] [CrossRef] [Green Version]
- Deane, R.; Du Yan, S.; Submamaryan, R.K.; LaRue, B.; Jovanovic, S.; Hogg, E.; Welch, D.; Manness, L.; Lin, C.; Yu, J. RAGE mediates amyloid-β peptide transport across the blood-brain barrier and accumulation in brain. Nat. Med. 2003, 9, 907. [Google Scholar] [CrossRef]
- Srikanth, V.; Maczurek, A.; Phan, T.; Steele, M.; Westcott, B.; Juskiw, D.; Münch, G. Advanced glycation endproducts and their receptor RAGE in Alzheimer’s disease. Neurobiol. Aging 2011, 32, 763–777. [Google Scholar] [CrossRef]
- Chaney, M.O.; Stine, W.B.; Kokjohn, T.A.; Kuo, Y.-M.; Esh, C.; Rahman, A.; Luehrs, D.C.; Schmidt, A.M.; Stern, D.; Du Yan, S. RAGE and amyloid beta interactions: Atomic force microscopy and molecular modeling. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2005, 1741, 199–205. [Google Scholar] [CrossRef] [Green Version]
- Xu, X.-Y.; Deng, C.-Q.; Wang, J.; Deng, X.-J.; Xiao, Q.; Li, Y.; He, Q.; Fan, W.-H.; Quan, F.-Y.; Zhu, Y.-P. Plasma levels of soluble receptor for advanced glycation end products in Alzheimer’s disease. Int. J. Neurosci. 2017, 127, 454–458. [Google Scholar] [CrossRef] [PubMed]
- Miller, M.C.; Tavares, R.; Johanson, C.E.; Hovanesian, V.; Donahue, J.E.; Gonzalez, L.; Silverberg, G.D.; Stopa, E.G. Hippocampal RAGE immunoreactivity in early and advanced Alzheimer’s disease. Brain Res. 2008, 1230, 273–280. [Google Scholar] [CrossRef] [Green Version]
- Arancio, O.; Zhang, H.P.; Chen, X.; Lin, C.; Trinchese, F.; Puzzo, D.; Liu, S.; Hegde, A.; Yan, S.F.; Stern, A. RAGE potentiates Aβ-induced perturbation of neuronal function in transgenic mice. EMBO J. 2004, 23, 4096–4105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fang, F.; Lue, L.-F.; Yan, S.; Xu, H.; Luddy, J.S.; Chen, D.; Walker, D.G.; Stern, D.M.; Yan, S.; Schmidt, A.M. RAGE-dependent signaling in microglia contributes to neuroinflammation, Aβ accumulation, and impaired learning/memory in a mouse model of Alzheimer’s disease. FASEB J. 2010, 24, 1043–1055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fang, F.; Yu, Q.; Arancio, O.; Chen, D.; Gore, S.S.; Yan, S.S.; Yan, S.F. RAGE mediates Aβ accumulation in a mouse model of Alzheimer’s disease via modulation of β-and γ-secretase activity. Hum. Mol. Genet. 2018, 27, 1002–1014. [Google Scholar] [CrossRef]
- Kang, J.Y.; Lee, J.-O. Structural biology of the Toll-like receptor family. Annu. Rev. Biochem. 2011, 80, 917–941. [Google Scholar] [CrossRef]
- Bode, J.G.; Ehlting, C.; Häussinger, D. The macrophage response towards LPS and its control through the p38MAPK–STAT3 axis. Cell. Signal. 2012, 24, 1185–1194. [Google Scholar] [CrossRef]
- Kurt-Jones, E.A.; Popova, L.; Kwinn, L.; Haynes, L.M.; Jones, L.P.; Tripp, R.A.; Walsh, E.E.; Freeman, M.W.; Golenbock, D.T.; Anderson, L.J. Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytial virus. Nat. Immunol. 2000, 1, 398. [Google Scholar] [CrossRef]
- Vaure, C.; Liu, Y. A comparative review of toll-like receptor 4 expression and functionality in different animal species. Front. Immunol. 2014, 5, 316. [Google Scholar] [CrossRef] [Green Version]
- Le Page, A.; Dupuis, G.; Frost, E.H.; Larbi, A.; Pawelec, G.; Witkowski, J.M.; Fulop, T. Role of the peripheral innate immune system in the development of Alzheimer’s disease. Exp. Gerontol. 2018, 107, 59–66. [Google Scholar] [CrossRef] [PubMed]
- Reed-Geaghan, E.G.; Savage, J.C.; Hise, A.G.; Landreth, G.E. CD14 and toll-like receptors 2 and 4 are required for fibrillar Aβ-stimulated microglial activation. J. Neurosci. 2009, 29, 11982–11992. [Google Scholar] [CrossRef] [PubMed]
- Ajit, D. Toll-like receptors 2 and 4 mediate Abeta (1-42) activation of the innate immune response in a human monocytic cell line. J. Neurochem. 2008, 104, 524–533. [Google Scholar]
- Jin, J.-J.; Kim, H.-D.; Maxwell, J.A.; Li, L.; Fukuchi, K.-I. Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer’s disease. J. Neuroinflamm. 2008, 5, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Long, H.; Zhong, G.; Wang, C.; Zhang, J.; Zhang, Y.; Luo, J.; Shi, S. TREM2 Attenuates Aβ1-42-Mediated Neuroinflammation in BV-2 Cells by Downregulating TLR Signaling. Neurochem. Res. 2019, 44, 1830–1839. [Google Scholar] [CrossRef] [PubMed]
- Ito, H.; Hamerman, J.A. TREM-2, triggering receptor expressed on myeloid cell-2, negatively regulates TLR responses in dendritic cells. Eur. J. Immunol. 2012, 42, 176–185. [Google Scholar] [CrossRef]
- Zhou, J.; Yu, W.; Zhang, M.; Tian, X.; Li, Y.; Lü, Y. Imbalance of Microglial TLR4/TREM2 in LPS-Treated APP/PS1 Transgenic Mice: A Potential Link Between Alzheimer’s Disease and Systemic Inflammation. Neurochem. Res. 2019, 44, 1138–1151. [Google Scholar] [CrossRef]
- Song, M.; Jin, J.; Lim, J.-E.; Kou, J.; Pattanayak, A.; Rehman, J.A.; Kim, H.-D.; Tahara, K.; Lalonde, R.; Fukuchi, K.-I. TLR4 mutation reduces microglial activation, increases Aβ deposits and exacerbates cognitive deficits in a mouse model of Alzheimer’s disease. J. Neuroinflamm. 2011, 8, 92. [Google Scholar] [CrossRef] [Green Version]
- Go, M.; Kou, J.; Lim, J.-E.; Yang, J.; Fukuchi, K.-I. Microglial response to LPS increases in wild-type mice during aging but diminishes in an Alzheimer’s mouse model: Implication of TLR4 signaling in disease progression. Biochem. Biophys. Res. Commun. 2016, 479, 331–337. [Google Scholar] [CrossRef] [Green Version]
- Qin, Y.; Liu, Y.; Hao, W.; Decker, Y.; Tomic, I.; Menger, M.D.; Liu, C.; Fassbender, K. Stimulation of TLR4 Attenuates Alzheimer’s Disease–Related Symptoms and Pathology in Tau-Transgenic Mice. J. Immunol. 2016, 197, 3281–3292. [Google Scholar] [CrossRef]
- Hickman, S.E.; El Khoury, J. TREM2 and the neuroimmunology of Alzheimer’s disease. Biochem. Pharmacol. 2014, 88, 495–498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tricker, E.; Cheng, G. With a little help from my friends: Modulation of phagocytosis through TLR activation. Cell Res. 2008, 18, 711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mackenzie, I.R.; Hao, C.; Munoz, D.G. Role of microglia in senile plaque formation. Neurobiol. Aging 1995, 16, 797–804. [Google Scholar] [CrossRef]
- Sheng, J.G.; Bora, S.H.; Xu, G.; Borchelt, D.R.; Price, D.L.; Koliatsos, V.E. Lipopolysaccharide-induced-neuroinflammation increases intracellular accumulation of amyloid precursor protein and amyloid β peptide in APPswe transgenic mice. Neurobiol. Dis. 2003, 14, 133–145. [Google Scholar] [CrossRef]
- Boutajangout, A.; Wisniewski, T. The innate immune system in Alzheimer’s disease. Int. J. Cell Biol. 2013, 2013, 576383. [Google Scholar] [CrossRef] [Green Version]
- Morgan, A.R.; Touchard, S.; Leckey, C.; O’Hagan, C.; Nevado-Holgado, A.J.; Barkhof, F.; Bertram, L.; Blin, O.; Bos, I.; Dobricic, V. Inflammatory biomarkers in Alzheimer’s disease plasma. Alzheimer’s Dement. 2019, 15, 776–787. [Google Scholar] [CrossRef]
- Bayer, A.J. The role of biomarkers and imaging in the clinical diagnosis of dementia. Age Ageing 2018, 47, 641–643. [Google Scholar] [CrossRef] [Green Version]
- Molinuevo, J.L.; Gispert, J.D.; Dubois, B.; Heneka, M.T.; Lleo, A.; Engelborghs, S.; Pujol, J.; de Souza, L.C.; Alcolea, D.; Jessen, F. The AD-CSF-index discriminates Alzheimer’s disease patients from healthy controls: A validation study. J. Alzheimer’s Dis. 2013, 36, 67–77. [Google Scholar] [CrossRef]
- Ritchie, C.; Smailagic, N.; Noel-Storr, A.H.; Ukoumunne, O.; Ladds, E.C.; Martin, S. CSF tau and the CSF tau/ABeta ratio for the diagnosis of Alzheimer’s disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst. Rev. 2017. [Google Scholar] [CrossRef] [Green Version]
- Zetterberg, H.; Schott, J.M. Biomarkers for Alzheimer’s disease beyond amyloid and tau. Nat. Med. 2019, 25, 201. [Google Scholar] [CrossRef]
- Festoff, B.W.; Sajja, R.K.; van Dreden, P.; Cucullo, L. HMGB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer’s disease. J. Neuroinflamm. 2016, 13, 194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sasaki, N.; Toki, S.; Chowei, H.; Saito, T.; Nakano, N.; Hayashi, Y.; Takeuchi, M.; Makita, Z. Immunohistochemical distribution of the receptor for advanced glycation end products in neurons and astrocytes in Alzheimer’s disease. Brain Res. 2001, 888, 256–262. [Google Scholar] [CrossRef]
- Mruthinti, S.; Buccafusco, J.J.; Hill, W.D.; Waller, J.L.; Jackson, T.W.; Zamrini, E.Y.; Schade, R.F. Autoimmunity in Alzheimer’s disease: Increased levels of circulating IgGs binding Aβ and RAGE peptides. Neurobiol. Aging 2004, 25, 1023–1032. [Google Scholar] [CrossRef] [PubMed]
- Jeynes, B.; Provias, J. Evidence for altered LRP/RAGE expression in Alzheimer lesion pathogenesis. Curr. Alzheimer Res. 2008, 5, 432–437. [Google Scholar] [CrossRef] [PubMed]
- Donahue, J.E.; Flaherty, S.L.; Johanson, C.E.; Duncan, J.A.; Silverberg, G.D.; Miller, M.C.; Tavares, R.; Yang, W.; Wu, Q.; Sabo, E. RAGE, LRP-1, and amyloid-beta protein in Alzheimer’s disease. Acta Neuropathol. 2006, 112, 405–415. [Google Scholar] [CrossRef]
- Emanuele, E.; D’Angelo, A.; Tomaino, C.; Binetti, G.; Ghidoni, R.; Politi, P.; Bernardi, L.; Maletta, R.; Bruni, A.C.; Geroldi, D. Circulating levels of soluble receptor for advanced glycation end products in Alzheimer disease and vascular dementia. Arch. Neurol. 2005, 62, 1734–1736. [Google Scholar] [CrossRef]
- Zhang, W.; Wang, L.-Z.; Yu, J.-T.; Chi, Z.-F.; Tan, L. Increased expressions of TLR2 and TLR4 on peripheral blood mononuclear cells from patients with Alzheimer’s disease. J. Neurol. Sci. 2012, 315, 67–71. [Google Scholar] [CrossRef]
- Kilic, U.; Elibol, B.; Uysal, O.; Kilic, E.; Yulug, B.; Sakul, A.S.; Yildiz, G.B. Specific alterations in the circulating levels of the SIRT1, TLR4, and IL7 proteins in patients with dementia. Exp. Gerontol. 2018, 111, 203–209. [Google Scholar] [CrossRef]
- Balistreri, C.; Grimaldi, M.; Chiappelli, M.; Licastro, F.; Castiglia, L.; Listì, F.; Vasto, S.; Lio, D.; Caruso, C.; Candore, G. Association between the polymorphisms of TLR4 and CD14 genes and Alzheimer’s disease. Curr. Pharm. Des. 2008, 14, 2672–2677. [Google Scholar] [CrossRef]
- Musumeci, D.; Roviello, G.N.; Montesarchio, D. An overview on HMGB1 inhibitors as potential therapeutic agents in HMGB1-related pathologies. Pharmacol. Ther. 2014, 141, 347–357. [Google Scholar] [CrossRef]
- Nishibori, M.; Mori, S.; Takahashi, H.K. Anti-HMGB1 monoclonal antibody therapy for a wide range of CNS and PNS diseases. J. Pharmacol. Sci. 2019, 140, 94–101. [Google Scholar] [CrossRef] [PubMed]
- Paudel, Y.N.; Angelopoulou, E.; Semple, B.; Piperi, C.; Othman, I.; Shaikh, M.F. Potential neuroprotective effect of the HMGB1 inhibitor Glycyrrhizin in neurological disorders. ACS Chem. Neurosci. 2020. [Google Scholar] [CrossRef] [PubMed]
- Okuma, Y.; Liu, K.; Wake, H.; Liu, R.; Nishimura, Y.; Hui, Z.; Teshigawara, K.; Haruma, J.; Yamamoto, Y.; Yamamoto, H. Glycyrrhizin inhibits traumatic brain injury by reducing HMGB1–RAGE interaction. Neuropharmacology 2014, 85, 18–26. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.J.; Wang, L.; Zhang, B.; Gao, F.; Yang, C.M. Glycyrrhizin, an HMGB1 inhibitor, exhibits neuroprotective effects in rats after lithium-pilocarpine-induced status epilepticus. J. Pharm. Pharmacol. 2019, 71, 390–399. [Google Scholar] [CrossRef] [PubMed]
- Webster, K.M.; Shultz, S.R.; Ozturk, E.; Dill, L.K.; Sun, M.; Casillas-Espinosa, P.; Jones, N.C.; Crack, P.J.; O’Brien, T.J.; Semple, B.D. Targeting high-mobility group box protein 1 (HMGB1) in pediatric traumatic brain injury: Chronic neuroinflammatory, behavioral, and epileptogenic consequences. Exp. Neurol. 2019, 320, 112979. [Google Scholar] [CrossRef]
- Santoro, M.; Maetzler, W.; Stathakos, P.; Martin, H.L.; Hobert, M.A.; Rattay, T.W.; Gasser, T.; Forrester, J.V.; Berg, D.; Tracey, K.J. In-vivo evidence that high mobility group box 1 exerts deleterious effects in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model and Parkinson’s disease which can be attenuated by glycyrrhizin. Neurobiol. Dis. 2016, 91, 59–68. [Google Scholar] [CrossRef] [Green Version]
- Song, J.-H.; Lee, J.-W.; Shim, B.; Lee, C.-Y.; Choi, S.; Kang, C.; Sohn, N.-W.; Shin, J.-W. Glycyrrhizin alleviates neuroinflammation and memory deficit induced by systemic lipopolysaccharide treatment in mice. Molecules 2013, 18, 15788–15803. [Google Scholar] [CrossRef] [Green Version]
- Liu, W.; Huang, S.; Li, Y.; Zhang, K.; Zheng, X. Suppressive effect of glycyrrhizic acid against lipopolysaccharide-induced neuroinflammation and cognitive impairment in C57 mice via toll-like receptor 4 signaling pathway. Food Nutr. Res. 2019, 63. [Google Scholar] [CrossRef] [Green Version]
- Chen, X.; Hua, H.-P.; Liang, L.; Liu, L.-J. The oral pretreatment of glycyrrhizin prevents surgery-induced cognitive impairment in aged mice by reducing neuroinflammation and Alzheimer’s-related pathology via HMGB1 inhibition. J. Mol. Neurosci. 2017, 63, 385–395. [Google Scholar]
- Yang, H.; Wang, H.; Chavan, S.S.; Andersson, U. High mobility group box protein 1 (HMGB1): The prototypical endogenous danger molecule. Mol. Med. 2015, 21, S6–S12. [Google Scholar] [CrossRef]
- Balducci, C.; Forloni, G. Novel targets in Alzheimer’s disease: A special focus on microglia. Pharmacol. Res. 2018, 130, 402–413. [Google Scholar] [CrossRef] [PubMed]
- Cirillo, C.; Capoccia, E.; Iuvone, T.; Cuomo, R.; Sarnelli, G.; Steardo, L.; Esposito, G. S100B inhibitor pentamidine attenuates reactive gliosis and reduces neuronal loss in a mouse model of Alzheimer’s disease. Biomed. Res. Int. 2015, 2015, 508342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cui, L.; Cai, Y.; Cheng, W.; Liu, G.; Zhao, J.; Cao, H.; Tao, H.; Wang, Y.; Yin, M.; Liu, T. A novel, multi-target natural drug candidate, matrine, improves cognitive deficits in Alzheimer’s disease transgenic mice by inhibiting Aβ aggregation and blocking the RAGE/Aβ axis. Mol. Neurobiol. 2017, 54, 1939–1952. [Google Scholar] [CrossRef] [PubMed]
- Deane, R.; Singh, I.; Sagare, A.P.; Bell, R.D.; Ross, N.T.; LaRue, B.; Love, R.; Perry, S.; Paquette, N.; Deane, R.J. A multimodal RAGE-specific inhibitor reduces amyloid β–mediated brain disorder in a mouse model of Alzheimer disease. J. Clin. Investig. 2012, 122, 1377–1392. [Google Scholar] [CrossRef] [Green Version]
- Kostura, M.J.; Kindy, M.S.; Burstein, A.; Valcarce, C.; Polisetti, D.; Andrews, R.; Mjalli, A.M. Efficacy of Rage Antagonists in Murine Model of Alzheimer’s Disease. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2014, 10, P638–P639. [Google Scholar] [CrossRef]
- Hu, Q.; Yu, B.; Chen, Q.; Wang, Y.; Ling, Y.; Sun, S.; Shi, Y.; Zhou, C. Effect of Linguizhugan decoction on neuroinflammation and expression disorder of the amyloid β-related transporters RAGE and LRP-1 in a rat model of Alzheimer’s disease. Mol. Med. Rep. 2018, 17, 827–834. [Google Scholar] [CrossRef] [Green Version]
- Hong, Y.; An, Z. Hesperidin attenuates learning and memory deficits in APP/PS1 mice through activation of Akt/Nrf2 signaling and inhibition of RAGE/NF-κB signaling. Arch. Pharm. Res. 2018, 41, 655–663. [Google Scholar] [CrossRef]
- Yousefi, N.; Sotoodehnejadnematalahi, F.; Heshmati-Fakhr, N.; Sayyah, M.; Hoseini, M.; Ghassemi, S.; Aliakbari, S.; Pourbadie, H.G. Prestimulation of Microglia Through TLR4 Pathway Promotes Interferon Beta Expression in a Rat Model of Alzheimer’s Disease. J. Mol. Neurosci. 2019, 67, 495–503. [Google Scholar] [CrossRef]
- Michaud, J.-P.; Hallé, M.; Lampron, A.; Thériault, P.; Préfontaine, P.; Filali, M.; Tribout-Jover, P.; Lanteigne, A.-M.; Jodoin, R.; Cluff, C. Toll-like receptor 4 stimulation with the detoxified ligand monophosphoryl lipid A improves Alzheimer’s disease-related pathology. Proc. Natl. Acad. Sci. USA 2013, 110, 1941–1946. [Google Scholar] [CrossRef] [Green Version]
- Shi, S.; Liang, D.; Chen, Y.; Xie, Y.; Wang, Y.; Wang, L.; Wang, Z.; Qiao, Z. Gx-50 reduces β-amyloid-induced TNF-α, IL-1β, NO, and PGE2 expression and inhibits NF-κB signaling in a mouse model of Alzheimer’s disease. Eur. J. Immunol. 2016, 46, 665–676. [Google Scholar] [CrossRef]
- Jin, X.; Liu, M.Y.; Zhang, D.F.; Zhong, X.; Du, K.; Qian, P.; Yao, W.F.; Gao, H.; Wei, M.J. Baicalin mitigates cognitive impairment and protects neurons from microglia-mediated neuroinflammation via suppressing NLRP 3 inflammasomes and TLR 4/NF-κB signaling pathway. CNS Neurosci. Ther. 2019, 25, 575–590. [Google Scholar] [CrossRef] [PubMed]
- Ikram, M.; Muhammad, T.; Rehman, S.U.; Khan, A.; Jo, M.G.; Ali, T.; Kim, M.O. Hesperetin confers neuroprotection by regulating Nrf2/TLR4/NF-κB signaling in an Aβ mouse model. Mol. Neurobiol. 2019, 6, 6293–6309. [Google Scholar] [CrossRef] [PubMed]
- Guan, F.; Zhou, X.; Li, P.; Wang, Y.; Liu, M.; Li, F.; Cui, Y.; Huang, T.; Yao, M.; Zhang, Y. MG53 attenuates lipopolysaccharide-induced neurotoxicity and neuroinflammation via inhibiting TLR4/NF-κB pathway in vitro and in vivo. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2019, 95, 109684. [Google Scholar] [CrossRef] [PubMed]
- Capiralla, H.; Vingtdeux, V.; Zhao, H.; Sankowski, R.; Al-Abed, Y.; Davies, P.; Marambaud, P. Resveratrol mitigates lipopolysaccharide-and Aβ-mediated microglial inflammation by inhibiting the TLR4/NF-κB/STAT signaling cascade. J. Neurochem. 2012, 120, 461–472. [Google Scholar] [CrossRef] [Green Version]
- Pourbadie, H.G.; Sayyah, M.; Khoshkholgh-Sima, B.; Choopani, S.; Nategh, M.; Motamedi, F.; Shokrgozar, M.A. Early minor stimulation of microglial TLR2 and TLR4 receptors attenuates Alzheimer’s disease–related cognitive deficit in rats: Behavioral, molecular, and electrophysiological evidence. Neurobiol. Aging 2018, 70, 203–216. [Google Scholar] [CrossRef]
- Querfurth, H.W.; LaFerla, F.M. Mechanisms of disease. N. Engl. J. Med. 2010, 362, 329–344. [Google Scholar] [CrossRef] [Green Version]
- Lahiri, D.K. Lessons from Alzheimer’s Disease (AD) Clinical Trials: Instead of “A-Drug”, AD-D prevention to Avert AD. Curr. Alzheimer Res. 2019, 16, 279–280. [Google Scholar] [CrossRef]
- McGeer, P.L.; McGeer, E.G. Inflammation, autotoxicity and Alzheimer disease. Neurobiol. Aging 2001, 22, 799–809. [Google Scholar] [CrossRef]
- McGeer, P.L.; McGeer, E.G. Local neuroinflammation and the progression of Alzheimer’s disease. J. Neurovirol. 2002, 8, 529–538. [Google Scholar] [CrossRef] [Green Version]
- Hampel, H.; Mesulam, M.-M.; Cuello, A.C.; Farlow, M.R.; Giacobini, E.; Grossberg, G.T.; Khachaturian, A.S.; Vergallo, A.; Cavedo, E.; Snyder, P.J. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 2018, 141, 1917–1933. [Google Scholar] [CrossRef]
- Papaevgeniou, N.; Sakellari, M.; Jha, S.; Tavernarakis, N.; Holmberg, C.I.; Gonos, E.S.; Chondrogianni, N. 18α-Glycyrrhetinic acid proteasome activator decelerates aging and Alzheimer’s disease progression in caenorhabditis elegans and neuronal cultures. Antioxid. Redox Signal. 2016, 25, 855–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oh, S.; Son, M.; Choi, J.; Lee, S.; Byun, K. sRAGE prolonged stem cell survival and suppressed RAGE-related inflammatory cell and T lymphocyte accumulations in an Alzheimer’s disease model. Biochem. Biophys. Res. Commun. 2018, 495, 807–813. [Google Scholar] [CrossRef] [PubMed]
- Criscuolo, C.; Fontebasso, V.; Middei, S.; Stazi, M.; Ammassari-Teule, M.; Yan, S.S.; Origlia, N. Entorhinal Cortex dysfunction can be rescued by inhibition of microglial RAGE in an Alzheimer’s disease mouse model. Sci. Rep. 2017, 7, 42370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sabbagh, M.N.; Agro, A.; Bell, J.; Aisen, P.S.; Schweizer, E.; Galasko, D. PF-04494700, an oral inhibitor of receptor for advanced glycation end products (RAGE), in Alzheimer’s disease. Alzheimer Dis. Assoc. Disord. 2011, 25, 206. [Google Scholar] [CrossRef]
- Galasko, D.; Bell, J.; Mancuso, J.Y.; Kupiec, J.W.; Sabbagh, M.N.; van Dyck, C.; Thomas, R.G.; Aisen, P.S. Clinical trial of an inhibitor of RAGE-Aβ interactions in Alzheimer disease. Neurology 2014, 82, 1536–1542. [Google Scholar] [CrossRef]
S.N. | Interventions | Model | Treatment Schedule | Observations | References |
---|---|---|---|---|---|
1 | HMGB1 short hairpin RNA (shRNA) | Aβ25–35-induced (25 μmol/L) neuroinflammation in hippocampal neuron cultures | Pre-treated for 24 h |
| [18] |
2 | Anti-HMGB1 mAb (1 mg/kg, S.C. injection) (1 injection/week) | 5xFAD transgenic mice overexpressing the mutant human APP | Administered for 1–6 months or 3–6 months |
| [37] |
3 | Glycyrrhizic acid (GA) (50 and 100 mg/kg, I.P.) | LPS (250 μg/kg) -induced neuroinflammation and cognitive impairment in the C57 mice (4–5 weeks old) | Once daily for 1 week |
| [109] |
4 | Glycyrrhizin (GL) (16.8 mg/kg, I.P.) | p35-/-/Tg2576 mice (p35 deletion in Tg2576 mice) | Every alternate day for 1 week |
| [42] |
5 | GL (30 mg/kg, orally) | Surgery induced cognitive decline in C57BL/6 mice | Once daily for 3 days pre-operatively |
| [110] |
6 | GL (30 and 50 mg/kg, orally) | LPS (3 mg/kg, I.P.)-induced neuroinflammation and cognitive impairment in the C57BL/6 mice | Once a day for 3 days prior to LPS injection |
| [108] |
7 | 18α-glycyrrhetinic acid (GA) (20 μg/mL) | AD nematode models (WT Caenorhabditis elegans) | - |
| [132] |
S.N. | Interventions | Model | Treatment Schedule | Observations | References |
---|---|---|---|---|---|
1 | TTP488 (RAGE antagonist) | Transgenic mice overexpressing APP/PS1 | Oral treatment with TTP488 starting at 12 months of age |
| [116] |
2 | sRAGE-mesenchymal stem cells (MSCs) | Aβ1–42 (5 μL; 200 μM) peptides induced AD model in SD rats | sRAGE-MSCs is transplanted for 4 months |
| [133] |
3 | Hesperidin (20, 40 and 80 mg/kg) | AD like pathology in APP/PS1 mice | Treatment for 90 days |
| [118] |
4 | Linguizhugan (2.4, 4.8, or 1.2 g/kg) | Aβ-induced (10 µg) AD model in SD rats | Linguizhugan treatment for 25 days |
| [117] |
5 | RAGE specific inhibitor (FPS-ZM1, 1 mg/kg/d, I.P.) | Male APPsw/0 mice (15 to 17 months old) overexpressing human APP | For 2 months starting at 8 or 15 months of age |
| [115] |
6 | DNMSR (dominant-negative form of RAGE lacking RAGE signaling targeted to microglia) | AD mouse model carrying human mutation of APP (mhAPP) expressing human Aβ | - |
| [134] |
7 | Pentamidine (0.05 μg/mL) (S100β inhibitor) | Aβ-induced (10 μg/mL) AD in C57BL/6J mice | Per day |
| [113] |
8 | Matrine (10 and 50 μM) | APP/PS1 transgenic mice model |
| [114] | |
9 | PF-04494700 (10 or 20 mg) (oral RAGE inhibitor) | Subjects with mild-to-moderate dementia of AD type meeting NINCDS-ADRDA criteria | 10 week randomized, double-blind, placebo-controlled trial with 2 doses of PF-04494700 (10 mg, after a 6-day loading dose of 30 mg/d); and PF-04494700 (20 mg, after a loading dose of 60 mg/d); |
| [135] |
10 | PF-04494700 (RAGE inhibitor) | Double-blind, placebo-controlled trial at 40 several centre, subjects assessed with AD assessment scale-cognitive-subscale | Treatment for 18 months using 2 doses of PF-04494700 60 mg/day for 6 days, then 20 mg daily and 15 mg/day for 6 days, then 5 mg daily |
| [136] |
S.N. | Interventions | Model | Treatment Schedule | Observations | References |
---|---|---|---|---|---|
1 | Monophosphoryl lipid A, LPS-derived TLR4 agonist (MPL, 50 μg, I.P.) | AD like pathology in APPswe/PS1 mice | Administered once a week for 12 weeks |
| [120] |
2 | MPL (1 μg/5 μL/rat) | Aβ1–42-induced (0.075 μg/hour, I.C.V. for 2 weeks) AD related cognitive decline in male Wistar rats | MPL treatment for 24 days (8 injections alternate 3 days) |
| [126] |
3 | Gx-50 (1 mg/kg) | APP transgenic model of AD | Gx-50 administered daily for 2 months at 5 months of age |
| [121] |
4 | Hesperetin (50 mg) | Aβ1–42-induced (5 μL/5min/mouse) AD model in (C57BL/6N, WT) mouse | Hesperetin (50 mg) treatment for 6 weeks |
| [123] |
5 | MG53 (2 mg/kg) | LPS-induced (0.25 mg/kg, I.P. once a day for 1 week) neuroinflammation and neurotoxicity (in vitro and in vivo) in male C57BL/6 mice. | MG53 (once a day for 2 weeks) was intravenously administrated through tail vein one week before LPS injection. |
| [124] |
6 | Resveratrol | In vitro study (RAW 264.7 cells stimulated with 10 ng/mL LPS, BV-2 cells 100 ng/mL LPS, and Ba/F3 cells with 50 ng/mL LPS) In vivo study in Aβ APP/PS1 transgenic mice | Orally administered for 15 weeks |
| [125] |
7 | Baicalin (BAI) (103 mg/kg administered intragastrically) | APP/PS1 transgenic mice | Treated with BAI once a day for 33 days |
| [122] |
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Paudel, Y.N.; Angelopoulou, E.; Piperi, C.; Othman, I.; Aamir, K.; Shaikh, M.F. Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s Disease (AD): From Risk Factors to Therapeutic Targeting. Cells 2020, 9, 383. https://doi.org/10.3390/cells9020383
Paudel YN, Angelopoulou E, Piperi C, Othman I, Aamir K, Shaikh MF. Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s Disease (AD): From Risk Factors to Therapeutic Targeting. Cells. 2020; 9(2):383. https://doi.org/10.3390/cells9020383
Chicago/Turabian StylePaudel, Yam Nath, Efthalia Angelopoulou, Christina Piperi, Iekhsan Othman, Khurram Aamir, and Mohd. Farooq Shaikh. 2020. "Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s Disease (AD): From Risk Factors to Therapeutic Targeting" Cells 9, no. 2: 383. https://doi.org/10.3390/cells9020383
APA StylePaudel, Y. N., Angelopoulou, E., Piperi, C., Othman, I., Aamir, K., & Shaikh, M. F. (2020). Impact of HMGB1, RAGE, and TLR4 in Alzheimer’s Disease (AD): From Risk Factors to Therapeutic Targeting. Cells, 9(2), 383. https://doi.org/10.3390/cells9020383