en Experimental Neurobiology

Cited by CrossRef (70)

  1. Joseph L. Bundy, Cynthia Vied, Crystal Badger, Richard S. Nowakowski. Sex‐biased hippocampal pathology in the 5XFAD mouse model of Alzheimer's disease: A multi‐omic analysis. J of Comparative Neurology 2019;527:462
    https://doi.org/10.1002/cne.24551
  2. Majbrit M. Jensen, Maria Arvaniti, Jens D. Mikkelsen, Dominik Michalski, Lars H. Pinborg, Wolfgang Härtig, Morten S. Thomsen. Prostate stem cell antigen interacts with nicotinic acetylcholine receptors and is affected in Alzheimer's disease. Neurobiology of Aging 2015;36:1629
    https://doi.org/10.1016/j.neurobiolaging.2015.01.001
  3. Margaret Ryan, Valerie T.Y. Tan, Nasya Thompson, Diane Guévremont, Bruce G. Mockett, Warren P. Tate, Wickliffe C. Abraham, Stephanie M. Hughes, Joanna Williams. Lentivirus-Mediated Expression of Human Secreted Amyloid Precursor Protein-Alpha Promotes Long-Term Induction of Neuroprotective Genes and Pathways in a Mouse Model of Alzheimer’s Disease. JAD 2021;79:1075
    https://doi.org/10.3233/JAD-200757
  4. Yao-Hsiang Shih, Shih-Ying Wu, Megan Yu, Sheng-Huai Huang, Chu-Wan Lee, Meei-Jyh Jiang, Pao-Yen Lin, Ting-Ting Yang, Yu-Min Kuo. Hypertension Accelerates Alzheimer’s Disease-Related Pathologies in Pigs and 3xTg Mice. Front. Aging Neurosci. 2018;10
    https://doi.org/10.3389/fnagi.2018.00073
  5. Yihe Wang, Tianrui Zhu, Min Wang, Feng Zhang, Guitao Zhang, Jing Zhao, Yuanyuan Zhang, Erxi Wu, Xiaohong Li. Icariin Attenuates M1 Activation of Microglia and Aβ Plaque Accumulation in the Hippocampus and Prefrontal Cortex by Up-Regulating PPARγ in Restraint/Isolation-Stressed APP/PS1 Mice. Front. Neurosci. 2019;13
    https://doi.org/10.3389/fnins.2019.00291
  6. Zhengjiang Qian, ZhiHao Wang, Bowei Li, Xin Meng, Zhonghua Kuang, Yanjiao Li, Yongfeng Yang, Keqiang Ye. Thy1-ApoE4/C/EBPβ double transgenic mice act as a sporadic model with Alzheimer’s disease. Mol Psychiatry 2024
    https://doi.org/10.1038/s41380-024-02565-x
  7. José Antonio Allué, Leticia Sarasa, María Izco, Virginia Pérez-Grijalba, Noelia Fandos, María Pascual-Lucas, Samuel Ogueta, Pedro Pesini, Manuel Sarasa. Outstanding Phenotypic Differences in the Profile of Amyloid-β between Tg2576 and APPswe/PS1dE9 Transgenic Mouse Models of Alzheimer’s Disease. JAD 2016;53:773
    https://doi.org/10.3233/JAD-160280
  8. Margaret M. Ryan, Diane Guévremont, Bruce G. Mockett, Wickliffe C. Abraham, Joanna M. Williams. Circulating Plasma microRNAs are Altered with Amyloidosis in a Mouse Model of Alzheimer’s Disease. JAD 2018;66:835
    https://doi.org/10.3233/JAD-180385
  9. Yingjun Zhao, Xilin Wu, Xiaoguang Li, Lu-Lin Jiang, Xun Gui, Yan Liu, Yu Sun, Bing Zhu, Juan C. Piña-Crespo, Muxian Zhang, Ningyan Zhang, Xiaochun Chen, Guojun Bu, Zhiqiang An, Timothy Y. Huang, Huaxi Xu. TREM2 Is a Receptor for β-Amyloid that Mediates Microglial Function. Neuron 2018;97:1023
    https://doi.org/10.1016/j.neuron.2018.01.031
  10. Sylvie L. Lesuis, Herve Maurin, Peter Borghgraef, Paul J. Lucassen, Fred Van Leuven, Harm J. Krugers. Positive and negative early life experiences differentially modulate long term survival and amyloid protein levels in a mouse model of Alzheimer's disease. Oncotarget 2016;7:39118
    https://doi.org/10.18632/oncotarget.9776
  11. Jan Rehker, Johanna Rodhe, Ryan R. Nesbitt, Evan A. Boyle, Beth K. Martin, Jenny Lord, Ilker Karaca, Adam Naj, Frank Jessen, Seppo Helisalmi, Hilkka Soininen, Mikko Hiltunen, Alfredo Ramirez, Martin Scherer, Lindsay A. Farrer, Jonathan L. Haines, Margaret A. Pericak-Vance, Wendy H. Raskind, Carlos Cruchaga, Gerard D. Schellenberg, Bertrand Joseph, Zoran Brkanac, Madepalli K. Lakshmana. Caspase-8, association with Alzheimer’s Disease and functional analysis of rare variants. PLoS ONE 2017;12:e0185777
    https://doi.org/10.1371/journal.pone.0185777
  12. Liqin Zhao, Sarah K. Woody, Anindit Chhibber. Estrogen receptor β in Alzheimer’s disease: From mechanisms to therapeutics. Ageing Research Reviews 2015;24:178
    https://doi.org/10.1016/j.arr.2015.08.001
  13. Jennifer D. Whitesell, Alex R. Buckley, Joseph E. Knox, Leonard Kuan, Nile Graddis, Andrew Pelos, Alice Mukora, Wayne Wakeman, Phillip Bohn, Anh Ho, Karla E. Hirokawa, Julie A. Harris. Whole brain imaging reveals distinct spatial patterns of amyloid beta deposition in three mouse models of Alzheimer's disease. J of Comparative Neurology 2019;527:2122
    https://doi.org/10.1002/cne.24555
  14. Leonidas Chouliaras, Roy Lardenoije, Gunter Kenis, Diego Mastroeni, Patrick R. Hof, Jim van Os, Harry W.M. Steinbusch, Fred W. van Leeuwen, Bart P.F. Rutten, Daniel L.A. van den Hove. Age-related disturbances in DNA (hydroxy)methylation in APP/PS1 mice. 2018;9:190
    https://doi.org/10.1515/tnsci-2018-0028
  15. Ji-Hun Shin, Young Sang Hwang, Bong-Kwang Jung, Seung-Hwan Seo, Do-Won Ham, Eun-Hee Shin. Reduction of Amyloid Burden by Proliferated Homeostatic Microglia in Toxoplasma gondii-Infected Alzheimer’s Disease Model Mice. IJMS 2021;22:2764
    https://doi.org/10.3390/ijms22052764
  16. Waiian Leong, Wei Xu, Bo Wang, Shuaiyun Gao, Xiuyun Zhai, Cuicui Wang, Eric Gilson, Jing Ye, Yiming Lu. PP2A subunit PPP2R2C is downregulated in the brains of Alzheimer’s transgenic mice. Aging 2020;12:6880
    https://doi.org/10.18632/aging.103048
  17. Hanna Bielarczyk, Agnieszka Jankowska-Kulawy, Corinna Höfling, Anna Ronowska, Sylwia Gul-Hinc, Steffen Roßner, Reinhard Schliebs, Tadeusz Pawelczyk, Andrzej Szutowicz. AβPP-Transgenic 2576 Mice Mimic Cell Type-Specific Aspects of Acetyl-CoA-Linked Metabolic Deficits in Alzheimer’s Disease. JAD 2015;48:1083
    https://doi.org/10.3233/JAD-150327
  18. Fabio Urbina, Ana C. Puhl, Sean Ekins. Recent advances in drug repurposing using machine learning. Current Opinion in Chemical Biology 2021;65:74
    https://doi.org/10.1016/j.cbpa.2021.06.001
  19. Yajing Liu, Jinjing Yao, Zhenpeng Song, Wenting Guo, Bo Sun, Jinhong Wei, John Paul Estillore, Thomas G. Back, S. R. Wayne Chen. Limiting RyR2 open time prevents Alzheimer's disease‐related deficits in the 3xTG‐AD mouse model. J of Neuroscience Research 2021;99:2906
    https://doi.org/10.1002/jnr.24936
  20. A-Hyeon Lee, Sung-Chul Hong, Inwook Park, Soljee Yoon, YoungSoo Kim, Jinsik Kim, Seung-Hoon Yang. Validation of Fucoxanthin from Microalgae Phaeodactylum tricornutum for the Detection of Amyloid Burden in Transgenic Mouse Models of Alzheimer’s Disease. Applied Sciences 2021;11:5878
    https://doi.org/10.3390/app11135878
  21. Jinjing Yao, S. R. Wayne Chen. RyR2‐dependent modulation of neuronal hyperactivity: A potential therapeutic target for treating Alzheimer's disease. The Journal of Physiology 2024;602:1509
    https://doi.org/10.1113/JP283824
  22. Iván Carrera, Ignacio Etcheverría, Lucía Fernández-Novoa, Valter Ruggero Maria Lombardi, Madepalli Krishnappa Lakshmana, Ramón Cacabelos, Carmen Vigo. A Comparative Evaluation of a Novel Vaccine in APP/PS1 Mouse Models of Alzheimer’s Disease. BioMed Research International 2015;2015:1
    https://doi.org/10.1155/2015/807146
  23. Miguel Ángel Ontiveros-Torres, María Luisa Labra-Barrios, Sofía Díaz-Cintra, Azucena Ruth Aguilar-Vázquez, Samadhi Moreno-Campuzano, Paola Flores-Rodríguez, Claudia Luna-Herrera, Raúl Mena, George Perry, Benjamín Florán-Garduño, José Luna-Muñoz, Juan Pedro Luna-Arias, Jesus Avila. Fibrillar Amyloid-β Accumulation Triggers an Inflammatory Mechanism Leading to Hyperphosphorylation of the Carboxyl-Terminal End of Tau Polypeptide in the Hippocampal Formation of the 3×Tg-AD Transgenic Mouse. JAD 2016;52:243
    https://doi.org/10.3233/JAD-150837
  24. Jean-Marie Billard. Changes in Serine Racemase-Dependent Modulation of NMDA Receptor: Impact on Physiological and Pathological Brain Aging. Front. Mol. Biosci. 2018;5
    https://doi.org/10.3389/fmolb.2018.00106
  25. Mickael Audrain, Romain Fol, Patrick Dutar, Brigitte Potier, Jean-Marie Billard, Julien Flament, Sandro Alves, Marie-Anne Burlot, Gaelle Dufayet-Chaffaud, Alexis-Pierre Bemelmans, Julien Valette, Philippe Hantraye, Nicole Déglon, Nathalie Cartier, Jérome Braudeau. Alzheimer’s disease-like APP processing in wild-type mice identifies synaptic defects as initial steps of disease progression. Mol Neurodegeneration 2016;11
    https://doi.org/10.1186/s13024-016-0070-y
  26. George D. Wilson, Brian Marples. A New Use for an Old Treatment: Radiation Therapy and Alzheimer's Disease. Radiation Research 2016;185:443
    https://doi.org/10.1667/RR14367.1
  27. Michael Klonarakis, Maya De Vos, Emma K. Woo, Liam T. Ralph, Jonathan S. Thacker, Joana Gil-Mohapel. The three sisters of fate: Genetics, pathophysiology and outcomes of animal models of neurodegenerative diseases. Neuroscience & Biobehavioral Reviews 2022;135:104541
    https://doi.org/10.1016/j.neubiorev.2022.104541
  28. Dong-Hee Kim, Yoon-Sun Jang, Won Kyung Jeon, Jung-Soo Han. Assessment of Cognitive Phenotyping in Inbred, Genetically Modified Mice, and Transgenic Mouse Models of Alzheimer's Disease. Exp Neurobiol 2019;28:146
    https://doi.org/10.5607/en.2019.28.2.146
  29. Yi‐Fan Chen, Tzu‐Yu Chou, I‐Hsuan Lin, Chung‐Guang Chen, Cheng‐Heng Kao, Guo‐Jen Huang, Liang‐Kung Chen, Pei‐Ning Wang, Ching‐Po Lin, Ting‐Fen Tsai. Upregulation of Cisd2 attenuates Alzheimer's‐related neuronal loss in mice. The Journal of Pathology 2020;250:299
    https://doi.org/10.1002/path.5374
  30. 旭旭 庄. Research Progress of APP/PS1 Mice on Neuronal Apoptosis. PI 2016;05:14
    https://doi.org/10.12677/PI.2016.52003
  31. Yuno Song, Hong-Duck Kim, Min-Kwon Lee, Il-Hwa Hong, Chung-Kil Won, Hyoung-Woo Bai, Seung Sik Lee, SungBeom Lee, Byung Yeoup Chung, Jae-Hyeon Cho, Maya Koronyo-Hamaoui. Maysin and Its Flavonoid Derivative from Centipedegrass Attenuates Amyloid Plaques by Inducting Humoral Immune Response with Th2 Skewed Cytokine Response in the Tg (APPswe, PS1dE9) Alzheimer’s Mouse Model. PLoS ONE 2017;12:e0169509
    https://doi.org/10.1371/journal.pone.0169509
  32. Smilja Todorovic, Natasa Loncarevic-Vasiljkovic, Milena Jovic, Srdjan Sokanovic, Selma Kanazir, Aleksandra Mladenovic Djordjevic. Frailty index and phenotype frailty score: Sex- and age-related differences in 5XFAD transgenic mouse model of Alzheimer’s disease. Mechanisms of Ageing and Development 2020;185:111195
    https://doi.org/10.1016/j.mad.2019.111195
  33. Chin-Chia Huang, Hsiang-Fan Cheng, Ben-Peng Zhu, Pei-Yu Chen, Suet Theng Beh, Yu-Min Kuo, Chih-Chung Huang. Studying Arterial Stiffness Using High-Frequency Ultrasound in Mice with Alzheimer Disease. Ultrasound in Medicine & Biology 2017;43:2054
    https://doi.org/10.1016/j.ultrasmedbio.2017.04.029
  34. Pedro Cisternas, Juan M. Zolezzi, Carolina Lindsay, Daniela S. Rivera, Alexis Martinez, Francisco Bozinovic, Nibaldo C. Inestrosa. New Insights into the Spontaneous Human Alzheimer’s Disease-Like Model Octodon degus: Unraveling Amyloid-β Peptide Aggregation and Age-Related Amyloid Pathology. JAD 2018;66:1145
    https://doi.org/10.3233/JAD-180729
  35. Tzeng Tsai-Teng, Chen Chin-Chu, Lee Li-Ya, Chen Wan-Ping, Lu Chung-Kuang, Shen Chien-Chang, Huang F. Chi-Ying, Chen Chien-Chih, Young-Ji Shiao. Erinacine A-enriched Hericium erinaceus mycelium ameliorates Alzheimer’s disease-related pathologies in APPswe/PS1dE9 transgenic mice. J Biomed Sci 2016;23
    https://doi.org/10.1186/s12929-016-0266-z
  36. Bridget Martinez, PhilipV Peplow. MicroRNAs as diagnostic and therapeutic tools for Alzheimer’s disease: advances and limitations. Neural Regen Res 2019;14:242
    https://doi.org/10.4103/1673-5374.244784
  37. Sarah E. Smith, Azad Bonni. The Molecular and Cellular Basis of Neurodegenerative Diseases. 2019.
    https://doi.org/10.1016/B978-0-12-811304-2.00014-6
  38. Sarah Logan, Thiago Arzua, Scott G. Canfield, Emily R. Seminary, Samantha L. Sison, Allison D. Ebert, Xiaowen Bai. Comprehensive Physiology. 2019.
    https://doi.org/10.1002/cphy.c180025
  39. Yu Qi, Xinhui Cheng, Guowei Gong, Tingxu Yan, Yiyang Du, Bo Wu, Kaishun Bi, Ying Jia. Synergistic neuroprotective effect of schisandrin and nootkatone on regulating inflammation, apoptosis and autophagy via the PI3K/AKT pathway. Food Funct. 2020;11:2427
    https://doi.org/10.1039/C9FO02927C
  40. Seon-Young Park, Juwon Yang, Hyejin Yang, Inhee Cho, Jae Yoon Kim, Hyunsu Bae. Therapeutic Effects of Aβ-Specific Regulatory T Cells in Alzheimer’s Disease: A Study in 5xFAD Mice. IJMS 2024;25:783
    https://doi.org/10.3390/ijms25020783
  41. Akhlaq A. Farooqui. Therapeutic Potentials of Curcumin for Alzheimer Disease. 2024.
    https://doi.org/10.1007/978-3-319-15889-1_2
  42. Zhi-Hao Wang, Yiyuan Xia, Zhourui Wu, Seong Su Kang, Ji-chun Zhang, Pai Liu, Xia Liu, Weihong Song, Vincent Huin, Claire-Marie Dhaenens, Shan Ping Yu, Xiao-Chuan Wang, Keqiang Ye. Neuronal ApoE4 stimulates C/EBPβ activation, promoting Alzheimer’s disease pathology in a mouse model. Progress in Neurobiology 2022;209:102212
    https://doi.org/10.1016/j.pneurobio.2021.102212
  43. Lu-Lu Xue, Li-Ren Huangfu, Ruo-Lan Du, Li Chen, Chang-Yin Yu, Liu-Lin Xiong, Ting-Hua Wang. The age-specific pathological changes of β-amyloid plaques in the cortex and hippocampus of APP/PS1 transgenic AD mice. Neurological Research 2022;44:1053
    https://doi.org/10.1080/01616412.2022.2112368
  44. Pierre Faucher, Nicole Mons, Jacques Micheau, Caroline Louis, Daniel J. Beracochea. Hippocampal Injections of Oligomeric Amyloid β-peptide (1–42) Induce Selective Working Memory Deficits and Long-lasting Alterations of ERK Signaling Pathway. Front. Aging Neurosci. 2016;7
    https://doi.org/10.3389/fnagi.2015.00245
  45. F. Schneider, K. Baldauf, W. Wetzel, K.G. Reymann. Behavioral and EEG changes in male 5xFAD mice. Physiology & Behavior 2014;135:25
    https://doi.org/10.1016/j.physbeh.2014.05.041
  46. Chang Tan, Yaqiong Dong, Jing Wang, Xiaoda Yang. Vanadyl acetylacetonate attenuates Aβ pathogenesis in APP/PS1 transgenic mice depending on the intervention stage. New J. Chem. 2019;43:17588
    https://doi.org/10.1039/C9NJ00820A
  47. Xinhua Zhou, Wei Xiao, Zhiyang Su, Jiehong Cheng, Chengyou Zheng, Zaijun Zhang, Yuqiang Wang, Liang Wang, Benhong Xu, Shupen Li, Xifei Yang, Maggie Pui Man Hoi. Hippocampal Proteomic Alteration in Triple Transgenic Mouse Model of Alzheimer’s Disease and Implication of PINK 1 Regulation in Donepezil Treatment. J. Proteome Res. 2019;18:1542
    https://doi.org/10.1021/acs.jproteome.8b00818
  48. Jin-Young Park, Juli Choi, Yunjin Lee, Jung-Eun Lee, Eun-Hwa Lee, Hye-Jin Kwon, Jinho Yang, Bo-Ri Jeong, Yoon-Keun Kim, Pyung-Lim Han. Metagenome Analysis of Bodily Microbiota in a Mouse Model of Alzheimer Disease Using Bacteria-derived Membrane Vesicles in Blood. Exp Neurobiol 2017;26:369
    https://doi.org/10.5607/en.2017.26.6.369
  49. Carolina B. Lindsay, Juan M. Zolezzi, Daniela S. Rivera, Pedro Cisternas, Francisco Bozinovic, Nibaldo C. Inestrosa. Andrographolide Reduces Neuroinflammation and Oxidative Stress in Aged Octodon degus. Mol Neurobiol 2020;57:1131
    https://doi.org/10.1007/s12035-019-01784-6
  50. Xiaoyan Tian, Chaonan Ji, Ying Luo, Yang Yang, Shengnan Kuang, Shaoshan Mai, Jie Ma, Junqing Yang. PGE2-EP3 signaling pathway contributes to protective effects of misoprostol on cerebral injury in APP/PS1 mice. Oncotarget 2016;7:25304
    https://doi.org/10.18632/oncotarget.8284
  51. Jinjing Yao, S. R. Wayne Chen. R-carvedilol, a potential new therapy for Alzheimer’s disease. Front. Pharmacol. 2022;13
    https://doi.org/10.3389/fphar.2022.1062495
  52. Amitha Muraleedharan, Noa Rotem‐Dai, Itai Strominger, Nikhil Ponnoor Anto, Noah Isakov, Alon Monsonego, Etta Livneh. Protein kinase C eta is activated in reactive astrocytes of an Alzheimer's disease mouse model: Evidence for its immunoregulatory function in primary astrocytes. Glia 2021;69:697
    https://doi.org/10.1002/glia.23921
  53. Nibaldo C. Inestrosa, Juvenal A. Ríos, Pedro Cisternas, Cheril Tapia‐Rojas, Daniela S. Rivera, Nady Braidy, Juan M. Zolezzi, Juan A. Godoy, Francisco J. Carvajal, Alvaro O. Ardiles, Francisco Bozinovic, Adrián G. Palacios, Perminder S. Sachdev. Age Progression of Neuropathological Markers in the Brain of the Chilean Rodent Octodon degus, a Natural Model of Alzheimer's Disease. Brain Pathology 2015;25:679
    https://doi.org/10.1111/bpa.12226
  54. G.S. Finnie, R. Gunnarsson, J. Manavis, P.C. Blumbergs, K.A. Mander, S. Edwards, C. Van den Heuvel, J.W. Finnie. Characterization of an ‘Amyloid Only’ Transgenic (B6C3-Tg(APPswe,PSEN1dE9)85Dbo/Mmjax) Mouse Model of Alzheimer's Disease. Journal of Comparative Pathology 2017;156:389
    https://doi.org/10.1016/j.jcpa.2017.03.001
  55. Lu Wang, Lan Guo, Lin Lu, Huili Sun, Muming Shao, Simon J. Beck, Lin Li, Janani Ramachandran, Yifeng Du, Heng Du, Stephen D Ginsberg. Synaptosomal Mitochondrial Dysfunction in 5xFAD Mouse Model of Alzheimer's Disease. PLoS ONE 2016;11:e0150441
    https://doi.org/10.1371/journal.pone.0150441
  56. Riikka Lampinen, Irina Belaya, Isabella Boccuni, Tarja Malm, Katja M. Kanninen. Astrocyte - Physiology and Pathology. 2016.
    https://doi.org/10.5772/intechopen.71825
  57. Shupeng Liu, Shuyang Li, Yudan Xia, Heng Zhang, Jing Tian, Chunlei Shan, Fufei Pang, Ying Wang, Yana Shang, Na Chen. Effects of multi-mode physical stimulation on APP/PS1 Alzheimer's disease model mice. Heliyon 2022;8:e12366
    https://doi.org/10.1016/j.heliyon.2022.e12366
  58. Sandra Amor, Laura A. N. Peferoen, Daphne Y. S. Vogel, Marjolein Breur, Paul van der Valk, David Baker, Johannes M. van Noort. Inflammation in neurodegenerative diseases – an update. Immunology 2014;142:151
    https://doi.org/10.1111/imm.12233
  59. Mujun Sun, Stuart J. McDonald, Rhys D. Brady, Lyndsey Collins-Praino, Glenn R. Yamakawa, Mastura Monif, Terence J. O’Brien, Geoffrey C. Cloud, Christopher G. Sobey, Richelle Mychasiuk, David J. Loane, Sandy R. Shultz. The need to incorporate aged animals into the preclinical modeling of neurological conditions. Neuroscience & Biobehavioral Reviews 2020;109:114
    https://doi.org/10.1016/j.neubiorev.2019.12.027
  60. Mar Cuadrado-Tejedor, Ana García-Osta. Current Animal Models of Alzheimer’s Disease: Challenges in Translational Research. Front. Neurol. 2014;5
    https://doi.org/10.3389/fneur.2014.00182
  61. Hong-Lei Gao, Ai-Hua Zhang, Jing-Bo Yu, Hui Sun, Ling Kong, Xiang-Qian Wang, Guang-li Yan, Liang Liu, Xi-Jun Wang. High-throughput lipidomics characterize key lipid molecules as potential therapeutic targets of Kaixinsan protects against Alzheimer's disease in APP/PS1 transgenic mice. Journal of Chromatography B 2018;1092:286
    https://doi.org/10.1016/j.jchromb.2018.06.032
  62. Áine M. Duffy, Jose Morales-Corraliza, Keria M. Bermudez-Hernandez, Michael J. Schaner, Alejandra Magagna-Poveda, Paul M. Mathews, Helen E. Scharfman. Entorhinal cortical defects in Tg2576 mice are present as early as 2–4 months of age. Neurobiology of Aging 2015;36:134
    https://doi.org/10.1016/j.neurobiolaging.2014.07.001
  63. Irina Belaya, Mariia Ivanova, Annika Sorvari, Marina Ilicic, Sanna Loppi, Hennariikka Koivisto, Alessandra Varricchio, Heikki Tikkanen, Frederick R. Walker, Mustafa Atalay, Tarja Malm, Alexandra Grubman, Heikki Tanila, Katja M. Kanninen. Astrocyte remodeling in the beneficial effects of long-term voluntary exercise in Alzheimer’s disease. J Neuroinflammation 2020;17
    https://doi.org/10.1186/s12974-020-01935-w
  64. Flavio H Beraldo, Daniel Palmer, Sara Memar, David I Wasserman, Wai-Jane V Lee, Shuai Liang, Samantha D Creighton, Benjamin Kolisnyk, Matthew F Cowan, Justin Mels, Talal S Masood, Chris Fodor, Mohammed A Al-Onaizi, Robert Bartha, Tom Gee, Lisa M Saksida, Timothy J Bussey, Stephen S Strother, Vania F Prado, Boyer D Winters, Marco AM Prado. MouseBytes, an open-access high-throughput pipeline and database for rodent touchscreen-based cognitive assessment. 2019;8
    https://doi.org/10.7554/eLife.49630
  65. Jesse Britz, Emmanuel Ojo, Asmita Dhukhwa, Takashi Saito, Takaomi C. Saido, Erin R. Hascup, Kevin N. Hascup, Shelley A. Tischkau. Assessing Sex-Specific Circadian, Metabolic, and Cognitive Phenotypes in the AβPP/PS1 and APPNL-F/NL-F Models of Alzheimer’s Disease. JAD 2022;85:1077
    https://doi.org/10.3233/JAD-210629
  66. Daxiang Na, Jingyuan Zhang, Holly J. Beaulac, Dorota Piekna-Przybylska, Paige R. Nicklas, Amy E. Kiernan, Patricia M. White. Increased central auditory gain in 5xFAD Alzheimer’s disease mice as an early biomarker candidate for Alzheimer’s disease diagnosis. Front. Neurosci. 2023;17
    https://doi.org/10.3389/fnins.2023.1106570
  67. Sara Porcellotti, Francesca Fanelli, Anna Fracassi, Sara Sepe, Francesco Cecconi, Cinzia Bernardi, AnnaMaria Cimini, Maria Paola Cerù, Sandra Moreno. Oxidative Stress during the Progression ofβ-Amyloid Pathology in the Neocortex of the Tg2576 Mouse Model of Alzheimer’s Disease. Oxidative Medicine and Cellular Longevity 2015;2015:1
    https://doi.org/10.1155/2015/967203
  68. Mickael Audrain, Benoit Souchet, Sandro Alves, Romain Fol, Arthur Viode, Alexis Haddjeri, Satoru Tada, Nicola S Orefice, Charlène Joséphine, Alexis-Pierre Bemelmans, Thierry Delzescaux, Nicole Déglon, Philippe Hantraye, Yvette Akwa, François Becher, Jean-Marie Billard, Brigitte Potier, Patrick Dutar, Nathalie Cartier, Jérôme Braudeau. βAPP Processing Drives Gradual Tau Pathology in an Age-Dependent Amyloid Rat Model of Alzheimer’s Disease. 2018;28:3976
    https://doi.org/10.1093/cercor/bhx260
  69. Wojciech Michno, Katie M. Stringer, Thomas Enzlein, Melissa K. Passarelli, Stephane Escrig, Karina Vitanova, Jack Wood, Kaj Blennow, Henrik Zetterberg, Anders Meibom, Carsten Hopf, Frances A. Edwards, Jörg Hanrieder. Following spatial Aβ aggregation dynamics in evolving Alzheimer’s disease pathology by imaging stable isotope labeling kinetics. Sci. Adv. 2021;7
    https://doi.org/10.1126/sciadv.abg4855
  70. Diego Fernández‐Fernández, Cornelia Dorner‐Ciossek, Katja S. Kroker, Holger Rosenbrock. Age‐related synaptic dysfunction in Tg2576 mice starts as a failure in early long‐term potentiation which develops into a full abolishment of late long‐term potentiation. J of Neuroscience Research 2016;94:266
    https://doi.org/10.1002/jnr.23701