1
|
Zhang NK, Zhang SK, Zhang LI, Tao HW, Zhang GW. The neural basis of neuropsychiatric symptoms in Alzheimer's disease. Front Aging Neurosci 2024; 16:1487875. [PMID: 39703925 PMCID: PMC11655510 DOI: 10.3389/fnagi.2024.1487875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/19/2024] [Indexed: 12/21/2024] Open
Abstract
Neuropsychiatric symptoms (NPS) such as depression, anxiety, apathy and aggression affect up to 90% of Alzheimer's disease (AD) patients. These symptoms significantly increase caregiver stress and institutionalization rates, and more importantly they are correlated with faster cognitive decline. However, the neuronal basis of NPS in AD remains largely unknown. Here, we review current understanding of NPS and related pathology in studies of AD patients and AD mouse models. Clinical studies indicate that NPS prevalence and severity vary across different AD stages and types. Neuroimaging and postmortem studies have suggested that pathological changes in the anterior cingulate cortex, hippocampus, prefrontal cortex, and amygdala are linked to NPS, although the precise mechanisms remain unclear. Studies of AD mouse models have indicated that amyloid-beta and tau-related neurodegeneration in the hippocampus, prefrontal cortex, and anterior cingulate cortex are correlated with NPS-like behavioral deficits. A better understanding of the NPS phenotypes and related pathological changes will pave the way for developing a better management strategy for NPS in AD patients.
Collapse
Affiliation(s)
- Nicole K. Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Selena K. Zhang
- Biomedical Engineering Program, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Li I. Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Huizhong W. Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
2
|
AlQot HE, Rylett RJ. Primate-specific 82-kDa choline acetyltransferase attenuates progression of Alzheimer's disease-like pathology in the APP NL-G-F knock-in mouse model. Sci Rep 2024; 14:27614. [PMID: 39528509 PMCID: PMC11555257 DOI: 10.1038/s41598-024-78751-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloidosis, neuroinflammation, cholinergic dysfunction and cognitive impairment. In AD, the cholinergic neuronal marker choline acetyltransferase (ChAT) is reduced and the primate-specific nuclear isoform, 82-kDa ChAT, is mislocalized to cytoplasm. Cell-based studies suggest a role for 82-kDa ChAT in regulating expression of AD-related genes with potential reductions in β-amyloid (Aβ) levels. To study this further, we crossed transgenic mice expressing human 82-kDa ChAT with the AD mouse model APPNL-G-F and used molecular techniques and neurobehavioral tests to study the impact of 82-kDa ChAT on AD pathology. These mice had altered expression of genes linked to Aβ clearance and inflammation, and reduced cognitive decline, amyloidosis and gliosis. These effects were inversely related to age and Aβ plaque load and correlated best with 82-kDa ChAT localized to nuclei of neurons. The study suggests a role for 82-kDa ChAT in decreasing AD-like pathology.
Collapse
Affiliation(s)
- Hadir E AlQot
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Robarts Research Institute, University of Western Ontario, London, ON, N6A 5K8, Canada
| | - Rebecca Jane Rylett
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Robarts Research Institute, University of Western Ontario, London, ON, N6A 5K8, Canada.
| |
Collapse
|
3
|
Mehla J, Deibel SH, Karem H, Hong NS, Hossain SR, Lacoursiere SG, Sutherland RJ, Mohajerani MH, McDonald RJ. Repeated multi-domain cognitive training prevents cognitive decline, anxiety and amyloid pathology found in a mouse model of Alzheimer disease. Commun Biol 2023; 6:1145. [PMID: 37950055 PMCID: PMC10638434 DOI: 10.1038/s42003-023-05506-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Education, occupation, and an active lifestyle, comprising enhanced social, physical, and mental components are associated with improved cognitive functions in aged people and may delay the progression of various neurodegenerative diseases including Alzheimer's disease. To investigate this protective effect, 3-month-old APPNL-G-F/NL-G-F mice were exposed to repeated single- or multi-domain cognitive training. Cognitive training was given at the age of 3, 6, & 9 months. Single-domain cognitive training was limited to a spatial navigation task. Multi-domain cognitive training consisted of a spatial navigation task, object recognition, and fear conditioning. At the age of 12 months, behavioral tests were completed for all groups. Then, mice were sacrificed, and their brains were assessed for pathology. APPNL-G-F/NL-G-F mice given multi-domain cognitive training compared to APPNL-G-F/NL-G-F control group showed an improvement in cognitive functions, reductions in amyloid load and microgliosis, and a preservation of cholinergic function. Additionally, multi-domain cognitive training improved anxiety in APPNL-G-F/NL-G-F mice as evidenced by measuring thigmotaxis behavior in the Morris water maze. There were mild reductions in microgliosis in the brain of APPNL-G-F/NL-G-F mice with single-domain cognitive training. These findings provide causal evidence for the potential of certain forms of cognitive training to mitigate the cognitive deficits in Alzheimer disease.
Collapse
Affiliation(s)
- Jogender Mehla
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Scott H Deibel
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
- Department of Psychology, University of New Brunswick, POB 4400, Fredericton, NB, E3B 3A1, Canada
| | - Hadil Karem
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Nancy S Hong
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Shakhawat R Hossain
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Sean G Lacoursiere
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Robert J Sutherland
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Majid H Mohajerani
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada.
| | - Robert J McDonald
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada.
| |
Collapse
|
4
|
Tan S, Tong WH, Vyas A. Impaired episodic-like memory in a mouse model of Alzheimer's disease is associated with hyperactivity in prefrontal-hippocampal regions. Dis Model Mech 2023; 16:297102. [PMID: 36897115 PMCID: PMC10040242 DOI: 10.1242/dmm.049945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/27/2023] [Indexed: 03/11/2023] Open
Abstract
Alzheimer's disease (AD) is a degenerative brain disorder with a long prodromal period. An APPNL-G-F knock-in mouse model is a preclinical model to study incipient pathologies during the early stages of AD. Despite behavioral tests revealing broad cognitive deficits in APPNL-G-F mice, detecting these impairments at the early disease phase has been challenging. In a cognitively demanding task that assessed episodic-like memory, 3-month-old wild-type mice could incidentally form and retrieve 'what-where-when' episodic associations of their past encounters. However, 3-month-old APPNL-G-F mice, corresponding to an early disease stage without prominent amyloid plaque pathology, displayed impairment in recalling 'what-where' information of past episodes. Episodic-like memory is also sensitive to the effect of age. Eight-month-old wild-type mice failed to retrieve conjunctive 'what-where-when' memories. This deficit was also observed in 8-month-old APPNL-G-F mice. c-Fos expression revealed that impaired memory retrieval in APPNL-G-F mice was accompanied by abnormal neuronal hyperactivity in the medial prefrontal cortex and CA1 dorsal hippocampus. These observations can be used for risk stratification during preclinical AD to detect and delay the progression into dementia.
Collapse
Affiliation(s)
- Sijie Tan
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232
| | - Wen Han Tong
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232
| | - Ajai Vyas
- School of Biological Sciences, Nanyang Technological University, Singapore 637551
| |
Collapse
|
5
|
Broadbelt T, Mutlu-Smith M, Carnicero-Senabre D, Saido TC, Saito T, Wang SH. Impairment in novelty-promoted memory via behavioral tagging and capture before apparent memory loss in a knock-in model of Alzheimer's disease. Sci Rep 2022; 12:22298. [PMID: 36566248 PMCID: PMC9789965 DOI: 10.1038/s41598-022-26113-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/09/2022] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) is associated with cognitive impairments and age-dependent memory deficits which have been studied using genetic models of AD. Whether the processes for modulating memory persistence are more vulnerable to the influence of amyloid pathology than the encoding and consolidation of the memory remains unclear. Here, we investigated whether early amyloid pathology would affect peri-learning novelty in promoting memory, through a process called behavioral tagging and capture (BTC). AppNL-G-F/NL-G-F mice and wild-type littermates were trained in an appetitive delayed matching-to-place (ADMP) task which allows for the assessment of peri-learning novelty in facilitating memory. The results show that novelty enabled intermediate-term memory in wild-type mice, but not in AppNL-G-F/NL-G-F mice in adulthood. This effect preceded spatial memory impairment in the ADMP task seen in middle age. Other memory tests in the Barnes maze, Y-maze, novel object or location recognition tasks remained intact. Together, memory modulation through BTC is impaired before apparent deficits in learning and memory. Relevant biological mechanisms underlying BTC and the implication in AD are discussed.
Collapse
Affiliation(s)
- Tabitha Broadbelt
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Menekse Mutlu-Smith
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Daniel Carnicero-Senabre
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,grid.5515.40000000119578126Present Address: Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
| | - Takaomi C. Saido
- grid.474690.8Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, 351-0198 Japan
| | - Takashi Saito
- grid.260433.00000 0001 0728 1069Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Aichi, 467-8601 Japan
| | - Szu-Han Wang
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| |
Collapse
|
6
|
Lebeau RH, Mendez-David I, Kucynski-Noyau L, Henry C, Attali D, Plaze M, Colle R, Corruble E, Gardier AM, Gaillard R, Guilloux JP, David DJ. Peripheral proteomic changes after electroconvulsive seizures in a rodent model of non-response to chronic fluoxetine. Front Pharmacol 2022; 13:993449. [DOI: 10.3389/fphar.2022.993449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/05/2022] [Indexed: 11/13/2022] Open
Abstract
Major depressive disorder (MDD) is the psychiatric disorder with the highest prevalence in the world. Pharmacological antidepressant treatment (AD), such as selective serotonin reuptake inhibitors [SSRI, i.e., fluoxetine (Flx)] is the first line of treatment for MDD. Despite its efficacy, lack of AD response occurs in numerous patients characterizing Difficult-to-treat Depression. ElectroConvulsive Therapy (ECT) is a highly effective treatment inducing rapid improvement in depressive symptoms and high remission rates of ∼50–63% in patients with pharmaco-resistant depression. Nevertheless, the need to develop reliable treatment response predictors to guide personalized AD strategies and supplement clinical observation is becoming a pressing clinical objective. Here, we propose to establish a proteomic peripheral biomarkers signature of ECT response in an anxio/depressive animal model of non-response to AD. Using an emotionality score based on the analysis complementary behavioral tests of anxiety/depression (Elevated Plus Maze, Novelty Suppressed Feeding, Splash Test), we showed that a 4-week corticosterone treatment (35 μg/ml, Cort model) in C57BL/6JRj male mice induced an anxiety/depressive-like behavior. A 28-day chronic fluoxetine treatment (Flx, 18 mg/kg/day) reduced corticosterone-induced increase in emotional behavior. A 50% decrease in emotionality score threshold before and after Flx, was used to separate Flx-responding mice (Flx-R, n = 18), or Flx non-responder mice (Flx-NR, n = 7). Then, Flx-NR mice received seven sessions of electroconvulsive seizure (ECS, equivalent to ECT in humans) and blood was collected before and after ECS treatment. Chronic ECS normalized the elevated emotionality observed in Flx-NR mice. Then, proteins were extracted from peripheral blood mononuclear cells (PBMCs) and isolated for proteomic analysis using a high-resolution MS Orbitrap. Data are available via ProteomeXchange with identifier PXD037392. The proteomic analysis revealed a signature of 33 peripheral proteins associated with response to ECS (7 down and 26 upregulated). These proteins were previously associated with mental disorders and involved in regulating pathways which participate to the depressive disorder etiology.
Collapse
|
7
|
Mehla J, Deibel SH, Karem H, Hossain S, Lacoursiere SG, Sutherland RJ, Mohajerani MH, McDonald RJ. Dramatic impacts on brain pathology, anxiety, and cognitive function in the knock-in APPNL-G-F mouse model of Alzheimer disease following long-term voluntary exercise. Alzheimers Res Ther 2022; 14:143. [PMID: 36180883 PMCID: PMC9526288 DOI: 10.1186/s13195-022-01085-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/13/2022] [Indexed: 11/22/2022]
Abstract
Background An active lifestyle is associated with improved cognitive functions in aged people and may prevent or slow down the progression of various neurodegenerative diseases including Alzheimer’s disease (AD). To investigate these protective effects, male APPNL-G-F mice were exposed to long-term voluntary exercise. Methods Three-month-old AD mice were housed in a cage supplemented with a running wheel for 9 months for long-term exercise. At the age of 12 months, behavioral tests were completed for all groups. After completing behavioral testing, their brains were assessed for amyloid pathology, microgliosis, and cholinergic cells. Results The results showed that APPNL-G-F mice allowed to voluntarily exercise showed an improvement in cognitive functions. Furthermore, long-term exercise also improved anxiety in APPNL-G-F mice as assessed by measuring thigmotaxis in the Morris water task. We also found reductions in amyloid load and microgliosis, and a preservation of cholinergic cells in the brain of APPNL-G-F mice allowed to exercise in their home cages. These profound reductions in brain pathology associated with AD are likely responsible for the observed improvement of learning and memory functions following extensive and regular exercise. Conclusion These findings suggest the potential of physical exercise to mitigate the cognitive deficits in AD.
Collapse
|
8
|
Error-prone protein synthesis recapitulates early symptoms of Alzheimer disease in aging mice. Cell Rep 2022; 40:111433. [PMID: 36170830 DOI: 10.1016/j.celrep.2022.111433] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/19/2022] [Accepted: 09/08/2022] [Indexed: 11/20/2022] Open
Abstract
Age-related neurodegenerative diseases (NDDs) are associated with the aggregation and propagation of specific pathogenic protein species (e.g., Aβ, α-synuclein). However, whether disruption of synaptic homeostasis results from protein misfolding per se rather than accumulation of a specific rogue protein is an unexplored question. Here, we show that error-prone translation, with its frequent outcome of random protein misfolding, is sufficient to recapitulate many early features of NDDs, including perturbed Ca2+ signaling, neuronal hyperexcitability, and mitochondrial dysfunction. Mice expressing the ribosomal ambiguity mutation Rps9 D95N exhibited disrupted synaptic homeostasis resulting in behavioral changes reminiscent of early Alzheimer disease (AD), such as learning and memory deficits, maladaptive emotional responses, epileptiform discharges, suppressed circadian rhythmicity, and sleep fragmentation, accompanied by hippocampal NPY expression and cerebral glucose hypometabolism. Collectively, our findings suggest that random protein misfolding may contribute to the pathogenesis of age-related NDDs, providing an alternative framework for understanding the initiation of AD.
Collapse
|
9
|
Wang S, Ichinomiya T, Savchenko P, Devulapalli S, Wang D, Beltz G, Saito T, Saido TC, Wagner SL, Patel HH, Head BP. Age-Dependent Behavioral and Metabolic Assessment of App NL-G-F/NL-G-F Knock-in (KI) Mice. Front Mol Neurosci 2022; 15:909989. [PMID: 35966019 PMCID: PMC9373872 DOI: 10.3389/fnmol.2022.909989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/13/2022] [Indexed: 02/03/2023] Open
Abstract
Mitochondria play a crucial role in Alzheimer's disease (AD) onset and progression. Traditional transgenic AD mouse models which were widely used in the past decades share a common limitation: The overexpression of APP and overproduction of amyloid-beta (Aβ) are accompanied by other APP peptide fragments, which could introduce artificial and non-clinically relevant phenotypes. Here, we performed an in-depth and time-resolved behavioral and metabolic characterization of a clinically relevant AD mouse model engineered to express normal physiological levels of APP harboring humanized Swedish (K670N/M671L), Beyreuther/Iberian (I716F), and Arctic (E693G) mutations (App NL-G-F/NL-G-F ), termed APP knock-in (APPKI) mice. Our result showed that APPKI mice exhibited fear learning deficits at 6-m age and contextual memory deficit at 12-m age. Histopathological analysis revealed mild amyloidosis (6E10) accompanied by microgliosis (Iba1) as early as 3 months, which progressed significantly together with significant astrocytosis at 6 and 12 m. We further analyzed hippocampal mitochondrial dysfunction by multiple assays, while 3-m APPKI mice brain mitochondrial function remains a similar level as WT mice. Significant mitochondrial dysfunction characterized by decreased ATP production and higher membrane potential with subsequent overproduction of reactive oxygen species (ROS) was observed in mitochondria isolated from 7-m APPKI mice hippocampal tissue. Morphologically, these mitochondria were larger in volume with a decreased level of mitochondrial fusion protein mitofusin-2 (MFN2). At 12 months, APPKI mice exhibit a significantly decreased total mitochondrial oxygen consumption rate (OCR) in isolated hippocampal mitochondria detected by high-resolution respirometry. These data indicate early mitochondrial dysfunction in the brain at pre-symptomatic age in the App NL-G-F/NL-G-mice, which may play a key role in the progression of the disease. Moreover, the identified behavioral and bioenergetic alterations in this clinically relevant AD mouse model provide a valuable tool to optimize the temporal component for therapeutic interventions to treat AD.
Collapse
Affiliation(s)
- Shanshan Wang
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States,Department of Anesthesia, University of California, San Diego, San Diego, CA, United States
| | - Taiga Ichinomiya
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States,Department of Anesthesia, University of California, San Diego, San Diego, CA, United States,Department of Anesthesiology and Intensive Care Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Paul Savchenko
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States,Department of Anesthesia, University of California, San Diego, San Diego, CA, United States
| | - Swetha Devulapalli
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States,Department of Anesthesia, University of California, San Diego, San Diego, CA, United States
| | - Dongsheng Wang
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States,Department of Anesthesia, University of California, San Diego, San Diego, CA, United States
| | - Gianna Beltz
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States,Department of Anesthesia, University of California, San Diego, San Diego, CA, United States
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan
| | - Steve L. Wagner
- Neurosciences Department, University of California, San Diego, San Diego, CA, United States
| | - Hemal H. Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States,Department of Anesthesia, University of California, San Diego, San Diego, CA, United States
| | - Brian P. Head
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States,Department of Anesthesia, University of California, San Diego, San Diego, CA, United States,*Correspondence: Brian P. Head
| |
Collapse
|
10
|
Tok S, Maurin H, Delay C, Crauwels D, Manyakov NV, Van Der Elst W, Moechars D, Drinkenburg WHIM. Pathological and neurophysiological outcomes of seeding human-derived tau pathology in the APP-KI NL-G-F and NL-NL mouse models of Alzheimer's Disease. Acta Neuropathol Commun 2022; 10:92. [PMID: 35739575 PMCID: PMC9219251 DOI: 10.1186/s40478-022-01393-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
The two main histopathological hallmarks that characterize Alzheimer’s Disease are the presence of amyloid plaques and neurofibrillary tangles. One of the current approaches to studying the consequences of amyloid pathology relies on the usage of transgenic animal models that incorporate the mutant humanized form of the amyloid precursor protein (hAPP), with animal models progressively developing amyloid pathology as they age. However, these mice models generally overexpress the hAPP protein to facilitate the development of amyloid pathology, which has been suggested to elicit pathological and neuropathological changes unrelated to amyloid pathology. In this current study, we characterized APP knock-in (APP-KI) animals, that do not overexpress hAPP but still develop amyloid pathology to understand the influence of protein overexpression. We also induced tau pathology via human-derived tau seeding material to understand the neurophysiological effects of amyloid and tau pathology. We report that tau-seeded APP-KI animals progressively develop tau pathology, exacerbated by the presence of amyloid pathology. Interestingly, older amyloid-bearing, tau-seeded animals exhibited more amyloid pathology in the entorhinal area, isocortex and hippocampus, but not thalamus, which appeared to correlate with impairments in gamma oscillations before seeding. Tau-seeded animals also featured immediate deficits in power spectra values and phase-amplitude indices in the hippocampus after seeding, with gamma power spectra deficits persisting in younger animals. Both deficits in hippocampal phase-amplitude coupling and gamma power differentiate tau-seeded, amyloid-positive animals from buffer controls. Based on our results, impairments in gamma oscillations appear to be strongly associated with the presence and development of amyloid and tau pathology, and may also be an indicator of neuropathology, network dysfunction, and even potential disposition to the future development of amyloid pathology.
Collapse
Affiliation(s)
- S Tok
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium.,Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - H Maurin
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - C Delay
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - D Crauwels
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - N V Manyakov
- Data Sciences, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - W Van Der Elst
- Quantitative Sciences Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - D Moechars
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - W H I M Drinkenburg
- Department of Neuroscience, Janssen Research and Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium. .,Groningen Institute for Evolutionary Life Sciences, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
11
|
Theta and gamma oscillatory dynamics in mouse models of Alzheimer's disease: A path to prospective therapeutic intervention. Neurosci Biobehav Rev 2022; 136:104628. [PMID: 35331816 DOI: 10.1016/j.neubiorev.2022.104628] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 12/26/2022]
Abstract
Understanding the neural basis of cognitive deficits, a key feature of Alzheimer's disease (AD), is imperative for achieving the therapy of the disease. Rhythmic oscillatory activities in neural systems are a fundamental mechanism for diverse brain functions, including cognition. In several neurological conditions like AD, aberrant neural oscillations have been shown to play a central role. Furthermore, manipulation of brain oscillations in animals has confirmed their impact on cognition and disease. In this article, we review the evidence from mouse models that shows how synchronized oscillatory activity is intricately linked to AD machinery. We primarily focus on recent reports showing abnormal oscillatory activities at theta and gamma frequencies in AD condition and their influence on cellular disturbances and cognitive impairments. A thorough comprehension of the role that neuronal oscillations play in AD pathology should pave the way to therapeutic interventions that can curb the disease.
Collapse
|
12
|
Niikura R, Miyazaki T, Takase K, Sasaguri H, Saito T, Saido TC, Goto T. Assessments of prolonged effects of desflurane and sevoflurane on motor learning deficits in aged App NL-G-F/NL-G-F mice. Mol Brain 2022; 15:32. [PMID: 35387663 PMCID: PMC8988377 DOI: 10.1186/s13041-022-00910-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/16/2022] [Indexed: 11/10/2022] Open
Abstract
As the proportion of elderly in society increases, so do the number of older patients undergoing surgical procedures. This is concerning as exposure to anesthesia has been identified as a risk factor for Alzheimer's disease (AD). However, the causal relationship between clinical AD development and anesthesia remains conjectural. Preclinical studies have demonstrated that anesthesia, such as halothane, isoflurane, and sevoflurane, induces AD-like pathophysiological changes and cognitive impairments in transgenic mouse models of AD. Desflurane does not have these effects and is expected to have more potential for use in elderly patients, yet little is known about its effects, especially on non-cognitive functions, such as motor and emotional functions. Thus, we examined the postanesthetic effects of desflurane and sevoflurane on motor and emotional function in aged AppNL-G-F/NL-G-F (App-KI) mice. This is a recently developed transgenic mouse model of AD exhibiting amyloid β peptide (Aβ) amyloidosis and a neuroinflammatory response in an age-dependent manner without non-physiological amyloid precursor protein (APP) overexpression. Mice were subjected to a short behavioral test battery consisting of an elevated plus maze, a balance beam test, and a tail suspension test seven days after exposure to 8.0% desflurane for 6 h or 2.8% sevoflurane for 2 h. App-KI mice showed significant increments in the percentage of entry and time spent in open arms in the elevated plus maze, increments in the number of slips and latency to traverse for the balance beam test, increments in the limb clasping score, increments in immobile duration, and decrements in latency to first immobile episode for the tail suspension test compared to age-matched wild type (WT) controls. Desflurane- and sevoflurane-exposed App-KI mice showed a delayed decrement in the number of slips for each trial in the balance beam test, while air-treated App-KI mice rapidly improved their performance, and increased their clasping behavior in the tail suspension test. Furthermore, App-KI inhibited the change in membrane GluA3 following exposure to anesthetics in the cerebellum. These results suggest high validity of App-KI mice as an animal model of AD.
Collapse
Affiliation(s)
- Ryo Niikura
- Department of Anesthesiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoyuki Miyazaki
- Department of Anesthesiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan. .,Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Kenkichi Takase
- Laboratory of Psychology, Jichi Medical University School of Medicine, Simotsuke, Tochigi, Japan.
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, Japan.,Department of Neurocognitive Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Takahisa Goto
- Department of Anesthesiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
13
|
MUTYH Actively Contributes to Microglial Activation and Impaired Neurogenesis in the Pathogenesis of Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8635088. [PMID: 34970419 PMCID: PMC8714343 DOI: 10.1155/2021/8635088] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/29/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022]
Abstract
Oxidative stress is a major risk factor for Alzheimer's disease (AD), which is characterized by brain atrophy, amyloid plaques, neurofibrillary tangles, and loss of neurons. 8-Oxoguanine, a major oxidatively generated nucleobase highly accumulated in the AD brain, is known to cause neurodegeneration. In mammalian cells, several enzymes play essential roles in minimizing the 8-oxoguanine accumulation in DNA. MUTYH with adenine DNA glycosylase activity excises adenine inserted opposite 8-oxoguanine in DNA. MUTYH is reported to actively contribute to the neurodegenerative process in Parkinson and Huntington diseases and some mouse models of neurodegenerative diseases by accelerating neuronal dysfunction and microgliosis under oxidative conditions; however, whether or not MUTYH is involved in AD pathogenesis remains unclear. In the present study, we examined the contribution of MUTYH to the AD pathogenesis. Using postmortem human brains, we showed that various types of MUTYH transcripts and proteins are expressed in most hippocampal neurons and glia in both non-AD and AD brains. We further introduced MUTYH deficiency into App NL-G-F/NL-G-F knock-in AD model mice, which produce humanized toxic amyloid-β without the overexpression of APP protein, and investigated the effects of MUTYH deficiency on the behavior, pathology, gene expression, and neurogenesis. MUTYH deficiency improved memory impairment in App NL-G-F/NL-G-F mice, accompanied by reduced microgliosis. Gene expression profiling strongly suggested that MUTYH is involved in the microglial response pathways under AD pathology and contributes to the phagocytic activity of disease-associated microglia. We also found that MUTYH deficiency ameliorates impaired neurogenesis in the hippocampus, thus improving memory impairment. In conclusion, we propose that MUTYH, which is expressed in the hippocampus of AD patients as well as non-AD subjects, actively contributes to memory impairment by inducing microgliosis with poor neurogenesis in the preclinical AD phase and that MUTYH is a novel therapeutic target for AD, as its deficiency is highly beneficial for ameliorating AD pathogenesis.
Collapse
|
14
|
Hashemi-Firouzi N, Shahidi S, Soleimani Asl S. Chronic stimulation of the serotonergic 5-HT4 receptor modulates amyloid-beta-related impairments in synaptic plasticity and memory deficits in male rats. Brain Res 2021; 1773:147701. [PMID: 34695393 DOI: 10.1016/j.brainres.2021.147701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by memory decline and impaired hippocampal synaptic plasticity. The serotonin 5-HT4 receptor is involved in learning and memory processes. This study explored the effects of chronic stimulation of 5-HT4R on cognition, memory, long-term potentiation (LTP), paired-pulse ratio (PPR), and neuronal apoptosis in a rat model of amyloid-beta (Aβ)-induced AD. Thirty-five male Wistar rats were randomly divided into three groups as follows: the sham, Aβ, and Aβ + BIMU8 groups. Aβ (6 µg/µl) was administrated by intracerebroventricular (icv) injection. The animals were treated with BIMU8 (1 μg/μL, ICV) as a 5-HT4R agonist for 30 days. Memory and behavioral changes were assessed by the passive avoidance learning, novel object recognition, open field, and elevated plus maze tests. Hippocampal synaptic plasticity was evaluated in the dentate gyrus (DG) in response to the stimulation applied to the perforant pathway. Furthermore, neuronal apoptosis was measured in the hippocampus. Data were analyzed by SPSS version 19 using one-way ANOVA, followed by Tukey's post hoc test. Aβ induced memory deficits and neuronal loss and inhibited LTP induction. Aβ also increased the normalized PPR. BIMU8 enhanced the slope of the field excitatory postsynaptic potential in LTP and improved cognition behavior. Paired-pulse inhibition or facilitation was not affected by LTP induction in Aβ animals receiving the BIMU8. It can be concluded that the stimulation of the 5-HT4 receptor modulated the Aβ-induced cognition and memory deficits, probably via a decrease in the hippocampal apoptotic neurons and an improvement in the hippocampal synaptic functions without involving its inhibitory interneurons.
Collapse
Affiliation(s)
- Nasrin Hashemi-Firouzi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Sara Soleimani Asl
- Anatomy Department, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
15
|
Ni R. Magnetic Resonance Imaging in Animal Models of Alzheimer's Disease Amyloidosis. Int J Mol Sci 2021; 22:12768. [PMID: 34884573 PMCID: PMC8657987 DOI: 10.3390/ijms222312768] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Amyloid-beta (Aβ) plays an important role in the pathogenesis of Alzheimer's disease. Aberrant Aβ accumulation induces neuroinflammation, cerebrovascular alterations, and synaptic deficits, leading to cognitive impairment. Animal models recapitulating the Aβ pathology, such as transgenic, knock-in mouse and rat models, have facilitated the understanding of disease mechanisms and the development of therapeutics targeting Aβ. There is a rapid advance in high-field MRI in small animals. Versatile high-field magnetic resonance imaging (MRI) sequences, such as diffusion tensor imaging, arterial spin labeling, resting-state functional MRI, anatomical MRI, and MR spectroscopy, as well as contrast agents, have been developed for preclinical imaging in animal models. These tools have enabled high-resolution in vivo structural, functional, and molecular readouts with a whole-brain field of view. MRI has been used to visualize non-invasively the Aβ deposits, synaptic deficits, regional brain atrophy, impairment in white matter integrity, functional connectivity, and cerebrovascular and glymphatic system in animal models of Alzheimer's disease amyloidosis. Many of the readouts are translational toward clinical MRI applications in patients with Alzheimer's disease. In this review, we summarize the recent advances in MRI for visualizing the pathophysiology in amyloidosis animal models. We discuss the outstanding challenges in brain imaging using MRI in small animals and propose future outlook in visualizing Aβ-related alterations in the brains of animal models.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
16
|
Arroyo-García LE, Isla AG, Andrade-Talavera Y, Balleza-Tapia H, Loera-Valencia R, Alvarez-Jimenez L, Pizzirusso G, Tambaro S, Nilsson P, Fisahn A. Impaired spike-gamma coupling of area CA3 fast-spiking interneurons as the earliest functional impairment in the App NL-G-F mouse model of Alzheimer's disease. Mol Psychiatry 2021; 26:5557-5567. [PMID: 34385602 PMCID: PMC8758494 DOI: 10.1038/s41380-021-01257-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/19/2021] [Accepted: 07/30/2021] [Indexed: 02/07/2023]
Abstract
In Alzheimer's disease (AD) the accumulation of amyloid-β (Aβ) correlates with degradation of cognition-relevant gamma oscillations. The gamma rhythm relies on proper neuronal spike-gamma coupling, specifically of fast-spiking interneurons (FSN). Here we tested the hypothesis that decrease in gamma power and FSN synchrony precede amyloid plaque deposition and cognitive impairment in AppNL-G-F knock-in mice (AppNL-G-F). The aim of the study was to evaluate the amyloidogenic pathology progression in the novel AppNL-G-F mouse model using in vitro electrophysiological network analysis. Using patch clamp of FSNs and pyramidal cells (PCs) with simultaneous gamma oscillation recordings, we compared the activity of the hippocampal network of wild-type mice (WT) and the AppNL-G-F mice at four disease stages (1, 2, 4, and 6 months of age). We found a severe degradation of gamma oscillation power that is independent of, and precedes Aβ plaque formation, and the cognitive impairment reported previously in this animal model. The degradation correlates with increased Aβ1-42 concentration in the brain. Analysis on the cellular level showed an impaired spike-gamma coupling of FSN from 2 months of age that correlates with the degradation of gamma oscillations. From 6 months of age PC firing becomes desynchronized also, correlating with reports in the literature of robust Aβ plaque pathology and cognitive impairment in the AppNL-G-F mice. This study provides evidence that impaired FSN spike-gamma coupling is one of the earliest functional impairment caused by the amyloidogenic pathology progression likely is the main cause for the degradation of gamma oscillations and consequent cognitive impairment. Our data suggests that therapeutic approaches should be aimed at restoring normal FSN spike-gamma coupling and not just removal of Aβ.
Collapse
Affiliation(s)
- Luis Enrique Arroyo-García
- Neuronal Oscillations Laboratory; Division of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden.
| | - Arturo G Isla
- Neuronal Oscillations Laboratory; Division of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Yuniesky Andrade-Talavera
- Neuronal Oscillations Laboratory; Division of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Hugo Balleza-Tapia
- Neuronal Oscillations Laboratory; Division of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Raúl Loera-Valencia
- Division of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Laura Alvarez-Jimenez
- Division of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Giusy Pizzirusso
- Neuronal Oscillations Laboratory; Division of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Simone Tambaro
- Division of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Per Nilsson
- Division of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - André Fisahn
- Neuronal Oscillations Laboratory; Division of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
17
|
Kaur H, Nookala S, Singh S, Mukundan S, Nagamoto-Combs K, Combs CK. Sex-Dependent Effects of Intestinal Microbiome Manipulation in a Mouse Model of Alzheimer's Disease. Cells 2021; 10:2370. [PMID: 34572019 PMCID: PMC8469717 DOI: 10.3390/cells10092370] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/21/2022] Open
Abstract
Mechanisms linking intestinal bacteria and neurodegenerative diseases such as Alzheimer's disease (AD) are still unclear. We hypothesized that intestinal dysbiosis might potentiate AD, and manipulating the microbiome to promote intestinal eubiosis and immune homeostasis may improve AD-related brain changes. This study assessed sex differences in the effects of oral probiotic, antibiotics, and synbiotic treatments in the AppNL-G-F mouse model of AD. The fecal microbiome demonstrated significant correlations between bacterial genera in AppNL-G-F mice and Aβ plaque load, gliosis, and memory performance. Female and not male AppNL-G-F mice fed probiotic but not synbiotic exhibited a decrease in Aβ plaques, microgliosis, brain TNF-α, and memory improvement compared to no treatment controls. Although antibiotics treatment did not produce these multiple changes in brain cytokines, memory, or gliosis, it did decrease Aβ plaque load and colon cytokines in AppNL-G-F males. The intestinal cytokine milieu and splenocyte phenotype of female but not male AppNL-G-F mice indicated a modest proinflammatory innate response following probiotic treatment compared to controls, with an adaptive response following antibiotics treatment in male AppNL-G-F mice. Overall, these results demonstrate the beneficial effects of probiotic only in AppNL-G-F females, with minimal benefits of antibiotics or synbiotic feeding in male or female mice.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA; (S.N.); (S.M.); (K.N.-C.)
| | - Suba Nookala
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA; (S.N.); (S.M.); (K.N.-C.)
| | - Surjeet Singh
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience (CCBN), University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada;
| | - Santhosh Mukundan
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA; (S.N.); (S.M.); (K.N.-C.)
| | - Kumi Nagamoto-Combs
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA; (S.N.); (S.M.); (K.N.-C.)
| | - Colin Kelly Combs
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA; (S.N.); (S.M.); (K.N.-C.)
| |
Collapse
|
18
|
Degawa T, Kawahata I, Izumi H, Shinoda Y, Fukunaga K. T-type Ca 2+ channel enhancer SAK3 administration improves the BPSD-like behaviors in App NL-G-F/NL-G-F knock-in mice. J Pharmacol Sci 2021; 146:1-9. [PMID: 33858649 DOI: 10.1016/j.jphs.2021.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 02/13/2021] [Accepted: 02/22/2021] [Indexed: 11/24/2022] Open
Abstract
Alzheimer's disease (AD) accounts for the majority of dementia among the elderly. In addition to cognitive impairment, behavioral and psychological symptoms (BPSD) such as depression tendency and increased aggression impose a great burden on the patient. However, there is still no rational therapeutic drug for BPSD. Recently, we developed a novel AD therapeutic candidate, SAK3, and demonstrated that it improved cognitive dysfunction in AppNL-G-F/NL-G-F knock-in (NL-G-F) mice. In this study, we investigated whether acute SAK3 administration improved BPSD in addition to cognitive improvement. Acute SAK3 administration improved BPSD, including anxiolytic and depressive-like behaviors, and ameliorated aggressive behaviors. Furthermore, continuous SAK3 administration improved anxiolytic and depressive-like behaviors. Intriguingly, the anti-anxiolytic and cognitive improvement lasted two weeks after the withdrawal of SAK3, whereas the anti-depressive action did not. Taken together, SAK3 had comprehensive beneficial effects on BPSD behavior.
Collapse
Affiliation(s)
- Tomohide Degawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ichiro Kawahata
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hisanao Izumi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
19
|
Kundu P, Torres ERS, Stagaman K, Kasschau K, Okhovat M, Holden S, Ward S, Nevonen KA, Davis BA, Saito T, Saido TC, Carbone L, Sharpton TJ, Raber J. Integrated analysis of behavioral, epigenetic, and gut microbiome analyses in App NL-G-F, App NL-F, and wild type mice. Sci Rep 2021; 11:4678. [PMID: 33633159 PMCID: PMC7907263 DOI: 10.1038/s41598-021-83851-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/09/2021] [Indexed: 12/19/2022] Open
Abstract
Epigenetic mechanisms occurring in the brain as well as alterations in the gut microbiome composition might contribute to Alzheimer’s disease (AD). Human amyloid precursor protein knock-in (KI) mice contain the Swedish and Iberian mutations (AppNL-F) or those two and also the Arctic mutation (AppNL-G-F). In this study, we assessed whether behavioral and cognitive performance in 6-month-old AppNL-F, AppNL-G-F, and C57BL/6J wild-type (WT) mice was associated with the gut microbiome, and whether the genotype modulates this association. The genotype effects observed in behavioral tests were test-dependent. The biodiversity and composition of the gut microbiome linked to various aspects of mouse behavioral and cognitive performance but differences in genotype modulated these relationships. These genotype-dependent associations include members of the Lachnospiraceae and Ruminococcaceae families. In a subset of female mice, we assessed DNA methylation in the hippocampus and investigated whether alterations in hippocampal DNA methylation were associated with the gut microbiome. Among other differentially methylated regions, we identified a 1 Kb region that overlapped ing 3′UTR of the Tomm40 gene and the promoter region of the Apoe gene that and was significantly more methylated in the hippocampus of AppNL-G-F than WT mice. The integrated gut microbiome hippocampal DNA methylation analysis revealed a positive relationship between amplicon sequence variants (ASVs) within the Lachnospiraceae family and methylation at the Apoe gene. Hence, these microbes may elicit an impact on AD-relevant behavioral and cognitive performance via epigenetic changes in AD-susceptibility genes in neural tissue or that such changes in the epigenome can elicit alterations in intestinal physiology that affect the growth of these taxa in the gut microbiome.
Collapse
Affiliation(s)
- Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Eileen Ruth S Torres
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Keaton Stagaman
- Department of Microbiology, Oregon State University, Corvallis, OR, 97331, USA
| | - Kristin Kasschau
- Department of Microbiology, Oregon State University, Corvallis, OR, 97331, USA
| | - Mariam Okhovat
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Sarah Holden
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Samantha Ward
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kimberly A Nevonen
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Brett A Davis
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Lucia Carbone
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA.,Departments of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, 97239, USA.,Departments of Medical Informatics and Clinical Epidemiology, Portland, OR, 97239, USA.,Division of Genetics, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Thomas J Sharpton
- Department of Microbiology, Oregon State University, Corvallis, OR, 97331, USA.,Department of Statistics, Oregon State University, Corvallis, OR, 97331, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA. .,Departments of Neurology, Psychiatry, and Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health & Science University, Portland, OR, 97239, USA. .,College of Pharmacy, Oregon State University, Corvallis, OR, 97331, USA. .,Department of Behavioral Neuroscience, L470, Oregon Health & Science University, 3181SW Sam Jackson Park Road, Portland, OR, 97239, USA.
| |
Collapse
|
20
|
Locci A, Orellana H, Rodriguez G, Gottliebson M, McClarty B, Dominguez S, Keszycki R, Dong H. Comparison of memory, affective behavior, and neuropathology in APP NLGF knock-in mice to 5xFAD and APP/PS1 mice. Behav Brain Res 2021; 404:113192. [PMID: 33607163 DOI: 10.1016/j.bbr.2021.113192] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 01/26/2021] [Accepted: 02/14/2021] [Indexed: 02/08/2023]
Abstract
Transgenic mouse models of Aβ amyloidosis generated by knock-in of a humanized Aβ sequence can offer some advantages over the transgenic models that overexpress amyloid precursor protein (APP). However, systematic comparison of memory, behavioral, and neuropathological phenotypes between these models has not been well documented. In this study, we compared memory and affective behavior in APPNLGF mice, an APP knock-in model, to two widely used mouse models of Alzheimer's disease, 5xFAD and APP/PS1 mice, at 10 months of age. We found that, despite similar deficits in working memory, object recognition, and social recognition memory, APPNLGF and 5xFAD mice but not APP/PS1 mice show compelling anxiety- and depressive-like behavior, and exhibited a marked impairment of social interaction. We quantified corticolimbic Aβ plaques, which were lowest in APPNLGF, intermediate in APP/PS1, and highest in 5xFAD mice. Interestingly, analysis of plaque size revealed that plaques were largest in APP/PS1 mice, intermediate in 5xFAD mice, and smallest in APPNLGF mice. Finally, we observed a significantly higher percentage of the area occupied by plaques in both 5xFAD and APP/PS1 relative to APPNLGF mice. Overall, our findings suggest that the severity of Aβ neuropathology is not directly correlated with memory and affective behavior impairments between these three transgenic mouse models. Additionally, APPNLGF may represent a valid mouse model for studying AD comorbid with anxiety and depression.
Collapse
Affiliation(s)
- Andrea Locci
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Hector Orellana
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Guadalupe Rodriguez
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Meredith Gottliebson
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Bryan McClarty
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sky Dominguez
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Rachel Keszycki
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
21
|
Jia Y, Zou J, Wang Y, Zhang X, Shi Y, Liang Y, Guo D, Yang M. Action mechanism of Roman chamomile in the treatment of anxiety disorder based on network pharmacology. J Food Biochem 2020; 45:e13547. [PMID: 33152801 DOI: 10.1111/jfbc.13547] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 11/25/2022]
Abstract
Anxiety disorder is a common psychiatric disease. Roman chamomile as medicine or tea has long been used as a mild tranquilizer to reduce anxiety, but the mechanism is unclear. This research is based on network pharmacology combined with database mining to find the ingredients, action pathways and key targets of Roman chamomile for the treatment of anxiety. About 126 common targets related to chamomile and anxiety were obtained, and these targets were involved in 56 KEGG pathways. GEO screened LRRK2 as a key protein, and molecular docking showed that the protein could stably bind to drug components. Roman chamomile has the characteristics of multi-target and multi-pathway in the treatment of anxiety disorder. Its possible mechanism is to intervene anxiety disorder in the process of disease development, such as neuroactive ligand-receptor interaction, serotonin synapse, and cAMP signaling pathway. LRRK2 may be an important gene for Roman chamomile in the treatment of anxiety disorder. PRACTICAL APPLICATIONS: Roman chamomile is well known for its use in medicine and tea making. It contains many nutrients, which can relieve people's anxiety, help sleep, antibacterial and anti-inflammatory. In this article, through network pharmacology combined with Gene Expression Omnibus data mining and molecular docking, the target and mechanism of Roman chamomile in the treatment of anxiety were discussed, and its efficacy was verified by model animals, which not only clarified its mechanism at the systematic level, but also proved to be effective at the biological level. It provides a reference for the further development and utilization of Roman chamomile.
Collapse
Affiliation(s)
- Yanzhuo Jia
- Department of Pharmaceutics, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Junbo Zou
- Department of Pharmaceutics, Shaanxi University of Chinese Medicine, Xianyang, China.,Department of Pharmaceutics, The Key Laboratory of Basicand New Drug Research of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yao Wang
- Department of Pharmaceutics, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaofei Zhang
- Department of Pharmaceutics, Shaanxi University of Chinese Medicine, Xianyang, China.,Department of Pharmaceutics, The Key Laboratory of Basicand New Drug Research of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, China.,Department of Pharmaceutics, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yajun Shi
- Department of Pharmaceutics, Shaanxi University of Chinese Medicine, Xianyang, China.,Department of Pharmaceutics, The Key Laboratory of Basicand New Drug Research of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yulin Liang
- Department of Pharmaceutics, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Dongyan Guo
- Department of Pharmaceutics, Shaanxi University of Chinese Medicine, Xianyang, China.,Department of Pharmaceutics, The Key Laboratory of Basicand New Drug Research of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Ming Yang
- Department of Pharmaceutics, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
22
|
Latif-Hernandez A, Sabanov V, Ahmed T, Craessaerts K, Saito T, Saido T, Balschun D. The two faces of synaptic failure in App NL-G-F knock-in mice. Alzheimers Res Ther 2020; 12:100. [PMID: 32838792 PMCID: PMC7445922 DOI: 10.1186/s13195-020-00667-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/10/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Intensive basic and preclinical research into Alzheimer's disease (AD) has yielded important new findings, but they could not yet been translated into effective therapies. One of the reasons is the lack of animal models that sufficiently reproduce the complexity of human AD and the response of human brain circuits to novel treatment approaches. As a step in overcoming these limitations, new App knock-in models have been developed that avoid transgenic APP overexpression and its associated side effects. These mice are proposed to serve as valuable models to examine Aß-related pathology in "preclinical AD." METHODS Since AD as the most common form of dementia progresses into synaptic failure as a major cause of cognitive deficits, the detailed characterization of synaptic dysfunction in these new models is essential. Here, we addressed this by extracellular and whole-cell patch-clamp recordings in AppNL-G-F mice compared to AppNL animals which served as controls. RESULTS We found a beginning synaptic impairment (LTP deficit) at 3-4 months in the prefrontal cortex of AppNL-G-F mice that is further aggravated and extended to the hippocampus at 6-8 months. Measurements of miniature EPSCs and IPSCs point to a marked increase in excitatory and inhibitory presynaptic activity, the latter accompanied by a moderate increase in postsynaptic inhibitory function. CONCLUSIONS Our data reveal a marked impairment of primarily postsynaptic processes at the level of synaptic plasticity but the dominance of a presumably compensatory presynaptic upregulation at the level of elementary miniature synaptic function.
Collapse
Affiliation(s)
- Amira Latif-Hernandez
- Brain and Cognition, KU Leuven, Tiensestraat 102, Box 3714, 3000, Leuven, Belgium
- Present Address: Neurology and Neurological Sciences, Stanford Medicine, Stanford, USA
| | - Victor Sabanov
- Brain and Cognition, KU Leuven, Tiensestraat 102, Box 3714, 3000, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Tariq Ahmed
- Brain and Cognition, KU Leuven, Tiensestraat 102, Box 3714, 3000, Leuven, Belgium
- Present Address: Qatar Biomedical Research Institute, Ar-Rayyan, Qatar
| | - Katleen Craessaerts
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for the Biology of Disease, Leuven, Belgium
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan
- Present Address: Department of Neurocognitive Science, Nagoya City University Graduate School of Medical Science, Nagoya, Aichi, Japan
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan
| | - Detlef Balschun
- Brain and Cognition, KU Leuven, Tiensestraat 102, Box 3714, 3000, Leuven, Belgium.
- Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
23
|
Ferreira S, Pitman KA, Wang S, Summers BS, Bye N, Young KM, Cullen CL. Amyloidosis is associated with thicker myelin and increased oligodendrogenesis in the adult mouse brain. J Neurosci Res 2020; 98:1905-1932. [PMID: 32557778 PMCID: PMC7540704 DOI: 10.1002/jnr.24672] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/03/2020] [Accepted: 05/25/2020] [Indexed: 12/15/2022]
Abstract
In Alzheimer's disease, amyloid plaque formation is associated with the focal death of oligodendrocytes and soluble amyloid β impairs the survival of oligodendrocytes in vitro. However, the response of oligodendrocyte progenitor cells (OPCs) to early amyloid pathology remains unclear. To explore this, we performed a histological, electrophysiological, and behavioral characterization of transgenic mice expressing a pathological form of human amyloid precursor protein (APP), containing three single point mutations associated with the development of familial Alzheimer's disease (PDGFB‐APPSw.Ind, also known as J20 mice). PDGFB‐APPSw.Ind transgenic mice had impaired survival from weaning, were hyperactive by 2 months of age, and developed amyloid plaques by 6 months of age, however, their spatial memory remained intact over this time course. Hippocampal OPC density was normal in P60‐P180 PDGFB‐APPSw.Ind transgenic mice and, by performing whole‐cell patch‐clamp electrophysiology, we found that their membrane properties, including their response to kainate (100 µM), were largely normal. However, by P100, the response of hippocampal OPCs to GABA was elevated in PDGFB‐APPSw.Ind transgenic mice. We also found that the nodes of Ranvier were shorter, the paranodes longer, and the myelin thicker for hippocampal axons in young adult PDGFB‐APPSw.Ind transgenic mice compared with wildtype littermates. Additionally, oligodendrogenesis was normal in young adulthood, but increased in the hippocampus, entorhinal cortex, and fimbria of PDGFB‐APPSw.Ind transgenic mice as pathology developed. As the new oligodendrocytes were not associated with a change in total oligodendrocyte number, these cells are likely required for cell replacement.
Collapse
Affiliation(s)
- Solène Ferreira
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Shiwei Wang
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Benjamin S Summers
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Nicole Bye
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
24
|
White D, de Sousa Abreu RP, Blake A, Murphy J, Showell S, Kitamoto T, Lawal HO. Deficits in the vesicular acetylcholine transporter alter lifespan and behavior in adult Drosophila melanogaster. Neurochem Int 2020; 137:104744. [PMID: 32315665 DOI: 10.1016/j.neuint.2020.104744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 10/24/2022]
Abstract
The neurotransmitter acetylcholine (ACh) is involved in critical organismal functions that include locomotion and cognition. Importantly, alterations in the cholinergic system are a key underlying factor in cognitive defects associated with aging. One essential component of cholinergic synaptic transmission is the vesicular ACh transporter (VAChT), which regulates the packaging of ACh into synaptic vesicles for extracellular release. Mutations that cause a reduction in either protein level or activity lead to diminished locomotion ability whereas complete loss of function of VAChT is lethal. While much is known about the function of VAChT, the direct role of altered ACh release and its association with either an impairment or an enhancement of cognitive function are still not fully understood. We hypothesize that point mutations in Vacht cause age-related deficits in cholinergic-mediated behaviors such as locomotion, and learning and memory. Using Drosophila melanogaster as a model system, we have studied several mutations within Vacht and observed their effect on survivability and locomotive behavior. Here we report for the first time a weak hypomorphic Vacht allele that shows a differential effect on ACh-linked behaviors. We also demonstrate that partially rescued Vacht point mutations cause an allele-dependent deficit in lifespan and defects in locomotion ability. Moreover, using a thorough data analytics strategy to identify exploratory behavioral patterns, we introduce new paradigms for measuring locomotion-related activities that could not be revealed or detected by a simple measure of the average speed alone. Together, our data indicate a role for VAChT in the maintenance of longevity and locomotion abilities in Drosophila and we provide additional measurements of locomotion that can be useful in determining subtle changes in Vacht function on locomotion-related behaviors.
Collapse
Affiliation(s)
- Daniel White
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, 19901, USA
| | - Raquel P de Sousa Abreu
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Andrew Blake
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, 19901, USA
| | - Jeremy Murphy
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, 19901, USA
| | - Shardae Showell
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, 19901, USA
| | - Toshihiro Kitamoto
- Department of Anesthesia, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Hakeem O Lawal
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, 19901, USA.
| |
Collapse
|
25
|
Lewandowski CT, Maldonado Weng J, LaDu MJ. Alzheimer's disease pathology in APOE transgenic mouse models: The Who, What, When, Where, Why, and How. Neurobiol Dis 2020; 139:104811. [PMID: 32087290 DOI: 10.1016/j.nbd.2020.104811] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/01/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
The focus on amyloid plaques and neurofibrillary tangles has yielded no Alzheimer's disease (AD) modifying treatments in the past several decades, despite successful studies in preclinical mouse models. This inconsistency has caused a renewed focus on improving the fidelity and reliability of AD mouse models, with disparate views on how this improvement can be accomplished. However, the interactive effects of the universal biological variables of AD, which include age, APOE genotype, and sex, are often overlooked. Age is the greatest risk factor for AD, while the ε4 allele of the human APOE gene, encoding apolipoprotein E, is the greatest genetic risk factor. Sex is the final universal biological variable of AD, as females develop AD at almost twice the rate of males and, importantly, female sex exacerbates the effects of APOE4 on AD risk and rate of cognitive decline. Therefore, this review evaluates the importance of context for understanding the role of APOE in preclinical mouse models. Specifically, we detail how human AD pathology is mirrored in current transgenic mouse models ("What") and describe the critical need for introducing human APOE into these mouse models ("Who"). We next outline different methods for introducing human APOE into mice ("How") and highlight efforts to develop temporally defined and location-specific human apoE expression models ("When" and "Where"). We conclude with the importance of choosing the human APOE mouse model relevant to the question being addressed, using the selection of transgenic models for testing apoE-targeted therapeutics as an example ("Why").
Collapse
Affiliation(s)
- Cutler T Lewandowski
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood St., Chicago, IL 60612, USA.
| | - Juan Maldonado Weng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S. Wood St., Chicago, IL 60612, USA.
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S. Wood St., Chicago, IL 60612, USA.
| |
Collapse
|
26
|
First person – Eleftheria Pervolaraki. Dis Model Mech 2019. [PMCID: PMC6765193 DOI: 10.1242/dmm.042408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
First Person is a series of interviews with the first authors of a selection of papers published in Disease Models & Mechanisms (DMM), helping early-career researchers promote themselves alongside their papers. Eleftheria Pervolaraki is first author on ‘Insoluble Aβ overexpression in an App knock-in mouse model alters microstructure and gamma oscillations in the prefrontal cortex, affecting anxiety-related behaviours’, published in DMM. Eleftheria conducted the research described in this article while a Research Fellow at the School of Biomedical Sciences, University of Leeds, UK. She is now Medical Science Lead for the UK at Cardinal Health, High Wycombe, Buckinghamshire, UK, working on medical devices for the management of cardiac rhythm whilst preventing infections in surgical sites.
Collapse
|