51
|
Attar A, Liu T, Chan WTC, Hayes J, Nejad M, Lei K, Bitan G. A shortened Barnes maze protocol reveals memory deficits at 4-months of age in the triple-transgenic mouse model of Alzheimer's disease. PLoS One 2013; 8:e80355. [PMID: 24236177 PMCID: PMC3827415 DOI: 10.1371/journal.pone.0080355] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/08/2013] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disease that manifests as memory loss, cognitive dysfunction, and dementia. Animal models of Alzheimer's disease have been instrumental in understanding the underlying pathological mechanism and in evaluation of potential therapies. The triple transgenic (3 × Tg) mouse model of AD is unique because it recapitulates both pathologic hallmarks of Alzheimer's disease--amyloid plaques and neurofibrillary tangles. The earliest cognitive deficits in this model have been shown at 6-m of age by most groups, necessitating aging of the mice to this age before initiating evaluation of the cognitive effects of therapies. To assess cognitive deficits in the 3 × Tg mice, originally we employed a typical Barnes maze protocol of 15 training trials, but found no significant deficits in aged mice. Therefore, we shortened the protocol to include only 5 training trials to increase difficulty. We found cognitive deficits using this protocol using mainly measures from the probe day, rather than the training trials. This also decreased the effort involved with data analysis. We compared 3 × Tg and wild-type mice at 4-m- and 15-m of age using both the original, long training, and the short training paradigms. We found that differences in learning between 3 × Tg and wild-type mice disappeared after the 4(th) training trial. Measures of learning and memory on the probe day showed significant differences between 3 × Tg and wild-type mice following the short, 5-training trial protocol but not the long, 15-training trial protocol. Importantly, we detected cognitive dysfunction already at 4-m of age in 3 × Tg mice using the short Barnes-maze protocol. The ability to test learning and memory in 4-m old 3 × Tg mice using a shortened Barnes maze protocol offers considerable time and cost savings and provides support for the utilization of this model at pre-pathology stages for therapeutic studies.
Collapse
Affiliation(s)
- Aida Attar
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tingyu Liu
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Wai-Ting Coco Chan
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jane Hayes
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Mona Nejad
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - KaiChyuan Lei
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Gal Bitan
- Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
52
|
Davis KE, Eacott MJ, Easton A, Gigg J. Episodic-like memory is sensitive to both Alzheimer's-like pathological accumulation and normal ageing processes in mice. Behav Brain Res 2013; 254:73-82. [DOI: 10.1016/j.bbr.2013.03.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 03/04/2013] [Accepted: 03/07/2013] [Indexed: 02/05/2023]
|
53
|
Ryan D, Koss D, Porcu E, Woodcock H, Robinson L, Platt B, Riedel G. Spatial learning impairments in PLB1Triple knock-in Alzheimer mice are task-specific and age-dependent. Cell Mol Life Sci 2013; 70:2603-19. [PMID: 23535719 PMCID: PMC11113905 DOI: 10.1007/s00018-013-1314-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 02/27/2013] [Accepted: 02/28/2013] [Indexed: 12/21/2022]
Abstract
We recently generated an advanced mouse model of Alzheimer's disease (AD) by targeted knock-in of single-copy mutated human amyloid precursor-protein (APP) and tau genes, crossed with a non-symptomatic presenilin (PS1A246E) over-expressing mouse line. These PLB1Triple mice presented with age-dependent and AD-relevant phenotypes. Homozygous PLB1Triple mice aged 4-12 months were assessed here in a battery of spatial learning tasks: Exp.1 radial-arm water maze (spatial reference and working memory) Exp.2 open-field water maze (spatial reference memory); Exp.3 home cage observation system with spatial learning (IntelliCage); Exp.4 spontaneous object recognition (SOR; novel object and spatial object shift). A separate test with high-expression transgenic APP mice matching the design of experiment 1 was also performed. Spatial deficits in PLB1Triple mice were confirmed at 12, but not 4 months in both water maze tasks. PSAPP mice, by contrast, presented with severe yet non-progressive spatial learning deficits already at 4 months. During tests of spatial learning in SOR and IntelliCage, PLB1Triple mice neither acquired the location of the water-rewarded corner, nor recognize novel or spatially shifted objects at 4 months, indicating these protocols to be more sensitive than the water maze. Collectively and in line with AD symptomatology, PLB1Triple mice present with a graded and progressive age-dependent loss of spatial memory that can be revealed by the use of a battery of tasks. With the emergence of subtle deficits progressively increasing in severity, PLB1Triple mice may offer a more patho-physiologically relevant model of dementia than aggressive expression models.
Collapse
Affiliation(s)
- D. Ryan
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - D. Koss
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - E. Porcu
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - H. Woodcock
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - L. Robinson
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - B. Platt
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| | - G. Riedel
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD Scotland, UK
| |
Collapse
|
54
|
Webster SJ, Bachstetter AD, Van Eldik LJ. Comprehensive behavioral characterization of an APP/PS-1 double knock-in mouse model of Alzheimer's disease. Alzheimers Res Ther 2013; 5:28. [PMID: 23705774 PMCID: PMC3706792 DOI: 10.1186/alzrt182] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 04/29/2013] [Accepted: 05/24/2013] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Despite the extensive mechanistic and pathological characterization of the amyloid precursor protein (APP)/presenilin-1 (PS-1) knock-in mouse model of Alzheimer's disease (AD), very little is known about the AD-relevant behavioral deficits in this model. Characterization of the baseline behavioral performance in a variety of functional tasks and identification of the temporal onset of behavioral impairments are important to provide a foundation for future preclinical testing of AD therapeutics. Here we perform a comprehensive behavioral characterization of this model, discuss how the observed behavior correlates with the mechanistic and pathological observations of others, and compare this model with other commonly used AD mouse models. METHODS FOUR DIFFERENT GROUPS OF MICE RANGING ACROSS THE LIFESPAN OF THIS MODEL (TEST GROUPS: 7, 11, 15, and 24 months old) were run in a behavioral test battery consisting of tasks to assess motor function (grip strength, rotor rod, beam walk, open field ambulatory movement), anxiety-related behavior (open field time spent in peripheral zone vs. center zone, elevated plus maze), and cognitive function (novel object recognition, radial arm water maze). RESULTS There were no differences in motor function or anxiety-related behavior between APP/PS-1 knock-in mice and wild-type counterpart mice for any age group. Cognitive deficits in both recognition memory (novel object recognition) and spatial reference memory (radial arm water maze) became apparent for the knock-in animals as the disease progressed. CONCLUSION This is the first reported comprehensive behavioral analysis of the APP/PS1 knock-in mouse model of AD. The lack of motor/coordination deficits or abnormal anxiety levels, coupled with the age/disease-related cognitive decline and high physiological relevance of this model, make it well suited for utilization in preclinical testing of AD-relevant therapeutics.
Collapse
Affiliation(s)
- Scott J Webster
- Sanders-Brown Center on Aging, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
- Department of Anatomy and Neurobiology, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
55
|
Prior M, Dargusch R, Ehren JL, Chiruta C, Schubert D. The neurotrophic compound J147 reverses cognitive impairment in aged Alzheimer's disease mice. ALZHEIMERS RESEARCH & THERAPY 2013; 5:25. [PMID: 23673233 PMCID: PMC3706879 DOI: 10.1186/alzrt179] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 03/01/2013] [Accepted: 03/28/2013] [Indexed: 12/12/2022]
Abstract
Introduction Despite years of research, there are no disease-modifying drugs for Alzheimer's disease (AD), a fatal, age-related neurodegenerative disorder. Screening for potential therapeutics in rodent models of AD has generally relied on testing compounds before pathology is present, thereby modeling disease prevention rather than disease modification. Furthermore, this approach to screening does not reflect the clinical presentation of AD patients which could explain the failure to translate compounds identified as beneficial in animal models to disease modifying compounds in clinical trials. Clearly a better approach to pre-clinical drug screening for AD is required. Methods To more accurately reflect the clinical setting, we used an alternative screening strategy involving the treatment of AD mice at a stage in the disease when pathology is already advanced. Aged (20-month-old) transgenic AD mice (APP/swePS1ΔE9) were fed an exceptionally potent, orally active, memory enhancing and neurotrophic molecule called J147. Cognitive behavioral assays, histology, ELISA and Western blotting were used to assay the effect of J147 on memory, amyloid metabolism and neuroprotective pathways. J147 was also investigated in a scopolamine-induced model of memory impairment in C57Bl/6J mice and compared to donepezil. Details on the pharmacology and safety of J147 are also included. Results Data presented here demonstrate that J147 has the ability to rescue cognitive deficits when administered at a late stage in the disease. The ability of J147 to improve memory in aged AD mice is correlated with its induction of the neurotrophic factors NGF (nerve growth factor) and BDNF (brain derived neurotrophic factor) as well as several BDNF-responsive proteins which are important for learning and memory. The comparison between J147 and donepezil in the scopolamine model showed that while both compounds were comparable at rescuing short term memory, J147 was superior at rescuing spatial memory and a combination of the two worked best for contextual and cued memory. Conclusion J147 is an exciting new compound that is extremely potent, safe in animal studies and orally active. J147 is a potential AD therapeutic due to its ability to provide immediate cognition benefits, and it also has the potential to halt and perhaps reverse disease progression in symptomatic animals as demonstrated in these studies.
Collapse
Affiliation(s)
- Marguerite Prior
- The Salk Institute for Biological Studies, Cellular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Richard Dargusch
- The Salk Institute for Biological Studies, Cellular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jennifer L Ehren
- The Salk Institute for Biological Studies, Cellular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Chandramouli Chiruta
- The Salk Institute for Biological Studies, Cellular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - David Schubert
- The Salk Institute for Biological Studies, Cellular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
56
|
Gerstein H, Hullinger R, Lindstrom MJ, Burger C. A behavioral paradigm to evaluate hippocampal performance in aged rodents for pharmacological and genetic target validation. PLoS One 2013; 8:e62360. [PMID: 23667471 PMCID: PMC3646843 DOI: 10.1371/journal.pone.0062360] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 03/21/2013] [Indexed: 11/19/2022] Open
Abstract
Aged-related cognitive ability is highly variable, ranging from unimpaired to severe impairments. The Morris water maze (a reliable tool for assessing memory) has been used to distinguish aged rodents that are superior learners from those that are learning impaired. This task, however, is not practical for pre- and post-pharmacological treatment, as the memory of the task is long lasting. In contrast, the object location memory task, also a spatial learning paradigm, results in a less robust memory that decays quickly. We demonstrate for the first time how these two paradigms can be used together to assess hippocampal cognitive impairments before and after pharmacological or genetic manipulations in rodents. Rats were first segregated into superior learning and learning impaired groups using the object location memory task, and their performance was correlated with future outcome on this task and on the Morris water maze. This method provides a tool to evaluate the effect of treatments on cognitive impairment associated with aging and neurodegenerative disorders.
Collapse
Affiliation(s)
- Hilary Gerstein
- Department of Neurology, University of Wisconsin-Madison, Medical Sciences Center, Madison, Wisconsin, United States of America
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Rikki Hullinger
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mary J. Lindstrom
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Corinna Burger
- Department of Neurology, University of Wisconsin-Madison, Medical Sciences Center, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
57
|
Subaiea GM, Adwan LI, Ahmed AH, Stevens KE, Zawia NH. Short-term treatment with tolfenamic acid improves cognitive functions in Alzheimer's disease mice. Neurobiol Aging 2013; 34:2421-30. [PMID: 23639209 DOI: 10.1016/j.neurobiolaging.2013.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/25/2013] [Accepted: 04/01/2013] [Indexed: 10/26/2022]
Abstract
Tolfenamic acid lowers the levels of the amyloid precursor protein (APP) and amyloid beta (Aβ) when administered to C57BL/6 mice by lowering their transcriptional regulator specificity protein 1 (SP1). To determine whether changes upstream in the amyloidogenic pathway that forms Aβ plaques would improve cognitive outcomes, we administered tolfenamic acid for 34 days to hemizygous R1.40 transgenic mice. After the characterization of cognitive deficits in these mice, assessment of spatial learning and memory functions revealed that treatment with tolfenamic acid attenuated long-term memory and working memory deficits, determined using Morris water maze and the Y-maze. These improvements occurred within a shorter period of exposure than that seen with clinically approved drugs. Cognitive enhancement was accompanied by reduction in the levels of the SP1 protein (but not messenger RNA [mRNA]), followed by lowering both the mRNA and the protein levels of APP and subsequent Aβ levels. These findings provide evidence that tolfenamic acid can disrupt the pathologic processes associated with Alzheimer's disease (AD) and are relevant to its scheduled biomarker study in AD patients.
Collapse
Affiliation(s)
- Gehad M Subaiea
- Neurodegeneration Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | | | | | | | | |
Collapse
|
58
|
Parrella E, Maxim T, Maialetti F, Zhang L, Wan J, Wei M, Cohen P, Fontana L, Longo VD. Protein restriction cycles reduce IGF-1 and phosphorylated Tau, and improve behavioral performance in an Alzheimer's disease mouse model. Aging Cell 2013; 12:257-68. [PMID: 23362919 PMCID: PMC3982836 DOI: 10.1111/acel.12049] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2013] [Indexed: 12/21/2022] Open
Abstract
In laboratory animals, calorie restriction (CR) protects against aging, oxidative stress, and neurodegenerative pathologies. Reduced levels of growth hormone and IGF-1, which mediate some of the protective effects of CR, can also extend longevity and/or protect against age-related diseases in rodents and humans. However, severely restricted diets are difficult to maintain and are associated with chronically low weight and other major side effects. Here we show that 4 months of periodic protein restriction cycles (PRCs) with supplementation of nonessential amino acids in mice already displaying significant cognitive impairment and Alzheimer's disease (AD)-like pathology reduced circulating IGF-1 levels by 30-70% and caused an 8-fold increase in IGFBP-1. Whereas PRCs did not affect the levels of β amyloid (Aβ), they decreased tau phosphorylation in the hippocampus and alleviated the age-dependent impairment in cognitive performance. These results indicate that periodic protein restriction cycles without CR can promote changes in circulating growth factors and tau phosphorylation associated with protection against age-related neuropathologies.
Collapse
Affiliation(s)
- Edoardo Parrella
- Longevity Institute, Davis School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Tom Maxim
- Longevity Institute, Davis School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | | | - Lu Zhang
- Longevity Institute, Davis School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Junxiang Wan
- Longevity Institute, Davis School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Min Wei
- Longevity Institute, Davis School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Pinchas Cohen
- Longevity Institute, Davis School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Luigi Fontana
- Division of Geriatrics and Nutritional Science, Washington University in St. Louis St. Louis, MO, USA
- Department of Medicine, Salerno University School of Medicine, Salerno, Italy
- Healthy Aging Platform, CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Valter D. Longo
- Longevity Institute, Davis School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
59
|
Dassie E, Andrews MR, Bensadoun JC, Cacquevel M, Schneider BL, Aebischer P, Wouters FS, Richardson JC, Hussain I, Howlett DR, Spillantini MG, Fawcett JW. Focal expression of adeno-associated viral-mutant tau induces widespread impairment in an APP mouse model. Neurobiol Aging 2012; 34:1355-68. [PMID: 23273572 DOI: 10.1016/j.neurobiolaging.2012.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 10/03/2012] [Accepted: 11/22/2012] [Indexed: 12/28/2022]
Abstract
Adeno-associated virus serotype 6 (AAV6) viral vectors encoding mutant and normal tau were used to produce focal tau pathology. Two mutant forms of tau were used; the P301S tau mutation is associated with neurofibrillary tangle formation in humans, and the 3PO mutation leads to rapid tau aggregation and is associated with pathogenic phosphorylation and cytotoxicity in vitro. We show that adeno-associated viral injection into entorhinal cortex of normal and tau knockout animals leads to local overexpression of tau, and the presence of human tau in axons projecting to and emanating from the entorhinal cortex. Starting at 2 months and increasing by 6 months post-injection neurons expressing mutant tau developed hyperphosphorylated tau pathology, in addition to dystrophic neurites. There was neuronal loss in tau-expressing regions, which was similar in normal and in TASTPM mice injected with mutant tau. There was neuroinflammation around plaques, and in regions expressing mutant tau. We saw no evidence that mutant tau had affected amyloid-beta pathology or vice versa. Morris water maze behavioral tests demonstrated mild memory impairment attributable to amyloid-beta pathology at 2 and 4 months, with severe impairment at 6 months in animals receiving adeno-associated viral-3PO. Therefore, TASTPM mice injected with mutant tau displayed many of the main features characteristic of human Alzheimer's disease patients and might be used as a model to test new drugs to ameliorate clinical features of Alzheimer's disease.
Collapse
Affiliation(s)
- Elisa Dassie
- Department of Clinical Neurosciences, Cambridge University Centre for Brain Repair, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Lalonde R, Fukuchi KI, Strazielle C. Neurologic and motor dysfunctions in APP transgenic mice. Rev Neurosci 2012; 23:363-79. [PMID: 23089603 DOI: 10.1515/revneuro-2012-0041] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 05/02/2012] [Indexed: 12/29/2022]
Abstract
The discovery of gene mutations underlying autosomal dominant Alzheimer's disease has enabled researchers to reproduce several hallmarks of this disorder in transgenic mice, notably the formation of Aβ plaques in brain and cognitive deficits. APP transgenic mutants have also been investigated with respect to survival rates, neurologic functions, and motor coordination, which are all susceptible to alteration in Alzheimer dementia. Several transgenic lines expressing human mutated or wild-type APP had higher mortality rates than non-transgenic controls with or without the presence of Aβ plaques. Mortality rates were also elevated in APP transgenic mice with vascular amyloid accumulation, thereby implicating cerebrovascular factors in the precocious death observed in all APP transgenic models. In addition, myoclonic jumping has been described in APP mutants, together with seizure activity, abnormal limb-flexion and paw-clasping reflexes, and motor coordination deficits. The neurologic signs resemble the myoclonic movements, epileptic seizures, pathological reflexes, and gait problems observed in late-stage Alzheimer's disease.
Collapse
Affiliation(s)
- Robert Lalonde
- Departement de Psychologie, Universite de Rouen, Mont-Saint-Aignan, France.
| | | | | |
Collapse
|
61
|
Foster TC, Defazio RA, Bizon JL. Characterizing cognitive aging of spatial and contextual memory in animal models. Front Aging Neurosci 2012; 4:12. [PMID: 22988436 PMCID: PMC3439636 DOI: 10.3389/fnagi.2012.00012] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 05/15/2012] [Indexed: 11/30/2022] Open
Abstract
Episodic memory, especially memory for contextual or spatial information, is particularly vulnerable to age-related decline in humans and animal models of aging. The continuing improvement of virtual environment technology for testing humans signifies that widely used procedures employed in the animal literature for examining spatial memory could be developed for examining age-related cognitive decline in humans. The current review examines cross species considerations for implementing these tasks and translating findings across different levels of analysis. The specificity of brain systems as well as gaps in linking human and animal laboratory models is discussed.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, Evelyn F. and William L. McKnight Brain Institute, University of Florida Gainesville, FL, USA
| | | | | |
Collapse
|
62
|
Chen SQ, Cai Q, Shen YY, Wang PJ, Teng GJ, Zhang W, Zang FC. Age-related changes in brain metabolites and cognitive function in APP/PS1 transgenic mice. Behav Brain Res 2012; 235:1-6. [PMID: 22828014 DOI: 10.1016/j.bbr.2012.07.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 07/08/2012] [Accepted: 07/10/2012] [Indexed: 12/23/2022]
Abstract
Proton magnetic resonance spectroscopy ((1)H-MRS) and the Morris water maze (MWM) have played an important role in Alzheimer's disease (AD) research. The aim of this study was to determine whether (1)H-MRS and the MWM can detect for early AD in APP/PS1 transgenic (tg) mice. (1)H-MRS was performed in 20 tg mice and 15 wild-type mice at 3, 5 and 8 months of age. The concentration of N-acetylaspartate (NAA), glutamate (Glu), myo-inositol (mI), choline (Cho) and creatine (Cr) in the hippocampus were measured, and the NAA/Cr, Glu/Cr, mI/Cr and Cho/Cr ratios were quantified. Additionally, the spatial learning and memory of the mice were evaluated by MWM. The (1)H-MRS revealed that mI levels in tg mice were significantly higher at 3 months of age compared to wt mice, while the NAA and Glu levels in 5- and 8-month-old tg mice were significantly decreased (p<0.05). Additionally, significant cognitive changes only occurred at 8 months of age in APP/PS1 tg mice. These results indicated that metabolic changes preceded overt cognitive dysfunctions in early-stage AD, suggesting that (1)H-MRS is a more sensitive biomarker for assessing early AD.
Collapse
Affiliation(s)
- Shuang-qing Chen
- Department of Radiology, Suzhou Hospital, Nanjing Medical University, Suzhou 215001, China.
| | | | | | | | | | | | | |
Collapse
|
63
|
Filali M, Lalonde R, Theriault P, Julien C, Calon F, Planel E. Cognitive and non-cognitive behaviors in the triple transgenic mouse model of Alzheimer's disease expressing mutated APP, PS1, and Mapt (3xTg-AD). Behav Brain Res 2012; 234:334-42. [PMID: 22796601 DOI: 10.1016/j.bbr.2012.07.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 06/19/2012] [Accepted: 07/06/2012] [Indexed: 01/01/2023]
Abstract
3xTg-AD mutant mice are characterized by parenchymal Aβ plaques and neurofibrillary tangles resembling those found in patients with Alzheimer's disease. The mutants were compared with non-transgenic controls in sensorimotor and learning tests. 3xTg-AD mutants were deficient in T-maze reversal, object recognition, and passive avoidance learning. In addition, the mutants showed hypoactivity in two open-field tests, fewer fecal boli in an observation jar, and reduced enclosed arm entries and head-dipping in the elevated plus-maze. On the contrary, the mutants did not differ from controls in pain thresholds, nest-building, and various reflexes determined by the SHIRPA primary screen and were even better on the rotorod test of motor coordination.
Collapse
Affiliation(s)
- Mohammed Filali
- Neurobehavioral Phenotyping Platform, Department of Molecular Medicine, Laval University and Laboratory of Endocrinology and Genomics, CHUQ Research Center, 2705 Blvd Laurier, Quebec, QC G1V 4G2, Canada.
| | | | | | | | | | | |
Collapse
|
64
|
Lalonde R, Fukuchi K, Strazielle C. APP transgenic mice for modelling behavioural and psychological symptoms of dementia (BPSD). Neurosci Biobehav Rev 2012; 36:1357-75. [PMID: 22373961 PMCID: PMC3340431 DOI: 10.1016/j.neubiorev.2012.02.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 02/10/2012] [Accepted: 02/13/2012] [Indexed: 12/17/2022]
Abstract
The discovery of gene mutations responsible for autosomal dominant Alzheimer's disease has enabled researchers to reproduce in transgenic mice several hallmarks of this disorder, notably Aβ accumulation, though in most cases without neurofibrillary tangles. Mice expressing mutated and wild-type APP as well as C-terminal fragments of APP exhibit variations in exploratory activity reminiscent of behavioural and psychological symptoms of Alzheimer dementia (BPSD). In particular, open-field, spontaneous alternation, and elevated plus-maze tasks as well as aggression are modified in several APP transgenic mice relative to non-transgenic controls. However, depending on the precise murine models, changes in open-field and elevated plus-maze exploration occur in either direction, either increased or decreased relative to controls. It remains to be determined which neurotransmitter changes are responsible for this variability, in particular with respect to GABA, 5HT, and dopamine.
Collapse
Affiliation(s)
- R Lalonde
- Département de Psychologie, Faculté des Sciences, Université de Rouen, 76821 Mont-Saint-Aignan Cedex, France.
| | | | | |
Collapse
|
65
|
Ferrington L, Palmer LE, Love S, Horsburgh KJ, Kelly PAT, Kehoe PG. Angiotensin II-inhibition: effect on Alzheimer's pathology in the aged triple transgenic mouse. Am J Transl Res 2012; 4:151-164. [PMID: 22611468 PMCID: PMC3353528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 04/06/2012] [Indexed: 06/01/2023]
Abstract
Reducing excessive accumulation of amyloid-β (Aβ) in Alzheimer's disease (AD) is a key objective of most AD therapies, and inhibition of angiotensin-converting enzyme (ACE) may delay onset or progression of AD. The effects of an ACE-inhibitor (ACE-I) and an angiotensin II receptor blocker (ARB) on Aβ and tau pathology in a triple transgenic (3xTGAD) mouse model of AD were investigated. 9-10month 3xTGAD mice were treated with ARB, ACE-I or vehicle for 6 months. Mean arterial blood pressure (MABP) was measured periodically and mice were assessed behaviourally. Aβ, phospho-tau, amyloid precursor protein (APP) and ACE activity were analysed. MABP was significantly reduced at 2 weeks and 3 months in the ACE-I group and at 3 months in the ARB group, compared to vehicle. Neither drug altered performance of 3xTGAD mice in Morris Water Maze or T-maze, nor were Aβ, tau immunolabelling or APP levels altered. ACE-I significantly reduced ACE activity in kidney. Prolonged treatment with ACE-I or ARB does not affect Aβ or phospho-tau accumulation in brains of aged 3xTGAD mice.
Collapse
Affiliation(s)
- Linda Ferrington
- University of Edinburgh, MRC Centre for Inflammation Research, The Queens Medical Research Institute47 Little France Crescent, Edinburgh, EH16 4TJ
| | - Laura E. Palmer
- Dementia Research Group, John James Laboratories, School of Clinical Sciences, University of Bristol, Frenchay HospitalBristol, BS16 1LE, United Kingdom
| | - Seth Love
- Department of Neuropathology, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Frenchay HospitalBristol, BS16 1LE, United Kingdom
| | - Karen J Horsburgh
- Centre for Cognitive Aging and Cognitive Epidemiology, Centre for Neuroregeneration, Chancellor’s building, 49 Little France Crescent, University of EdinburghEdinburgh, EH16 4SB, United Kingdom
| | - Paul AT Kelly
- University of Edinburgh, MRC Centre for Inflammation Research, The Queens Medical Research Institute47 Little France Crescent, Edinburgh, EH16 4TJ
| | - Patrick G Kehoe
- Dementia Research Group, John James Laboratories, School of Clinical Sciences, University of Bristol, Frenchay HospitalBristol, BS16 1LE, United Kingdom
| |
Collapse
|
66
|
Foster TC. Dissecting the age-related decline on spatial learning and memory tasks in rodent models: N-methyl-D-aspartate receptors and voltage-dependent Ca2+ channels in senescent synaptic plasticity. Prog Neurobiol 2012; 96:283-303. [PMID: 22307057 DOI: 10.1016/j.pneurobio.2012.01.007] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 01/09/2012] [Accepted: 01/19/2012] [Indexed: 10/14/2022]
Abstract
In humans, heterogeneity in the decline of hippocampal-dependent episodic memory is observed during aging. Rodents have been employed as models of age-related cognitive decline and the spatial water maze has been used to show variability in the emergence and extent of impaired hippocampal-dependent memory. Impairment in the consolidation of intermediate-term memory for rapidly acquired and flexible spatial information emerges early, in middle-age. As aging proceeds, deficits may broaden to include impaired incremental learning of a spatial reference memory. The extent and time course of impairment has been be linked to senescence of calcium (Ca²⁺) regulation and Ca²⁺-dependent synaptic plasticity mechanisms in region CA1. Specifically, aging is associated with altered function of N-methyl-D-aspartate receptors (NMDARs), voltage-dependent Ca²⁺ channels (VDCCs), and ryanodine receptors (RyRs) linked to intracellular Ca²⁺ stores (ICS). In young animals, NMDAR activation induces long-term potentiation of synaptic transmission (NMDAR-LTP), which is thought to mediate the rapid consolidation of intermediate-term memory. Oxidative stress, starting in middle-age, reduces NMDAR function. In addition, VDCCs and ICS can actively inhibit NMDAR-dependent LTP and oxidative stress enhances the role of VDCC and RyR-ICS in regulating synaptic plasticity. Blockade of L-type VDCCs promotes NMDAR-LTP and memory in older animals. Interestingly, pharmacological or genetic manipulations to reduce hippocampal NMDAR function readily impair memory consolidation or rapid learning, generally leaving incremental learning intact. Finally, evidence is mounting to indicate a role for VDCC-dependent synaptic plasticity in associative learning and the consolidation of remote memories. Thus, VDCC-dependent synaptic plasticity and extrahippocampal systems may contribute to incremental learning deficits observed with advanced aging.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, Evelyn F. and William L. McKnight Brain Institute, University of Florida, PO Box 100244, Gainesville, FL 32610-0244, USA. ,
| |
Collapse
|
67
|
Sun L, Lam WP, Wong YW, Lam LH, Tang HC, Wai MS, Mak YT, Pan F, Yew DT. Permanent deficits in brain functions caused by long-term ketamine treatment in mice. Hum Exp Toxicol 2010; 30:1287-96. [PMID: 21056951 DOI: 10.1177/0960327110388958] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ketamine, an injectable anesthetic, is also a popular recreational drug used by young adults worldwide. Ketamine is a non-competitive antagonist of N-methyl-d-aspartate receptor, which plays important roles in synaptic plasticity and neuronal learning. Most previous studies have examined the immediate and short-term effects of ketamine, which include learning and cognitive deficits plus impairment of working memory, whereas little is known about the long-term effects of repeated ketamine injections of common or usual recreational doses. Therefore, we aimed to evaluate the deficits in brain functions with behavioral tests, including wire hang, hot plate and water maze tests, plus examine prefrontal cortex apoptotic markers, including Bax, Bcl-2 and caspase-3, in mice treated with 6 months of daily ketamine administration. In our study, following 6 months of ketamine injection, mice showed significant deterioration in neuromuscular strength and nociception 4 hours post-dose, but learning and working memory were not affected nor was there significant apoptosis in the prefrontal cortex. Our research revealed the important clinical finding that long-term ketamine abuse with usual recreational doses can detrimentally affect neuromuscular strength and nociception as part of measurable, stable and persistent deficits in brain function.
Collapse
Affiliation(s)
- L Sun
- Department of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
A novel, rapidly acquired and persistent spatial memory task that induces immediate early gene expression. Behav Brain Funct 2010; 6:35. [PMID: 20594357 PMCID: PMC2911393 DOI: 10.1186/1744-9081-6-35] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 07/02/2010] [Indexed: 02/02/2023] Open
Abstract
Background The Morris water maze task is a hippocampus-dependent learning and memory test that typically takes between 3 days to 2 weeks of training. This task is used to assess spatial learning and induces the expression of genes known to be crucial to learning and memory in the hippocampus. A major caveat in the protocol is the prolonged duration of training, and difficulty of assessing the time during training in which animals have learned the task. We introduce here a condensed version of the task that like traditional water maze tasks, creates lasting hippocampus-dependent spatial cognitive maps and elicits gene expression following learning. Methods This paradigm was designed for rats to quickly acquire a hippocampus-dependent spatial cognitive map and retain this memory for at least 24 hours. To accomplish this, we interspersed visible and hidden training trials, delivering them in a massed fashion so training takes a maximum of 15 minutes. Learning was assessed based on latencies to the platform during each training trial, as well as time spent in the goal quadrant during probe testing 30 minutes and 24 hours after training. Normal rats were compared to two impaired cohorts (rats with fimbria-fornix lesions and rats administered NMDA receptor antagonist (CPP)). To quantitate hippocampal expression of known learning genes, real-time polymerase chain reaction (RT-PCR) was performed on hippocampal cDNA. Results We show that massed training using alternating visible and hidden training trials generates robust short-term working and long-term reference memories in rats. Like the traditional Morris water maze paradigm, this task requires proper hippocampal function, as rats with fimbria-fornix lesions and rats administered CPP fail to learn the spatial component of the task. Furthermore, training in this paradigm elicits hippocampal expression of genes upregulated following learning in a variety of spatial tasks: homer1a, cfos and zif268. Conclusions We introduce here a condensed version of the Morris water maze, which is like a traditional water maze paradigm, in that it is hippocampus-dependent, and elicits hippocampal expression of learning genes. However, this task is administered in 15 minutes and induces spatial memory for at least 24 hours.
Collapse
|