Comparison of Oleocanthal-Low EVOO and Oleocanthal against Amyloid-β and Related Pathology in a Mouse Model of Alzheimer’s Disease
Abstract
:1. Introduction
2. Results
2.1. EVOO and OC Treatments Reduced Aβ Burden in 5xFAD Mouse Brains
2.2. EVOO and OC Enhanced BBB Function in 5xFAD Mice Brains
2.3. EVOO and OC Reduced the Amyloidogenic and Enhanced the Non-Amyloidogenic Pathways in 5xFAD Mouse Brains
2.4. EVOO and OC Increased the Expression of Synaptic Markers in 5xFAD Mouse Brains
2.5. EVOO and OC Reduced Neuroinflammation in 5xFAD Mouse Brains
2.6. OC Reduced RAGE and HMGB1 Expressions in 5xFAD Mouse Brains
2.7. EVOO- and OC-Reduced Neuroinflammation Is Mediated by NF-κB Pathway in 5xFAD Mouse Brains
3. Discussion
4. Materials and Methods
4.1. Materials
4.2. Animals
4.3. Western Blot Analysis
4.4. Immunofluorescence Staining and Analysis
4.5. Enzyme-Linked Immunosorbent Assay (ELISA)
4.6. Statistical Analysis
Author Contributions
Funding
Institutional Review Board Statement
Data Availability Statement
Conflicts of Interest
Sample Availability
References
- Breijyeh, Z.; Karaman, R. Comprehensive review on Alzheimer’s disease: Causes and treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef] [PubMed]
- Mebane-Sims, I. Alzheimer’s Association, 2018 Alzheimer’s disease facts and figures. Alzheimer’s Dement 2018, 14, 367–429. [Google Scholar]
- Adav, S.S.; Sze, S.K. Insight of brain degenerative protein modifications in the pathology of neurodegeneration and dementia by proteomic profiling. Mol. Brain 2016, 9, 1–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murpy, M.; LeVine III, H. Alzheimer’s disease and the β-amyloid peptide. J. Alzheimer’s Dis 2010, 19, 311–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alzheimer’s Association. 2019 Alzheimer’s disease facts and figures. Alzheimer’s Dement 2019, 15, 321–387. [Google Scholar] [CrossRef]
- Long, J.M.; Holtzman, D.M. Alzheimer disease: An update on pathobiology and treatment strategies. Cell 2019, 179, 312–339. [Google Scholar] [CrossRef]
- DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 1–18. [Google Scholar] [CrossRef] [Green Version]
- O’brien, R.J.; Wong, P.C. Amyloid precursor protein processing and Alzheimer’s disease. Annu. Rev. Neurosci. 2011, 34, 185–204. [Google Scholar] [CrossRef] [Green Version]
- Chen, G.-f.; Xu, T.-h.; Yan, Y.; Zhou, Y.-r.; Jiang, Y.; Melcher, K.; Xu, H.E. Amyloid beta: Structure, biology and structure-based therapeutic development. Acta Pharmacol. Sin. 2017, 38, 1205–1235. [Google Scholar] [CrossRef] [Green Version]
- Tillement, L.; Lecanu, L.; Papadopoulos, V. Alzheimer’s disease: Effects of β-amyloid on mitochondria. Mitochondrion 2011, 11, 13–21. [Google Scholar] [CrossRef]
- Holtzman, D.M.; Morris, J.C.; Goate, A.M. Alzheimer’s disease: The challenge of the second century. Sci. Transl. Med. 2011, 3, 77sr71. [Google Scholar] [CrossRef]
- Kinney, J.W.; Bemiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, 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]
- Zotova, E.; Nicoll, J.A.; Kalaria, R.; Holmes, C.; Boche, D. Inflammation in Alzheimer’s disease: Relevance to pathogenesis and therapy. Alzheimer’s Res. Ther. 2010, 2, 1. [Google Scholar] [CrossRef]
- Onyango, I.G.; Jauregui, G.V.; Čarná, M.; Bennett, J.P., Jr.; Stokin, G.B. Neuroinflammation in Alzheimer’s disease. Biomedicines 2021, 9, 524. [Google Scholar] [CrossRef]
- Yoon, S.-S.; Jo, S.A. Mechanisms of amyloid-β peptide clearance: Potential therapeutic targets for Alzheimer’s disease. Biomol. Ther. 2012, 20, 245. [Google Scholar] [CrossRef] [Green Version]
- Zhang, G.; Wang, Z.; Hu, H.; Zhao, M.; Sun, L. Microglia in Alzheimer’s Disease: A Target for Therapeutic Intervention. Front. Cell. Neurosci. 2021, 15, 749587. [Google Scholar] [CrossRef]
- Uddin, M.S.; Kabir, M.T.; Al Mamun, A.; Barreto, G.E.; Rashid, M.; Perveen, A.; Ashraf, G.M. Pharmacological approaches to mitigate neuroinflammation in Alzheimer’s disease. Int. Immunopharmacol. 2020, 84, 106479. [Google Scholar] [CrossRef]
- Saha, R.N.; Pahan, K. Regulation of inducible nitric oxide synthase gene in glial cells. Antioxid. Redox Signal. 2006, 8, 929–947. [Google Scholar] [CrossRef]
- Morgan, M.J.; Liu, Z.-g. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [Green Version]
- Hensley, K. Neuroinflammation in Alzheimer’s disease: Mechanisms, pathologic consequences, and potential for therapeutic manipulation. J. Alzheimer’s Dis. 2010, 21, 1–14. [Google Scholar] [CrossRef] [Green Version]
- Abbott, N.J.; Patabendige, A.A.; Dolman, D.E.; Yusof, S.R.; Begley, D.J. Structure and function of the blood–brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef] [PubMed]
- Wisniewski, H.M.; Vorbrodt, A.W.; Wegiel, J. Amyloid Angiopathy and Blood–Brain Barrier Changes in Alzheimer’s Disease a, b. Ann. N. Y. Acad. Sci. 1997, 826, 161–172. [Google Scholar] [CrossRef] [PubMed]
- Cirrito, J.R.; Deane, R.; Fagan, A.M.; Spinner, M.L.; Parsadanian, M.; Finn, M.B.; Jiang, H.; Prior, J.L.; Sagare, A.; Bales, K.R. P-glycoprotein deficiency at the blood-brain barrier increases amyloid-β deposition in an Alzheimer disease mouse model. J. Clin. Investig. 2005, 115, 3285–3290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qosa, H.; Abuasal, B.S.; Romero, I.A.; Weksler, B.; Couraud, P.-O.; Keller, J.N.; Kaddoumi, A. Differences in amyloid-β clearance across mouse and human blood–brain barrier models: Kinetic analysis and mechanistic modeling. Neuropharmacology 2014, 79, 668–678. [Google Scholar] [CrossRef] [Green Version]
- Scarmeas, N.; Stern, Y.; Tang, M.X.; Mayeux, R.; Luchsinger, J.A. Mediterranean diet and risk for Alzheimer’s disease. Ann. Neurol. Off. J. Am. Neurol. Assoc. Child Neurol. Soc. 2006, 59, 912–921. [Google Scholar] [CrossRef] [Green Version]
- Gu, Y.; Nieves, J.W.; Stern, Y.; Luchsinger, J.A.; Scarmeas, N. Food combination and Alzheimer disease risk: A protective diet. Arch. Neurol. 2010, 67, 699–706. [Google Scholar] [CrossRef] [Green Version]
- Roman, G.C.; Jackson, R.; Reis, J.; Román, A.; Toledo, J.; Toledo, E. Extra-virgin olive oil for potential prevention of Alzheimer disease. Rev. Neurol. 2019, 175, 705–723. [Google Scholar] [CrossRef]
- Abuznait, A.H.; Qosa, H.; Busnena, B.A.; El Sayed, K.A.; Kaddoumi, A. Olive-oil-derived oleocanthal enhances β-amyloid clearance as a potential neuroprotective mechanism against Alzheimer’s disease: In vitro and in vivo studies. ACS Chem. Neurosci. 2013, 4, 973–982. [Google Scholar] [CrossRef] [Green Version]
- Qosa, H.; Mohamed, L.A.; Batarseh, Y.S.; Alqahtani, S.; Ibrahim, B.; LeVine III, H.; Keller, J.N.; Kaddoumi, A. Extra-virgin olive oil attenuates amyloid-β and tau pathologies in the brains of TgSwDI mice. J. Nutr. Biochem. 2015, 26, 1479–1490. [Google Scholar] [CrossRef] [Green Version]
- Al Rihani, S.B.; Darakjian, L.I.; Kaddoumi, A. Oleocanthal-rich extra-virgin olive oil restores the blood–brain barrier function through NLRP3 inflammasome inhibition simultaneously with autophagy induction in TgSwDI mice. ACS Chem. Neurosci. 2019, 10, 3543–3554. [Google Scholar] [CrossRef]
- Batarseh, Y.S.; Kaddoumi, A. Oleocanthal-rich extra-virgin olive oil enhances donepezil effect by reducing amyloid-β load and related toxicity in a mouse model of Alzheimer’s disease. J. Nutr. Biochem. 2018, 55, 113–123. [Google Scholar] [CrossRef]
- Kaddoumi, A.; Denney, T.S., Jr.; Deshpande, G.; Robinson, J.L.; Beyers, R.J.; Redden, D.T.; Praticò, D.; Kyriakides, T.C.; Lu, B.; Kirby, A.N. Extra-Virgin Olive Oil Enhances the Blood–Brain Barrier Function in Mild Cognitive Impairment: A Randomized Controlled Trial. Nutrients 2022, 14, 5102. [Google Scholar] [CrossRef]
- Tsolaki, M.; Lazarou, E.; Kozori, M.; Petridou, N.; Tabakis, I.; Lazarou, I.; Karakota, M.; Saoulidis, I.; Melliou, E.; Magiatis, P. A randomized clinical trial of greek high phenolic early harvest extra virgin olive oil in mild cognitive impairment: The MICOIL pilot study. J. Alzheimer’s Dis. 2020, 78, 801–817. [Google Scholar] [CrossRef]
- Tripoli, E.; Giammanco, M.; Tabacchi, G.; Di Majo, D.; Giammanco, S.; La Guardia, M. The phenolic compounds of olive oil: Structure, biological activity and beneficial effects on human health. Nutr. Res. Rev. 2005, 18, 98–112. [Google Scholar] [CrossRef]
- Reboredo-Rodríguez, P.; Varela-López, A.; Forbes-Hernández, T.Y.; Gasparrini, M.; Afrin, S.; Cianciosi, D.; Zhang, J.; Manna, P.P.; Bompadre, S.; Quiles, J.L.; et al. Phenolic Compounds Isolated from Olive Oil as Nutraceutical Tools for the Prevention and Management of Cancer and Cardiovascular Diseases. Int. J. Mol. Sci. 2018, 19, 2305. [Google Scholar] [CrossRef] [Green Version]
- Rodríguez-Morató, J.; Xicota, L.; Fitó, M.; Farré, M.; Dierssen, M.; De la Torre, R. Potential role of olive oil phenolic compounds in the prevention of neurodegenerative diseases. Molecules 2015, 20, 4655–4680. [Google Scholar] [CrossRef] [Green Version]
- Beauchamp, G.K.; Keast, R.S.; Morel, D.; Lin, J.; Pika, J.; Han, Q.; Lee, C.-H.; Smith, A.B.; Breslin, P.A. Ibuprofen-like activity in extra-virgin olive oil. Nature 2005, 437, 45–46. [Google Scholar] [CrossRef]
- Qosa, H.; Batarseh, Y.S.; Mohyeldin, M.M.; El Sayed, K.A.; Keller, J.N.; Kaddoumi, A. Oleocanthal enhances amyloid-β clearance from the brains of TgSwDI mice and in vitro across a human blood-brain barrier model. ACS Chem. Neurosci. 2015, 6, 1849–1859. [Google Scholar] [CrossRef] [Green Version]
- Romero-Márquez, J.M.; Navarro-Hortal, M.D.; Jiménez-Trigo, V.; Muñoz-Ollero, P.; Forbes-Hernández, T.Y.; Esteban-Muñoz, A.; Giampieri, F.; Delgado Noya, I.; Bullón, P.; Vera-Ramírez, L. An Olive-Derived Extract 20% Rich in Hydroxytyrosol Prevents β-Amyloid Aggregation and Oxidative Stress, Two Features of Alzheimer Disease, via SKN-1/NRF2 and HSP-16.2 in Caenorhabditis elegans. Antioxidants 2022, 11, 629. [Google Scholar] [CrossRef]
- Leri, M.; Bertolini, A.; Stefani, M.; Bucciantini, M. EVOO Polyphenols Relieve Synergistically Autophagy Dysregulation in a Cellular Model of Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 7225. [Google Scholar] [CrossRef]
- Delgado, A.; Cholevas, C.; Theoharides, T.C. Neuroinflammation in Alzheimer’s disease and beneficial action of luteolin. Biofactors 2021, 47, 207–217. [Google Scholar] [CrossRef]
- Qin, C.; Hu, S.; Zhang, S.; Zhao, D.; Wang, Y.; Li, H.; Peng, Y.; Shi, L.; Xu, X.; Wang, C. Hydroxytyrosol Acetate Improves the Cognitive Function of APP/PS1 Transgenic Mice in ERβ-dependent Manner. Mol. Nutr. Food Res. 2021, 65, 2000797. [Google Scholar] [CrossRef] [PubMed]
- Grewal, R.; Reutzel, M.; Dilberger, B.; Hein, H.; Zotzel, J.; Marx, S.; Tretzel, J.; Sarafeddinov, A.; Fuchs, C.; Eckert, G.P. Purified oleocanthal and ligstroside protect against mitochondrial dysfunction in models of early Alzheimer’s disease and brain ageing. Exp. Neurol. 2020, 328, 113248. [Google Scholar] [CrossRef] [PubMed]
- Leri, M.; Natalello, A.; Bruzzone, E.; Stefani, M.; Bucciantini, M. Oleuropein aglycone and hydroxytyrosol interfere differently with toxic Aβ1-42 aggregation. Food Chem. Toxicol. 2019, 129, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Nardiello, P.; Pantano, D.; Lapucci, A.; Stefani, M.; Casamenti, F. Diet supplementation with hydroxytyrosol ameliorates brain pathology and restores cognitive functions in a mouse model of amyloid-β deposition. J. Alzheimer’s Dis. 2018, 63, 1161–1172. [Google Scholar] [CrossRef]
- Abdallah, I.M.; Al-Shami, K.M.; Yang, E.; Wang, J.; Guillaume, C.; Kaddoumi, A. Oleuropein-Rich Olive Leaf Extract Attenuates Neuroinflammation in the Alzheimer’s Disease Mouse Model. ACS Chem. Neurosci. 2022, 13, 1002–1013. [Google Scholar] [CrossRef]
- Casamenti, F.; Grossi, C.; Rigacci, S.; Pantano, D.; Luccarini, I.; Stefani, M. Oleuropein aglycone: A possible drug against degenerative conditions. In vivo evidence of its effectiveness against Alzheimer’s disease. J. Alzheimer’s Dis. 2015, 45, 679–688. [Google Scholar] [CrossRef]
- Castejón, M.L.; Montoya, T.; Ortega-Vidal, J.; Altarejos, J.; Alarcón-de-la-Lastra, C. Ligstroside aglycon, an extra virgin olive oil secoiridoid, prevents inflammation by regulation of MAPKs, JAK/STAT, NF-κB, Nrf2/HO-1, and NLRP3 inflammasome signaling pathways in LPS-stimulated murine peritoneal macrophages. Food Funct. 2022, 13, 10200–10209. [Google Scholar] [CrossRef]
- Citron, M. Alzheimer’s disease: Strategies for disease modification. Nat. Rev. Drug Discov. 2010, 9, 387–398. [Google Scholar] [CrossRef]
- Féart, C.; Samieri, C.; Barberger-Gateau, P. Mediterranean diet and cognitive function in older adults. Curr. Opin. Clin. Nutr. Metab. Care 2010, 13, 14–18. [Google Scholar] [CrossRef] [Green Version]
- Hardman, R.J.; Kennedy, G.; Macpherson, H.; Scholey, A.B.; Pipingas, A. Adherence to a Mediterranean-style diet and effects on cognition in adults: A qualitative evaluation and systematic review of longitudinal and prospective trials. Front. Nutr. 2016, 3, 22. [Google Scholar] [CrossRef]
- El Riachy, M.; Priego-Capote, F.; León, L.; Rallo, L.; Luque de Castro, M.D. Hydrophilic antioxidants of virgin olive oil. Part 1: Hydrophilic phenols: A key factor for virgin olive oil quality. Eur. J. Lipid Sci. Technol. 2011, 113, 678–691. [Google Scholar] [CrossRef]
- Lauretti, E.; Iuliano, L.; Praticò, D. Extra-virgin olive oil ameliorates cognition and neuropathology of the 3xTg mice: Role of autophagy. Ann. Clin. Transl. Neurol. 2017, 4, 564–574. [Google Scholar] [CrossRef]
- Lauretti, E.; Nenov, M.; Dincer, O.; Iuliano, L.; Praticò, D. Extra virgin olive oil improves synaptic activity, short-term plasticity, memory, and neuropathology in a tauopathy model. Aging Cell 2020, 19, e13076. [Google Scholar] [CrossRef] [Green Version]
- Tzekaki, E.E.; Tsolaki, M.; Geromichalos, G.D.; Pantazaki, A.A. Extra Virgin Olive Oil consumption from Mild Cognitive Impairment patients attenuates oxidative and nitrative stress reflecting on the reduction of the PARP levels and DNA damage. Exp. Gerontol. 2021, 156, 111621. [Google Scholar] [CrossRef]
- Zlokovic, B.V. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat. Rev. Neurosci. 2011, 12, 723–738. [Google Scholar] [CrossRef]
- Baloyannis, S.J. Brain capillaries in Alzheimer’s disease. Hell. J. Nucl. Med. 2015, 18, 152. [Google Scholar] [CrossRef]
- Hawkins, B.T.; Davis, T.P. The blood-brain barrier/neurovascular unit in health and disease. Pharmacol. Rev. 2005, 57, 173–185. [Google Scholar] [CrossRef]
- Hardy, J.A.; Higgins, G.A. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef]
- Bates, K.; Verdile, G.; Li, Q.; Ames, D.; Hudson, P.; Masters, C.; Martins, R. Clearance mechanisms of Alzheimer’s amyloid-β peptide: Implications for therapeutic design and diagnostic tests. Mol. Psychiatry 2009, 14, 469–486. [Google Scholar] [CrossRef]
- Jung, Y.-J.; Chung, W.-S. Phagocytic roles of glial cells in healthy and diseased brains. Biomol. Ther. 2018, 26, 350. [Google Scholar] [CrossRef] [PubMed]
- Sasaki, N.; Fukatsu, R.; Tsuzuki, K.; Hayashi, Y.; Yoshida, T.; Fujii, N.; Koike, T.; Wakayama, I.; Yanagihara, R.; Garruto, R. Advanced glycation end products in Alzheimer’s disease and other neurodegenerative diseases. Am. J. Pathol. 1998, 153, 1149–1155. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Takeuchi, M.; Yamagishi, S. TAGE (toxic AGEs) hypothesis in various chronic diseases. Med. Hypotheses 2004, 63, 449–452. [Google Scholar] [CrossRef] [PubMed]
- Takeuchi, M.; Yamagishi, S.-i. Possible involvement of advanced glycation end-products (AGEs) in the pathogenesis of Alzheimer’s disease. Curr. Pharm. Des. 2008, 14, 973–978. [Google Scholar] [CrossRef]
- Krautwald, M.; Münch, G. Advanced glycation end products as biomarkers and gerontotoxins–a basis to explore methylglyoxal-lowering agents for Alzheimer’s disease? Exp. Gerontol. 2010, 45, 744–751. [Google Scholar] [CrossRef]
- Yan, S.D.; 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, 685–691. [Google Scholar] [CrossRef]
- Chen, J.; Song, M.; Yu, S.; Gao, P.; Yu, Y.; Wang, H.; Huang, L. Advanced glycation endproducts alter functions and promote apoptosis in endothelial progenitor cells through receptor for advanced glycation endproducts mediate overpression of cell oxidant stress. Mol. Cell. Biochem. 2010, 335, 137–146. [Google Scholar] [CrossRef]
- Chavakis, E.; Hain, A.; Vinci, M.; Carmona, G.; Bianchi, M.E.; Vajkoczy, P.; Zeiher, A.M.; Chavakis, T.; Dimmeler, S. High-mobility group box 1 activates integrin-dependent homing of endothelial progenitor cells. Circ. Res. 2007, 100, 204–212. [Google Scholar] [CrossRef] [Green Version]
- Hofmann, M.A.; Drury, S.; Fu, C.; Qu, W.; Taguchi, A.; Lu, Y.; Avila, C.; Kambham, N.; Bierhaus, A.; Nawroth, P. RAGE mediates a novel proinflammatory axis: A central cell surface receptor for S100/calgranulin polypeptides. Cell 1999, 97, 889–901. [Google Scholar] [CrossRef] [Green Version]
- Scaffidi, P.; Misteli, T.; Bianchi, M.E. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 2002, 418, 191–195. [Google Scholar] [CrossRef]
- Bianchi, M.E.; Manfredi, A.A. High-mobility group box 1 (HMGB1) protein at the crossroads between innate and adaptive immunity. Immunol. Rev. 2007, 220, 35–46. [Google Scholar] [CrossRef]
- Gao, H.-M.; Zhou, H.; Zhang, F.; Wilson, B.C.; Kam, W.; Hong, J.-S. HMGB1 acts on microglia Mac1 to mediate chronic neuroinflammation that drives progressive neurodegeneration. J. Neurosci. 2011, 31, 1081–1092. [Google Scholar] [CrossRef] [Green Version]
- Son, M.; Lee, S.; Byun, K. Ligands receptor for advanced glycation end products produced by activated microglia are critical in neurodegenerative diseases. J. Alzheimers Dis Park. 2017, 7, 318. [Google Scholar] [CrossRef]
- Mosquera, J.A. Role of the receptor for advanced glycation end products (RAGE) in inflammation. Investig. Clin. 2010, 51, 257–268. [Google Scholar]
- Tan, M.-S.; Yu, J.-T.; Jiang, T.; Zhu, X.-C.; Tan, L. The NLRP3 inflammasome in Alzheimer’s disease. Mol. Neurobiol. 2013, 48, 875–882. [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. 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]
- Bauernfeind, F.G.; Horvath, G.; Stutz, A.; Alnemri, E.S.; MacDonald, K.; Speert, D.; Fernandes-Alnemri, T.; Wu, J.; Monks, B.G.; Fitzgerald, K.A. Cutting edge: NF-κB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J. Immunol. 2009, 183, 787–791. [Google Scholar] [CrossRef] [Green Version]
- Li, Q.; Chen, L.; Liu, X.; Li, X.; Cao, Y.; Bai, Y.; Qi, F. Pterostilbene inhibits amyloid-β-induced neuroinflammation in a microglia cell line by inactivating the NLRP3/caspase-1 inflammasome pathway. J. Cell. Biochem. 2018, 119, 7053–7062. [Google Scholar] [CrossRef]
- Yang, F.; Wang, Z.; Wei, X.; Han, H.; Meng, X.; Zhang, Y.; Shi, W.; Li, F.; Xin, T.; Pang, Q. NLRP3 deficiency ameliorates neurovascular damage in experimental ischemic stroke. J. Cereb. Blood Flow Metab. 2014, 34, 660–667. [Google Scholar] [CrossRef] [Green Version]
- Kaushal, V.; Dye, R.; Pakavathkumar, P.; Foveau, B.; Flores, J.; Hyman, B.; Ghetti, B.; Koller, B.; LeBlanc, A. Neuronal NLRP1 inflammasome activation of Caspase-1 coordinately regulates inflammatory interleukin-1-beta production and axonal degeneration-associated Caspase-6 activation. Cell Death Differ. 2015, 22, 1676–1686. [Google Scholar] [CrossRef] [Green Version]
- Gurung, P.; Kanneganti, T.-D. Novel roles for caspase-8 in IL-1β and inflammasome regulation. Am. J. Pathol. 2015, 185, 17–25. [Google Scholar] [CrossRef] [PubMed]
- Oakley, H.; Cole, S.L.; Logan, S.; Maus, E.; Shao, P.; Craft, J.; Guillozet-Bongaarts, A.; Ohno, M.; Disterhoft, J.; Van Eldik, L. Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: Potential factors in amyloid plaque formation. J. Neurosci. 2006, 26, 10129–10140. [Google Scholar] [CrossRef] [Green Version]
- Richard, B.C.; Kurdakova, A.; Baches, S.; Bayer, T.A.; Weggen, S.; Wirths, O. Gene dosage dependent aggravation of the neurological phenotype in the 5xFAD mouse model of Alzheimer’s disease. J. Alzheimer’s Dis. 2015, 45, 1223–1236. [Google Scholar] [CrossRef] [PubMed]
- Tuck, K.L.; Hayball, P.J. Major phenolic compounds in olive oil: Metabolism and health effects. J. Nutr. Biochem. 2002, 13, 636–644. [Google Scholar] [CrossRef] [PubMed]
Antibody | Company |
---|---|
Western blot | |
Anti-mouse IgG (H+L) secondary antibody, HRP-labeled | Invitrogen (Waltham, MA, USA) |
Anti-rabbit IgG (H+L) secondary antibody, HRP-labeled | Invitrogen |
ADAM10 | Santa Cruz Biotechnology (Dallas, TX, USA) |
Claudin-5 | Invitrogen |
GAPDH | Invitrogen |
HMGB1 | Santa Cruz Biotechnology |
IκB-α | Cell Signaling (Danvers, MA, USA) |
LRP1 | Abcam (Waltham, MA, USA) |
NLRP3 | Cell Signaling |
Occludin | Invitrogen |
p- IκB-α | Cell Signaling |
P-gp | BioLegend (San Diego, CA, USA) |
p-IKK | Cell Signaling |
Pro-caspase 1 | Santa Cruz Biotechnology |
Pro-caspase 8 | Santa Cruz Biotechnology |
PSD-95 | Invitrogen |
RAGE | Santa Cruz Biotechnology |
sAPP-α | IBL America (Minneapolis, MN, USA) |
sAPP-β | IBL America |
SNAP-25 | Invitrogen |
Synapsin-1 | Cell Signaling |
VE-cadherin | Santa Cruz Biotechnology |
Immunofluorescence staining | |
Alexa-fluor 488-labeled 6E10 | BioLegend |
Anti-goat IgG−CFL594 | Santa Cruz Biotechnology |
Anti-goat HRP-labeled secondary | R&D Systems (Minneapolis, MN, USA) |
Anti-collagen-IV | EDM-Millipore (Burlington, MA, USA) |
Anti-rabbit IgG-CFL594 | Santa Cruz Biotechnology |
GFAP | Santa Cruz Biotechnology |
Biophenols (mg/kg) | ROO | EVOO |
---|---|---|
Hydroxytyrosol | 5.1 | |
Tyrosol | 3.1 | |
Vanillic acid and Caffeic acid | 1.8 | |
Vanillin | 1.5 | |
Para-coumaric acid | 2.7 | |
Ferulic acid | 4.2 | |
Decarboxymethyl oleuropein aglycone oxidized dialdehyde form | 34.1 | |
Oleacein | 85.2 | |
Oleuropein | 29.9 | |
Oleuropein aglycone, dialdehyde form | 49.3 | |
Tyrosol acetate | 24.9 | |
Decarboxymethyl ligstroside aglycone, oxidised dialdehyde form | 37.1 | |
Oleocanthal | 33.9 | |
Cinnamic acid | 59.1 | |
Ligstroside aglycone, dialdehyde form | 12.7 | |
Oleuropein aglycone, oxidized aldehyde, and hydroxylic form | 17.4 | |
Luteolin | 36 | |
Oleuropein aglycone, aldehyde, and hydroxylic form | 58.2 | |
Ligstroside aglycone, oxidised aldehyde, and hyroxylic form | 15.1 | |
Apigenin | 16.6 | |
Ligstoside aglycone, aldehyde and hydroxylic form | 11.4 | |
Total biophenol content mg/kg | <10 | 539.3 |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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
Abdallah, I.M.; Al-Shami, K.M.; Alkhalifa, A.E.; Al-Ghraiybah, N.F.; Guillaume, C.; Kaddoumi, A. Comparison of Oleocanthal-Low EVOO and Oleocanthal against Amyloid-β and Related Pathology in a Mouse Model of Alzheimer’s Disease. Molecules 2023, 28, 1249. https://doi.org/10.3390/molecules28031249
Abdallah IM, Al-Shami KM, Alkhalifa AE, Al-Ghraiybah NF, Guillaume C, Kaddoumi A. Comparison of Oleocanthal-Low EVOO and Oleocanthal against Amyloid-β and Related Pathology in a Mouse Model of Alzheimer’s Disease. Molecules. 2023; 28(3):1249. https://doi.org/10.3390/molecules28031249
Chicago/Turabian StyleAbdallah, Ihab M., Kamal M. Al-Shami, Amer E. Alkhalifa, Nour F. Al-Ghraiybah, Claudia Guillaume, and Amal Kaddoumi. 2023. "Comparison of Oleocanthal-Low EVOO and Oleocanthal against Amyloid-β and Related Pathology in a Mouse Model of Alzheimer’s Disease" Molecules 28, no. 3: 1249. https://doi.org/10.3390/molecules28031249
APA StyleAbdallah, I. M., Al-Shami, K. M., Alkhalifa, A. E., Al-Ghraiybah, N. F., Guillaume, C., & Kaddoumi, A. (2023). Comparison of Oleocanthal-Low EVOO and Oleocanthal against Amyloid-β and Related Pathology in a Mouse Model of Alzheimer’s Disease. Molecules, 28(3), 1249. https://doi.org/10.3390/molecules28031249