1
|
Cushing SD, Moseley SC, Stimmell AC, Schatschneider C, Wilber AA. Rescuing impaired hippocampal-cortical interactions and spatial reorientation learning and memory during sleep in a mouse model of Alzheimer's disease using hippocampal 40 Hz stimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599921. [PMID: 38979221 PMCID: PMC11230253 DOI: 10.1101/2024.06.20.599921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
In preclinical Alzheimer's disease (AD), spatial learning and memory is impaired. We reported similar impairments in 3xTg-AD mice on a virtual maze (VM) spatial-reorientation-task that requires using landmarks to navigate. Hippocampal (HPC)-cortical dysfunction during sleep (important for memory consolidation) is a potential mechanism for memory impairments in AD. We previously found deficits in HPC-cortical coordination during sleep coinciding with VM impairments the next day. Some forms of 40 Hz stimulation seem to clear AD pathology in mice, and improve functional connectivity in AD patients. Thus, we implanted a recording array targeting parietal cortex (PC) and HPC to assess HPC-PC coordination, and an optical fiber targeting HPC for 40 Hz or sham optogenetic stimulation in 3xTg/PV cre mice. We assessed PC delta waves (DW) and HPC sharp wave ripples (SWRs). In sham mice, SWR-DW cross-correlations were reduced, similar to 3xTg-AD mice. In 40 Hz mice, this phase-locking was rescued, as was performance on the VM. However, rescued HPC-PC coupling no longer predicted performance as in NonTg animals. Instead, DWs and SWRs independently predicted performance in 40 Hz mice. Thus, 40 Hz stimulation of HPC rescued functional interactions in the HPC-PC network, and rescued impairments in spatial navigation, but did not rescue the correlation between HPC-PC coordination during sleep and learning and memory. Together this pattern of results could inform AD treatment timing by suggesting that despite applying 40 Hz stimulation before significant tau and amyloid aggregation, pathophysiological processes led to brain changes that were not fully reversed even though cognition was recovered. Significance Statement One of the earliest symptoms of Alzheimer's disease (AD) is getting lost in space or experiencing deficits in spatial navigation, which involve navigation computations as well as learning and memory. We investigated cross brain region interactions supporting memory formation as a potential causative factor of impaired spatial learning and memory in AD. To assess this relationship between AD pathophysiology, brain changes, and behavioral alterations, we used a targeted approach for clearing amyloid beta and tau to rescue functional interactions in the brain. This research strongly connects brain activity patterns during sleep to tau and amyloid accumulation, and will aid in understanding the mechanisms underlying cognitive dysfunction in AD. Furthermore, the results offer insight for improving early identification and treatment strategies.
Collapse
|
2
|
Duggan MR, Steinberg Z, Peterson T, Francois TJ, Parikh V. Cognitive trajectories in longitudinally trained 3xTg-AD mice. Physiol Behav 2024; 275:114435. [PMID: 38103626 PMCID: PMC10872326 DOI: 10.1016/j.physbeh.2023.114435] [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: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
Preclinical studies in Alzheimer's disease (AD) often rely on cognitively naïve animal models in cross-sectional designs that can fail to reflect the cognitive exposures across the lifespan and heterogeneous neurobehavioral features observed in humans. To determine whether longitudinal cognitive training may affect cognitive capacities in a well-characterized AD mouse model, 3xTg and wild-type mice (n = 20) were exposed daily to a training variant of the Go-No-Go (GNG) operant task from 3 to 9 months old. At 3, 6, and 9 months, performance on a testing variant of the GNG task and anxiety-like behaviors were measured, while long-term recognition memory was also assessed at 9 months. In general, GNG training improved performance with increasing age across genotypes. At 3 months old, 3xTg mice showed slight deficits in inhibitory control that were accompanied by minor improvements in signal detection and decreased anxiety-like behavior, but these differences did not persist at 6 and 9 months old. At 9 months old, 3xTg mice displayed minor deficits in signal detection, and long-term recognition memory capacity was comparable with wild-type subjects. Our findings indicate that longitudinal cognitive training can render 3xTg mice with cognitive capacities that are on par with their wild-type counterparts, potentially reflecting functional compensation in subjects harboring AD genetic mutations.
Collapse
Affiliation(s)
- Michael R Duggan
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Zoe Steinberg
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Tara Peterson
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Tara-Jade Francois
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States
| | - Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, United States.
| |
Collapse
|
3
|
Reyna NC, Clark BJ, Hamilton DA, Pentkowski NS. Anxiety and Alzheimer's disease pathogenesis: focus on 5-HT and CRF systems in 3xTg-AD and TgF344-AD animal models. Front Aging Neurosci 2023; 15:1251075. [PMID: 38076543 PMCID: PMC10699143 DOI: 10.3389/fnagi.2023.1251075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/25/2023] [Indexed: 02/12/2024] Open
Abstract
Dementia remains one of the leading causes of morbidity and mortality in older adults. Alzheimer's disease (AD) is the most common type of dementia, affecting over 55 million people worldwide. AD is characterized by distinct neurobiological changes, including amyloid-beta protein deposits and tau neurofibrillary tangles, which cause cognitive decline and subsequent behavioral changes, such as distress, insomnia, depression, and anxiety. Recent literature suggests a strong connection between stress systems and AD progression. This presents a promising direction for future AD research. In this review, two systems involved in regulating stress and AD pathogenesis will be highlighted: serotonin (5-HT) and corticotropin releasing factor (CRF). Throughout the review, we summarize critical findings in the field while discussing common limitations with two animal models (3xTg-AD and TgF344-AD), novel pharmacotherapies, and potential early-intervention treatment options. We conclude by highlighting promising future pharmacotherapies and translational animal models of AD and anxiety.
Collapse
Affiliation(s)
- Nicole C. Reyna
- Department of Psychology, University of New Mexico, Albuquerque, NM, United States
| | | | | | | |
Collapse
|
4
|
Torraville SE, Flynn CM, Kendall TL, Yuan Q. Life Experience Matters: Enrichment and Stress Can Influence the Likelihood of Developing Alzheimer's Disease via Gut Microbiome. Biomedicines 2023; 11:1884. [PMID: 37509523 PMCID: PMC10377385 DOI: 10.3390/biomedicines11071884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease, characterized by the presence of β-amyloid (Aβ) plaques and neurofibrillary tangles (NFTs) formed from abnormally phosphorylated tau proteins (ptau). To date, there is no cure for AD. Earlier therapeutic efforts have focused on the clinical stages of AD. Despite paramount efforts and costs, pharmaceutical interventions including antibody therapies targeting Aβ have largely failed. This highlights the need to alternate treatment strategies and a shift of focus to early pre-clinical stages. Approximately 25-40% of AD cases can be attributed to environmental factors including chronic stress. Gut dysbiosis has been associated with stress and the pathogenesis of AD and can increase both Aβ and NFTs in animal models of the disease. Both stress and enrichment have been shown to alter AD progression and gut health. Targeting stress-induced gut dysbiosis through probiotic supplementation could provide a promising intervention to delay disease progression. In this review, we discuss the effects of stress, enrichment, and gut dysbiosis in AD models and the promising evidence from probiotic intervention studies.
Collapse
Affiliation(s)
- Sarah E Torraville
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Cassandra M Flynn
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Tori L Kendall
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Qi Yuan
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| |
Collapse
|
5
|
Baeta-Corral R, De la Fuente M, Giménez-Llort L. Sex-dependent worsening of NMDA-induced responses, anxiety, hypercortisolemia, and organometry of early peripheral immunoendocrine impairment in adult 3xTg-AD mice and their long-lasting ontogenic modulation by neonatal handling. Behav Brain Res 2023; 438:114189. [PMID: 36343697 DOI: 10.1016/j.bbr.2022.114189] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 10/23/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
The neuroimmunomodulation hypothesis for Alzheimer's disease (AD) postulates that alterations in the innate immune system triggered by damage signals result in adverse effects on neuronal functions. The peripheral immune system and neuroimmunoendocrine communication are also impaired. Here we provide further evidence using a longitudinal design that also studied the long-lasting effects of an early life sensorial intervention (neonatal handling, from postnatal day 1-21) in 6-month-old (early stages of the disease) male and female 3xTg-AD mice compared to age- and sex-matched non-transgenic (NTg) mice with normal aging. The behavioral patterns elicited by the direct exposure to an open field, and the motor depression response evoked by NMDA (25 mg/kg, i.p) were found correlated to the organometry of peripheral immune-endocrine organs (thymus involution, splenomegaly, and adrenal glands' hypertrophy) and increased corticosterone levels, suggesting their potential value for diagnostic and biomonitoring.The NMDA-induced immediate and depressant motor activity and endocrine (corticosterone) responses were sensitive to sex and AD-genotype, suggesting worse endogenous susceptibility/neuroprotective response to glutamatergic excitotoxicity in males and in the AD-genotype. 3xTg-AD females showed a reduced immediate response, whereas the NTg showed higher responsiveness to subsequent NMDA-induced depressant effect than their male counterparts. The long-lasting ontogenic modulation by handling was shown as a potentiation of NMDA-depressant effect in NTg males and females, while sex × treatment effects were found in 3xTg-AD mice. Finally, NMDA-induced corticosterone showed sex, genotype and interaction effects with sexual dimorphism enhanced in the AD-genotype, suggesting different endogenous vulnerability/neuroprotective capacities and modulation of the neuroimmunoendocrine system.
Collapse
Affiliation(s)
- R Baeta-Corral
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - M De la Fuente
- Department of Genetics, Physiology, and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain
| | - L Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain.
| |
Collapse
|
6
|
Sex- and Neuropsychiatric-Dependent Circadian Alterations in Daily Voluntary Physical Activity Engagement and Patterns in Aged 3xTg-AD Mice. Int J Mol Sci 2022; 23:ijms232213671. [PMID: 36430150 PMCID: PMC9696337 DOI: 10.3390/ijms232213671] [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: 10/02/2022] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Alzheimer's disease (AD) patients suffer from circadian rhythm alterations affecting their daily physical activity patterns with less willingness to perform a voluntary exercise. In preclinical studies, there is no clarity on whether animal models of AD can replicate these impairments. Here, we provide a proof of concept of the performance and behavioral effects of four weeks of voluntary wheel running (VWR) in a group of 14-month-old male and female 3xTg-AD mice at advanced stages of AD and the daily variance (behavioral circadian rhythmicity) of VWR associated with sex and their neuropsychiatric-like phenotype. Higher levels of horizontal exploration in the open field (OF) test were found in mice submitted to exercise. A linear mixed effect model showed significant sex-dependent differences in the VWR activity performed on the first night of follow-up, with high-NIBI males running less than high-NIBI females. Thus, an influence of NPS-like symptoms on the circadian patterns of VWR may account for such differences. In addition, males remained more active than females during diurnal periods. We hypothesize that this increment in energy expenditure during resting periods may be related to hyperactive behavior, similar to that observed in humans' exacerbated agitation or sundowning behavior. These findings support the usage of the 3xTg-AD mouse as a reliable model for studying circadian rhythm alterations in AD and, at the translational level, the importance of tailored and individualized physical activity programs in clinical settings.
Collapse
|
7
|
Effects of early life adversities upon memory processes and cognition in rodent models. Neuroscience 2022; 497:282-307. [PMID: 35525496 DOI: 10.1016/j.neuroscience.2022.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 01/14/2023]
Abstract
Exposure to stressors in early postnatal life induces long-lasting modifications in brainfunction.Thisplasticity,an essential characteristic of the brain that enables adaptation to the environment, may also induce impairments in some psychophysiological functions, including learning and memory. Early life stress (ELS) has long-term effects on thehypothalamic-pituitary-adrenal axisresponse to stressors, and has been reported to lead toneuroinflammation,altered levelsof neurotrophic factors, modifications inneurogenesis andsynaptic plasticity,with changes in neurotransmitter systems and network functioning. In this review, we focus on early postnatal stress in animal models and their effects on learning and memory.Many studies have reported ELS-induced impairments in different types of memories, including spatial memory, fear memory, recognition (both for objects and social) memory, working memory and reversal learning. Studies are not always in agreement, however, no effects, or sometimes facilitation, being reported, depending on the nature and intensity of the early intervention, as well as the age when the outcome was evaluated and the sex of the animals. When considering processes occurring after consolidation, related with memory maintenance or modification, there are a very reduced number of reports. Future studies addressing the mechanisms underlying memory changes for ELS should shed some light on the understanding of the different effects induced by stressors of different types and intensities on cognitive functions.
Collapse
|
8
|
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: 8.5] [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
|
9
|
Xu B, He Y, Liu L, Ye G, Chen L, Wang Q, Chen M, Chen Y, Long D. The Effects of Physical Running on Dendritic Spines and Amyloid-beta Pathology in 3xTg-AD Male Mice. Aging Dis 2022; 13:1293-1310. [PMID: 35855335 PMCID: PMC9286906 DOI: 10.14336/ad.2022.0110] [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: 10/20/2021] [Accepted: 01/10/2022] [Indexed: 11/01/2022] Open
Abstract
Memory loss is the key symptom of Alzheimer's disease (AD). As successful drug treatments have not yet been identified, non-pharmaceutical interventions such as physical exercise and training have been employed to improve the memory function of people with dementia. We investigated the effect of prolonged physical running on hippocampal-dependent spatial memory and its underlying mechanisms using a well-established rodent model of AD. 3xTg-AD transgenic mice and non-transgenic mice were subjected to voluntary wheel running for 5 months (1 hour per day, 5 days per week), followed by spatial memory testing. After the behavioral testing, dendritic spines, synapses, and synaptic proteins as well as amyloid-beta (Aβ) pathology were analyzed in the dorsal hippocampi. Running improved hippocampal-dependent spatial memory in 3xTg-AD mice. This running strategy prevented both thin and mushroom-type spines on CA1 pyramidal cells in 3xTg-AD mice, whereas the effects of running in non-transgenic mice were limited to thin spines. The enormous effects of running on spines were accompanied by an increased number of synapses and upregulated expression of synaptic proteins. Notably, running downregulated the processing of amyloid precursor protein, decreasing intracellular APP expression and extracellular Aβ accumulation, and spatial memory performance correlated with levels of Aβ peptides Aβ1-40 and Aβ1-42. These data suggest that prolonged running may improve memory in preclinical AD via slowing down the amyloid pathology and preventing the loss of synaptic contacts.
Collapse
Affiliation(s)
- Benke Xu
- Department of Human Anatomy, School of Basic Medical Sciences, Yangtze University, Hubei 434023, China.
| | - Yun He
- Department of Human Anatomy, School of Basic Medical Sciences, Yangtze University, Hubei 434023, China.
| | - Lian Liu
- Department of Pharmacology, School of Basic Medical Sciences, Yangtze University, Hubei 434023, China.
| | - Guosheng Ye
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Lulu Chen
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Qingning Wang
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Michael Chen
- University of California, Los Angeles, CA 90095, USA.
| | - Yuncai Chen
- Department of Pediatrics, University of California, Irvine, CA 92697, USA.
- Correspondence should be addressed to: Dr. Dahong Long, Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China. E-mail: or Dr. Yuncai Chen, Department of Pediatrics, University of California-Irvine, Irvine, California 92697, USA. E-mail:
| | - Dahong Long
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
- Correspondence should be addressed to: Dr. Dahong Long, Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China. E-mail: or Dr. Yuncai Chen, Department of Pediatrics, University of California-Irvine, Irvine, California 92697, USA. E-mail:
| |
Collapse
|
10
|
Gloria Y, Ceyzériat K, Tsartsalis S, Millet P, Tournier BB. Dopaminergic dysfunction in the 3xTg-AD mice model of Alzheimer's disease. Sci Rep 2021; 11:19412. [PMID: 34593951 PMCID: PMC8484608 DOI: 10.1038/s41598-021-99025-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/16/2021] [Indexed: 11/09/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid (Aβ) protein aggregation and neurofibrillary tangles accumulation, accompanied by neuroinflammation. With all the therapeutic attempts targeting these biomarkers having been unsuccessful, the understanding of early mechanisms involved in the pathology is of paramount importance. Dopaminergic system involvement in AD has been suggested, particularly through the appearance of dopaminergic dysfunction-related neuropsychiatric symptoms and an overall worsening of cognitive and behavioral symptoms. In this study, we reported an early dopaminergic dysfunction in a mouse model presenting both amyloid and Tau pathology. 3xTg-AD mice showed an increase of postsynaptic D2/3R receptors density in the striatum and D2/3-autoreceptors in SN/VTA cell bodies. Functionally, a reduction of anxiety-like behavior, an increase in locomotor activity and D2R hyper-sensitivity to quinpirole stimulation have been observed. In addition, microglial cells in the striatum showed an early inflammatory response, suggesting its participation in dopaminergic alterations. These events are observed at an age when tau accumulation and Aβ deposits in the hippocampus are low. Thus, our results suggest that early dopaminergic dysfunction could have consequences in behavior and cognitive function, and may shed light on future therapeutic pathways of AD.
Collapse
Affiliation(s)
- Yesica Gloria
- Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Kelly Ceyzériat
- Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Division of Nuclear Medicine, Diagnostic Department, University Hospitals of Geneva, Geneva, Switzerland.,Division of Radiation Oncology, Department of Oncology, University Hospitals of Geneva, Geneva, Switzerland
| | - Stergios Tsartsalis
- Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland.,Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Philippe Millet
- Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Benjamin B Tournier
- Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland. .,Department of Psychiatry, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
11
|
Sex-Dependent Signatures, Time Frames and Longitudinal Fine-Tuning of the Marble Burying Test in Normal and AD-Pathological Aging Mice. Biomedicines 2021; 9:biomedicines9080994. [PMID: 34440198 PMCID: PMC8391620 DOI: 10.3390/biomedicines9080994] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 12/19/2022] Open
Abstract
The marble burying (MB) test, a classical test based on the natural tendency of rodents to dig in diverse substrates and to bury small objects, is sensitive to some intrinsic and extrinsic factors. Here, under emerging neuroethological quantitative and qualitative analysis, the MB performance of 12-month-old male and female 3xTg-AD mice for Alzheimer’s disease and age-matched counterparts of gold-standard C57BL6 strain with normal aging unveiled sex-dependent signatures. In addition, three temporal analyses, through the (1) time course of the performance, and (2) a repeated test schedule, identified the optimal time frames and schedules to detect sex- and genotype-dependent differences. Besides, a (3) longitudinal design from 12 to 16 months of age monitored the changes in the performance with aging, worsening in AD-mice, and modulation through the repeated test. In summary, the present results allow us to conclude that (1) the marble burying test is responsive to genotype, sex, aging, and its interactions; (2) the male sex was more sensitive to showing the AD-phenotype; (3) longitudinal assessment shows a reduction in females with AD pathology; (4) burying remains stable in repeated testing; (5) the time-course of marbles burying is useful; and (6) burying behavior most likely represents perseverative and/or stereotyped-like behavior rather than anxiety-like behavior in 3xTg-AD mice.
Collapse
|
12
|
Social Nesting, Animal Welfare, and Disease Monitoring. Animals (Basel) 2021; 11:ani11041079. [PMID: 33918975 PMCID: PMC8070261 DOI: 10.3390/ani11041079] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Most standardized tools to evaluate welfare and disease progression in animals assess the individuals, while social behaviors are scarcely monitored, despite being useful to detecting acute illness and chronic and mental health problems. The main reason is that social behavior is complex and time-consuming. We are currently using the nests built by animals living together, a species-typical behavior naturally occurring in standard housing conditions, to monitor them. Here, we provide an example of its use to evaluate social deficits and the long-term effects of a neonatal tactile-proprioceptive sensorial treatment from postnatal day 1 to 21, in male and female adult mice modeling Alzheimer’s disease compared to mice with normal aging. Social nesting was worse in the mutants, mostly in males, since the number of days needed to build a perfect nest was longer or unsuccessful in a three-day test. Early life intervention was successful. Social nesting, easily included in housing routines, can be a useful tool to assess animal welfare, monitor disease progress, and evaluate potential risk factors and effects of preventive/therapeutical strategies. Other advantages, such as being a noninvasive, painless, simple, short, and low-cost, rend social nesting feasible to be implemented in most animal department settings. Abstract The assessment of welfare and disease progression in animal models is critical. Most tools rely on evaluating individual subjects, whereas social behaviors, also sensitive to acute illness, chronic diseases, or mental health, are scarcely monitored because they are complex and time-consuming. We propose the evaluation of social nesting, a species-typical behavior naturally occurring in standard housing conditions, for such behavioral monitoring. We provide an example of its use to evaluate social deficits and the long-term effects of neonatal tactile-proprioceptive sensorial stimulation from postnatal day 1 to 21, in male and female adult 3xTg-AD mice for Alzheimer’s disease compared to sex- and age-matched non-transgenic (NTg) counterparts with normal aging. Social nesting was sensitive to genotype (worse in 3xTg-AD mice), sex (worse in males), profile, and treatment (distinct time to observe the maximum score and incidence of the perfect nest). Since social nesting can be easily included in housing routines, this neuroethological approach can be useful for animal welfare, monitoring the disease’s progress, and evaluating potential risk factors and effects of preventive/therapeutical strategies. Finally, the noninvasive, painless, simple, short time, and low-cost features of this home-cage monitoring are advantages that make social nesting feasible to be successfully implemented in most animal department settings.
Collapse
|
13
|
Clock/Sleep-Dependent Learning and Memory in Male 3xTg-AD Mice at Advanced Disease Stages and Extrinsic Effects of Huprine X and the Novel Multitarget Agent AVCRI104P3. Brain Sci 2021; 11:brainsci11040426. [PMID: 33810622 PMCID: PMC8065516 DOI: 10.3390/brainsci11040426] [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: 01/29/2021] [Revised: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 01/20/2023] Open
Abstract
A new hypothesis highlights sleep-dependent learning/memory consolidation and regards the sleep-wake cycle as a modulator of β-amyloid and tau Alzheimer's disease (AD) pathologies. Sundowning behavior is a common neuropsychiatric symptom (NPS) associated with dementia. Sleep fragmentation resulting from disturbances in sleep and circadian rhythms in AD may have important consequences on memory processes and exacerbate the other AD-NPS. The present work studied the effect of training time schedules on 12-month-old male 3xTg-AD mice modeling advanced disease stages. Their performance in two paradigms of the Morris water maze for spatial-reference and visual-perceptual learning and memory were found impaired at midday, after 4 h of non-active phase. In contrast, early-morning trained littermates, slowing down from their active phase, exhibited better performance and used goal-directed strategies and non-search navigation described for normal aging. The novel multitarget anticholinesterasic compound AVCRI104P3 (0.6 µmol·kg-1, 21 days i.p.) exerted stronger cognitive benefits than its in vitro equipotent dose of AChEI huprine X (0.12 μmol·kg-1, 21 days i.p.). Both compounds showed streamlined drug effectiveness, independently of the schedule. Their effects on anxiety-like behaviors were moderate. The results open a question of how time schedules modulate the capacity to respond to task demands and to assess/elucidate new drug effectiveness.
Collapse
|
14
|
Dennison JL, Ricciardi NR, Lohse I, Volmar CH, Wahlestedt C. Sexual Dimorphism in the 3xTg-AD Mouse Model and Its Impact on Pre-Clinical Research. J Alzheimers Dis 2021; 80:41-52. [PMID: 33459720 PMCID: PMC8075398 DOI: 10.3233/jad-201014] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Female sex is a leading risk factor for developing Alzheimer’s disease (AD). Sexual dimorphism in AD is gaining attention as clinical data show that women are not only more likely to develop AD but also to experience worse pathology and faster cognitive decline. Pre-clinical AD research in animal models often neglects to address sexual dimorphism in evaluation of behavioral or molecular characteristics and outcomes. This can compromise its translation to a clinical setting. The triple-transgenic AD mouse model (3xTg-AD) is a commonly used but unique AD model because it exhibits both amyloid and tau pathology, essential features of the human AD phenotype. Mounting evidence has revealed important sexually dimorphic characteristics of this animal model that have yet to be reviewed and thus, are often overlooked in studies using the 3xTg-AD model. In this review we conduct a thorough analysis of reports of sexual dimorphism in the 3xTg-AD model including findings of molecular, behavioral, and longevity-related sex differences in original research articles through August 2020. Importantly, we find results to be inconsistent, and that strain source and differing methodologies are major contributors to lack of consensus regarding traits of each sex. We first touch on the nature of sexual dimorphism in clinical AD, followed by a brief summary of sexual dimorphism in other major AD murine models before discussing the 3xTg-AD model in depth. We conclude by offering four suggestions to help unify pre-clinical mouse model AD research inspired by the NIH expectations for considering sex as a biological variable.
Collapse
Affiliation(s)
- Jessica L Dennison
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Natalie R Ricciardi
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ines Lohse
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Claude-Henry Volmar
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Claes Wahlestedt
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
15
|
Sex-Dependent End-of-Life Mental and Vascular Scenarios for Compensatory Mechanisms in Mice with Normal and AD-Neurodegenerative Aging. Biomedicines 2021; 9:biomedicines9020111. [PMID: 33498895 PMCID: PMC7911097 DOI: 10.3390/biomedicines9020111] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
Life expectancy decreases with aging, with cardiovascular, mental health, and neurodegenerative disorders strongly contributing to the total disability-adjusted life years. Interestingly, the morbidity/mortality paradox points to females having a worse healthy life expectancy. Since bidirectional interactions between cardiovascular and Alzheimer’s diseases (AD) have been reported, the study of this emerging field is promising. In the present work, we further explored the cardiovascular–brain interactions in mice survivors of two cohorts of non-transgenic and 3xTg-AD mice, including both sexes, to investigate the frailty/survival through their life span. Survival, monitored from birth, showed exceptionally worse mortality rates in females than males, independently of the genotype. This mortality selection provided a “survivors” cohort that could unveil brain–cardiovascular interaction mechanisms relevant for normal and neurodegenerative aging processes restricted to long-lived animals. The results show sex-dependent distinct physical (worse in 3xTg-AD males), neuropsychiatric-like and cognitive phenotypes (worse in 3xTg-AD females), and hypothalamic–pituitary–adrenal (HPA) axis activation (higher in females), with higher cerebral blood flow and improved cardiovascular phenotype in 3xTg-AD female mice survivors. The present study provides an experimental scenario to study the suggested potential compensatory hemodynamic mechanisms in end-of-life dementia, which is sex-dependent and can be a target for pharmacological and non-pharmacological interventions.
Collapse
|
16
|
Differential effects of chronic immunosuppression on behavioral, epigenetic, and Alzheimer's disease-associated markers in 3xTg-AD mice. ALZHEIMERS RESEARCH & THERAPY 2021; 13:30. [PMID: 33472690 PMCID: PMC7818784 DOI: 10.1186/s13195-020-00745-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Circulating autoantibodies and sex-dependent discrepancy in prevalence are unexplained phenomena of Alzheimer's disease (AD). Using the 3xTg-AD mouse model, we reported that adult males show early manifestations of systemic autoimmunity, increased emotional reactivity, enhanced expression of the histone variant macroH2A1 in the cerebral cortex, and loss of plaque/tangle pathology. Conversely, adult females display less severe autoimmunity and retain their AD-like phenotype. This study examines the link between immunity and other traits of the current 3xTg-AD model. METHODS Young 3xTg-AD and wild-type mice drank a sucrose-laced 0.4 mg/ml solution of the immunosuppressant cyclophosphamide on weekends for 5 months. After behavioral phenotyping at 2 and 6 months of age, we assessed organ mass, serologic markers of autoimmunity, molecular markers of early AD pathology, and expression of genes associated with neurodegeneration. RESULTS Chronic immunosuppression prevented hematocrit drop and reduced soluble Aβ in 3xTg-AD males while normalizing the expression of histone variant macroH2A1 in 3xTg-AD females. This treatment also reduced hepatosplenomegaly, lowered autoantibody levels, and increased the effector T cell population while decreasing the proportion of regulatory T cells in both sexes. Exposure to cyclophosphamide, however, neither prevented reduced brain mass and BDNF expression nor normalized increased tau and anxiety-related behaviors. CONCLUSION The results suggest that systemic autoimmunity increases soluble Aβ production and affects transcriptional regulation of macroH2A1 in a sex-related manner. Despite the complexity of multisystem interactions, 3xTg-AD mice can be a useful in vivo model for exploring the regulatory role of autoimmunity in the etiology of AD-like neurodegenerative disorders.
Collapse
|
17
|
Ahmed T, Van der Jeugd A, Caillierez R, Buée L, Blum D, D'Hooge R, Balschun D. Chronic Sodium Selenate Treatment Restores Deficits in Cognition and Synaptic Plasticity in a Murine Model of Tauopathy. Front Mol Neurosci 2020; 13:570223. [PMID: 33132838 PMCID: PMC7578417 DOI: 10.3389/fnmol.2020.570223] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/26/2020] [Indexed: 12/18/2022] Open
Abstract
A major goal in diseases is identifying a potential therapeutic agent that is cost-effective and can remedy some, if not all, disease symptoms. In Alzheimer’s disease (AD), aggregation of hyperphosphorylated tau protein is one of the neuropathological hallmarks, and Tau pathology correlates better with cognitive impairments in AD patients than amyloid-β load, supporting a key role of tau-related mechanisms. Selenium is a non-metallic trace element that is incorporated in the brain into selenoproteins. Chronic treatment with sodium selenate, a non-toxic selenium compound, was recently reported to rescue behavioral phenotypes in tau mouse models. Here, we focused on the effects of chronic selenate application on synaptic transmission and synaptic plasticity in THY-Tau22 mice, a transgenic animal model of tauopathies. Three months with a supplement of sodium selenate in the drinking water (12 μg/ml) restored not only impaired neurocognitive functions but also rescued long-term depression (LTD), a major form of synaptic plasticity. Furthermore, selenate reduced the inactive demethylated catalytic subunit of protein phosphatase 2A (PP2A) in THY-Tau22 without affecting total PP2A.Our study provides evidence that chronic dietary selenate rescues functional synaptic deficits of tauopathy and identifies activation of PP2A as the putative mechanism.
Collapse
Affiliation(s)
- Tariq Ahmed
- Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium
| | - Ann Van der Jeugd
- Leuven Brain Institute, Leuven, Belgium.,Laboratory of Biological Psychology, Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium
| | - Raphaëlle Caillierez
- Univ. Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- Univ. Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Rudi D'Hooge
- Leuven Brain Institute, Leuven, Belgium.,Laboratory of Biological Psychology, Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium
| | - Detlef Balschun
- Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
18
|
Muntsant A, Giménez-Llort L. Impact of Social Isolation on the Behavioral, Functional Profiles, and Hippocampal Atrophy Asymmetry in Dementia in Times of Coronavirus Pandemic (COVID-19): A Translational Neuroscience Approach. Front Psychiatry 2020; 11:572583. [PMID: 33329110 PMCID: PMC7732415 DOI: 10.3389/fpsyt.2020.572583] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/28/2020] [Indexed: 01/10/2023] Open
Abstract
The impact of COVID-19 on the elderly is devastating, and nursing homes are struggling to provide the best care to the most fragile. The urgency and severity of the pandemic forces the use of segregation in restricted areas and confinement in individual rooms as desperate strategies to avoid the spread of disease and the worst-case scenario of becoming a deadly trap. The conceptualization of the post-COVID-19 era implies strong efforts to redesign all living conditions, care/rehabilitation interventions, and management of loneliness forced by social distance measures. Recently, a study of gender differences in COVID-19 found that men are more likely to suffer more severe effects of the disease and are over twice as likely to die. It is well-known that dementia is associated with increased mortality, and males have worse survival and deranged neuro-immuno-endocrine systems than females. The present study examines the impact of long-term isolation in male 3xTg-AD mice modeling advanced stages of Alzheimer's disease (AD) and as compared to age-matched counterparts with normal aging. We used a battery of ethological and unconditioned tests resembling several areas in nursing homes. The main findings refer to an exacerbated (two-fold increase) hyperactivity and emergence of bizarre behaviors in isolated 3xTg-AD mice, worrisome results since agitation is a challenge in the clinical management of dementia and an important cause of caregiver burden. This increase was consistently shown in gross (activity in most of the tests) and fine (thermoregulatory nesting) motor functions. Isolated animals also exhibited re-structured anxiety-like patterns and coping-with-stress strategies. Bodyweight and kidney weight loss were found in AD-phenotypes and increased by isolation. Spleen weight loss was isolation dependent. Hippocampal tau pathology was not modified, but asymmetric atrophy of the hippocampus, recently described in human patients with dementia and modeled here for the first time in an animal model of AD, was found to increase with isolation. Overall, the results show awareness of the impact of isolation in elderly patients with dementia, offering some guidance from translational neuroscience in these times of coronavirus and post-COVID-19 pandemic. They also highlight the relevance of personalized-based interventions tailored to the heterogeneous and complex clinical profile of the individuals with dementia and to consider the implications on caregiver burden.
Collapse
Affiliation(s)
- Aida Muntsant
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lydia Giménez-Llort
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Joseph DJ, Liu C, Peng J, Liang G, Wei H. Isoflurane mediated neuropathological and cognitive impairments in the triple transgenic Alzheimer's mouse model are associated with hippocampal synaptic deficits in an age-dependent manner. PLoS One 2019; 14:e0223509. [PMID: 31600350 PMCID: PMC6786564 DOI: 10.1371/journal.pone.0223509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/22/2019] [Indexed: 12/17/2022] Open
Abstract
Many in vivo studies suggest that inhalational anesthetics can accelerate or prevent the progression of neuropathology and cognitive impairments in Alzheimer Disease (AD), but the synaptic mechanisms mediating these ambiguous effects are unclear. Here, we show that repeated exposures of neonatal and old triple transgenic AD (3xTg) and non-transgenic (NonTg) mice to isoflurane (Iso) distinctly increased neurodegeneration as measured by S100β levels, intracellular Aβ, Tau oligomerization, and apoptotic markers. Spatial cognition measured by reference and working memory testing in the Morris Water Maze (MWM) were altered in young NonTg and 3xTg. Field recordings in the cornu ammonis 1 (CA1) hippocampus showed that neonatal control 3xTg mice exhibited hypo-excitable synaptic transmission, reduced paired-pulse facilitation (PPF), and normal long-term potentiation (LTP) compared to NonTg controls. By contrast, the old control 3xTg mice exhibited hyper-excitable synaptic transmission, enhanced PPF, and unstable LTP compared to NonTg controls. Repeated Iso exposures reduced synaptic transmission and PPF in neonatal NonTg and old 3xTg mice. LTP was normalized in old 3xTg mice, but reduced in neonates. By contrast, LTP was reduced in old but not neonatal NonTg mice. Our results indicate that Iso-mediated neuropathologic and cognitive defects in AD mice are associated with synaptic pathologies in an age-dependent manner. Based on these findings, the extent of this association with age and, possibly, treatment paradigms warrant further study.
Collapse
Affiliation(s)
- Donald J. Joseph
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Chunxia Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Jun Peng
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Anesthesiology, sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ge Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
20
|
Caruso A, Nicoletti F, Gaetano A, Scaccianoce S. Risk Factors for Alzheimer's Disease: Focus on Stress. Front Pharmacol 2019; 10:976. [PMID: 31551781 PMCID: PMC6746823 DOI: 10.3389/fphar.2019.00976] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022] Open
Abstract
In vulnerable individuals, chronic and persistent stress is an established risk factor for disorders that are comorbid with Alzheimer’s disease (AD), such as hypertension, obesity and metabolic syndrome, and psychiatric disorders. There are no disease-modifying drugs in the treatment of AD, and all phase-3 clinical trials with anti-amyloid drugs (e.g., β- or γ-secretase inhibitors and monoclonal antibodies) did not meet the primary endpoints. There are many reasons for the lack of efficacy of anti-amyloid drugs in AD, the most likely being a late start of treatment, considering that pathophysiological mechanisms underlying synaptic dysfunction and neuronal death begin several decades before the clinical onset of AD. The identification of risk factors is, therefore, an essential step for early treatment of AD with candidate disease-modifying drugs. Preclinical studies suggest that stress, and the resulting activation of the hypothalamic–pituitary–adrenal axis, can induce biochemical abnormalities reminiscent to those found in autoptic brain samples from individuals affected by AD (e.g., increases amyloid precursor protein and tau hyperphosphorylation). In this review, we will critically analyze the current knowledge supporting stress as a potential risk factor for AD.
Collapse
Affiliation(s)
- Alessandra Caruso
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy.,Neuropharmacology Research Unit, I.R.C.C.S. Neuromed, Pozzilli, Italy
| | - Alessandra Gaetano
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| | - Sergio Scaccianoce
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
21
|
Teruel AF. Meet Our Editorial Board Member. Curr Neuropharmacol 2019. [PMCID: PMC6520590 DOI: 10.2174/1570159x1705190405151423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
22
|
Río-Álamos C, Piludu MA, Gerbolés C, Barroso D, Oliveras I, Sánchez-González A, Cañete T, Tapias-Espinosa C, Sampedro-Viana D, Torrubia R, Tobeña A, Fernández-Teruel A. Volumetric brain differences between the Roman rat strains: Neonatal handling effects, sensorimotor gating and working memory. Behav Brain Res 2019; 361:74-85. [DOI: 10.1016/j.bbr.2018.12.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/23/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022]
|
23
|
Muntsant A, Shrivastava K, Recasens M, Giménez-Llort L. Severe Perinatal Hypoxic-Ischemic Brain Injury Induces Long-Term Sensorimotor Deficits, Anxiety-Like Behaviors and Cognitive Impairment in a Sex-, Age- and Task-Selective Manner in C57BL/6 Mice but Can Be Modulated by Neonatal Handling. Front Behav Neurosci 2019; 13:7. [PMID: 30814939 PMCID: PMC6381068 DOI: 10.3389/fnbeh.2019.00007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022] Open
Abstract
Perinatal brain injury (PBI) leads to neurological disabilities throughout life, from motor deficits, cognitive limitations to severe cerebral palsy. Yet, perinatal brain damage has limited therapeutic outcomes. Besides, the immature brain of premature children is at increased risk of hypoxic/ischemic (HI) injury, with males being more susceptible to it and less responsive to protective/therapeutical interventions. Here, we model in male and female C57BL/6 mice, the impact of neonatal HI and the protective effects of neonatal handling (NH), an early life tactile and proprioceptive sensory stimulation. From postnatal day 1 (PND1, modeling pre-term) to PND21 randomized litters received either NH or left undisturbed. HI brain damage occurred by permanent left carotid occlusion followed by hypoxia at PND7 (modeling full-term) in half of the animals. The behavioral and functional screening of the pups at weaning (PND23) and their long-term outcomes (adulthood, PND70) were evaluated in a longitudinal study, as follows: somatic development (weight), sensorimotor functions (reflexes, rods and hanger tests), exploration [activity (ACT) and open-field (OF) test], emotional and anxiety-like behaviors [corner, open-field and dark-light box (DLB) tests], learning and memory [T-maze (TM) and Morris Water-Maze (MWM)]. HI induced similar brain damage in both sexes but affected motor development, sensorimotor functions, induced hyperactivity at weaning, and anxiety-like behaviors and cognitive deficits at adulthood, in a sex- and age-dependent manner. Thus, during ontogeny, HI affected equilibrium especially in females and prehensility in males, but only reflexes at adulthood. Hyperactivity of HI males was normalized at adulthood. HI increased neophobia and other anxiety-like behaviors in males but emotionality in females. Both sexes showed worse short/long-term learning, but memory was more affected in males. Striking neuroprotective effects of NH were found, with significantly lower injury scores, mostly in HI males. At the functional level, NH reversed the impaired reflex responses and improved memory performances in hippocampal-dependent spatial-learning tasks, especially in males. Finally, neuropathological correlates referred to atrophy, neuronal densities and cellularity in the affected areas [hippocampal-CA, caudate/putamen, thalamus, neocortex and corpus callosum (CC)] point out distinct neuronal substrates underlying the sex- and age- functional impacts of these risk/protection interventions on sensorimotor, behavioral and cognitive outcomes from ontogeny to adulthood.
Collapse
Affiliation(s)
- Aida Muntsant
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kalpana Shrivastava
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology & Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mireia Recasens
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology & Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lydia Giménez-Llort
- Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
24
|
Stimmell AC, Baglietto-Vargas D, Moseley SC, Lapointe V, Thompson LM, LaFerla FM, McNaughton BL, Wilber AA. Impaired Spatial Reorientation in the 3xTg-AD Mouse Model of Alzheimer's Disease. Sci Rep 2019; 9:1311. [PMID: 30718609 PMCID: PMC6361963 DOI: 10.1038/s41598-018-37151-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/15/2018] [Indexed: 01/08/2023] Open
Abstract
In early Alzheimer's disease (AD) spatial navigation is impaired; however, the precise cause of this impairment is unclear. Recent evidence suggests that getting lost is one of the first impairments to emerge in AD. It is possible that getting lost represents a failure to use distal cues to get oriented in space. Therefore, we set out to look for impaired use of distal cues for spatial orientation in a mouse model of amyloidosis (3xTg-AD). To do this, we trained mice to shuttle to the end of a track and back to an enclosed start box to receive a water reward. Then, mice were trained to stop in an unmarked reward zone to receive a brain stimulation reward. The time required to remain in the zone for a reward was increased across training, and the track was positioned in a random start location for each trial. We found that 6-month female, but not 3-month female, 6-month male, or 12-month male, 3xTg-AD mice were impaired. 6-month male and female mice had only intracellular pathology and male mice had less pathology, particularly in the dorsal hippocampus. Thus, AD may cause spatial disorientation as a result of impaired use of landmarks.
Collapse
Affiliation(s)
- Alina C Stimmell
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA.
| | | | - Shawn C Moseley
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Valérie Lapointe
- Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Lauren M Thompson
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Frank M LaFerla
- Neurobiology and Behavior, University of California Irvine, Irvine, California, USA
| | - Bruce L McNaughton
- Neurobiology and Behavior, University of California Irvine, Irvine, California, USA
- Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Aaron A Wilber
- Department of Psychology, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA.
| |
Collapse
|
25
|
Nyarko JNK, Quartey MO, Baker GB, Mousseau DD. Can Animal Models Inform on the Relationship between Depression and Alzheimer Disease? CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2019; 64:18-29. [PMID: 29685068 PMCID: PMC6364140 DOI: 10.1177/0706743718772514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The focus on the β-amyloid (Aβ) peptide in clinical Alzheimer disease (AD) as well as in animal models of AD has perhaps biased our understanding of what contributes to the heterogeneity in disease onset and progression. Part of this heterogeneity could reflect the various neuropsychiatric risk factors that present with common symptomatology and can predispose the brain to AD-like changes. One such risk factor is depression. Animal models, particularly mouse models carrying variants of AD-related gene(s), many of which lead to an accumulation of Aβ, suggest that a fundamental shift in depression-related monoaminergic systems (including serotonin and noradrenaline) is a strong indicator of the altered cellular function associated with the earlier(est) stages of AD-related pathology. These changes in monoaminergic neurochemistry could provide for relevant targets for intervention in clinical AD and/or could support a polypharmacy strategy, which might include the targeting of Aβ, in vulnerable populations. Future studies must also include female mice as well as male mice in animal model studies on the relationship between depression and AD.
Collapse
Affiliation(s)
- Jennifer N K Nyarko
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Maa O Quartey
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Glen B Baker
- 2 Department of Psychiatry, Neuroscience and Mental Health Institute, Neurochemical Research Unit, University of Alberta, Edmonton, Alberta, Canada
| | - Darrell D Mousseau
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
26
|
Lesuis SL, Hoeijmakers L, Korosi A, de Rooij SR, Swaab DF, Kessels HW, Lucassen PJ, Krugers HJ. Vulnerability and resilience to Alzheimer's disease: early life conditions modulate neuropathology and determine cognitive reserve. Alzheimers Res Ther 2018; 10:95. [PMID: 30227888 PMCID: PMC6145191 DOI: 10.1186/s13195-018-0422-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/15/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder with a high prevalence among the elderly and a huge personal and societal impact. Recent epidemiological studies have indicated that the incidence and age of onset of sporadic AD can be modified by lifestyle factors such as education, exercise, and (early) stress exposure. Early life adversity is known to promote cognitive decline at a later age and to accelerate aging, which are both primary risk factors for AD. In rodent models, exposure to 'negative' or 'positive' early life experiences was recently found to modulate various measures of AD neuropathology, such as amyloid-beta levels and cognition at later ages. Although there is emerging interest in understanding whether experiences during early postnatal life also modulate AD risk in humans, the mechanisms and possible substrates underlying these long-lasting effects remain elusive. METHODS We review literature and discuss the role of early life experiences in determining later age and AD-related processes from a brain and cognitive 'reserve' perspective. We focus on rodent studies and the identification of possible early determinants of later AD vulnerability or resilience in relation to early life adversity/enrichment. RESULTS Potential substrates and mediators of early life experiences that may influence the development of AD pathology and cognitive decline are: programming of the hypothalamic-pituitary-adrenal axis, priming of the neuroinflammatory response, dendritic and synaptic complexity and function, overall brain plasticity, and proteins such as early growth response protein 1 (EGR1), activity regulated cytoskeleton-associated protein (Arc), and repressor element-1 silencing transcription factor (REST). CONCLUSIONS We conclude from these rodent studies that the early postnatal period is an important and sensitive phase that influences the vulnerability to develop AD pathology. Yet translational studies are required to investigate whether early life experiences also modify AD development in human studies, and whether similar molecular mediators can be identified in the sensitivity to develop AD in humans.
Collapse
Affiliation(s)
- Sylvie L. Lesuis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Lianne Hoeijmakers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Aniko Korosi
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Susanne R. de Rooij
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
- Department of Clinical Epidemiology, Biostatistics & Bio informatics, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Dick F. Swaab
- The Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Helmut W. Kessels
- The Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
- Department of Cellular and Computational Neuroscience, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
27
|
Blockade of adenosine A2A receptors recovers early deficits of memory and plasticity in the triple transgenic mouse model of Alzheimer's disease. Neurobiol Dis 2018; 117:72-81. [DOI: 10.1016/j.nbd.2018.05.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 05/08/2018] [Accepted: 05/30/2018] [Indexed: 11/23/2022] Open
|
28
|
Klakotskaia D, Agca C, Richardson RA, Stopa EG, Schachtman TR, Agca Y. Memory deficiency, cerebral amyloid angiopathy, and amyloid-β plaques in APP+PS1 double transgenic rat model of Alzheimer's disease. PLoS One 2018; 13:e0195469. [PMID: 29641600 PMCID: PMC5895023 DOI: 10.1371/journal.pone.0195469] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/22/2018] [Indexed: 01/31/2023] Open
Abstract
Transgenic rat models of Alzheimer's disease were used to examine differences in memory and brain histology. Double transgenic female rats (APP+PS1) over-expressing human amyloid precursor protein (APP) and presenilin 1 (PS1) and single transgenic rats (APP21) over-expressing human APP were compared with wild type Fischer rats (WT). The Barnes maze assessed learning and memory and showed that both APP21 and APP+PS1 rats made significantly more errors than the WT rats during the acquisition phase, signifying slower learning. Additionally, the APP+PS1 rats made significantly more errors following a retention interval, indicating impaired memory compared to both the APP21 and WT rats. Immunohistochemistry using an antibody against amyloid-β (Aβ) showed extensive and mostly diffuse Aβ plaques in the hippocampus and dense plaques that contained tau in the cortex of the brains of the APP+PS1 rats. Furthermore, the APP+PS1 rats also showed vascular changes, including cerebral amyloid angiopathy with extensive Aβ deposits in cortical and leptomeningeal blood vessel walls and venous collagenosis. In addition to the Aβ accumulation observed in arterial, venous, and capillary walls, APP+PS1 rats also displayed enlarged blood vessels and perivascular space. Overall, the brain histopathology and behavioral assessment showed that the APP+PS1 rats demonstrated behavioral characteristics and vascular changes similar to those commonly observed in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Diana Klakotskaia
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Cansu Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Rachel A. Richardson
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Edward G. Stopa
- Department of Pathology, Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Todd R. Schachtman
- Department of Psychological Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Yuksel Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
29
|
Hoeijmakers L, Amelianchik A, Verhaag F, Kotah J, Lucassen PJ, Korosi A. Early-Life Stress Does Not Aggravate Spatial Memory or the Process of Hippocampal Neurogenesis in Adult and Middle-Aged APP/PS1 Mice. Front Aging Neurosci 2018; 10:61. [PMID: 29563870 PMCID: PMC5845884 DOI: 10.3389/fnagi.2018.00061] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/20/2018] [Indexed: 01/13/2023] Open
Abstract
Life-time experiences are thought to influence the risk to develop the neurodegenerative disorder Alzheimer’s disease (AD). In particular, early-life stress (ES) may modulate the onset and progression of AD. There is recent evidence by our group and others that AD-related neuropathological progression and the associated neuroimmune responses are modulated by ES in the classic APPswe/PS1dE9 mouse model for AD. We here extend our previous study on ES mediated modulation of neuropathology and neuroinflammation and address in the same cohort of mice whether ES accelerates and/or aggravates AD-induced cognitive decline and alterations in the process of adult hippocampal neurogenesis (AHN), a form of brain plasticity. Chronic ES was induced by limiting bedding and nesting material during the first postnatal week and is known to induce cognitive deficits by 4 months in wild type (WT) mice. The onset of cognitive decline in APP/PS1 mice generally starts around 6 months of age. We here tested mice at ages 2–4 months to study acceleration and at ages 8–10 months for aggravation of the APP/PS1 phenotype. ES-exposed WT and APP/PS1 mice were able to perform the object recognition (ORT) and location tasks (OLT) at 2 months of age. Interestingly, at 3 months, ES induced impairments in the performance of the OLT in WT, but not in APP/PS1 mice. APP/PS1 mice exhibited alterations in hippocampal cell proliferation and differentiation, but ES exposure did not further change this. At 9 months, APP/PS1 mice exhibited impaired performance in the Morris Water Maze (MWM) task, as well as reductions in markers of the AHN process, which were not further modulated by ES exposure. In addition, we observed a so far unreported hyperactivity in ES-exposed mice at 8 months of age, which hampered assessment of cognitive functions in the ORT and OLT. In conclusion, while ES has been reported to modulate AD neuropathology and neuroinflammation before, it failed to accelerate or aggravate the decline in cognition or the process of AHN in APP/PS1 mice at ages 2–4 and 8–10 months. Future studies are needed to unravel how ES might affect the vulnerability to develop AD.
Collapse
Affiliation(s)
- Lianne Hoeijmakers
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Anna Amelianchik
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Fleur Verhaag
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Janssen Kotah
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - A Korosi
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
30
|
Baeta-Corral R, Johansson B, Giménez-Llort L. Long-term Treatment with Low-Dose Caffeine Worsens BPSD-Like Profile in 3xTg-AD Mice Model of Alzheimer's Disease and Affects Mice with Normal Aging. Front Pharmacol 2018; 9:79. [PMID: 29497377 PMCID: PMC5818407 DOI: 10.3389/fphar.2018.00079] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/24/2018] [Indexed: 12/26/2022] Open
Abstract
Coffee or caffeine has recently been suggested as prophylaxis for dementia. Although memory problems are hallmarks of Alzheimer's disease, this dementia is also characterized by neuropsychiatric symptoms called Behavioral and Psychological Symptoms of Dementia (BPSD). The impact of preventive/therapeutic strategies on both cognitive and non-cognitive symptoms can be addressed in the 3xTg-AD mice, since they exhibit cognitive but also BPSD-like profiles. Here, we studied the long-term effects of a low dose of caffeine in male 3xTg-AD mice and as compared to age-matched non-transgenic (NTg) counterparts with normal aging. Animals were treated (water or caffeine in drinking water) from adulthood (6 months of age) until middle-aged (13 months of age), that in 3xTg-AD mice correspond to onset of cognitive impairment and advanced stages, respectively. The low caffeine dosing used (0.3 mg/ml) was previously found to give a plasma concentration profile in mice roughly equivalent to that of a human coffee drinker. There were significant effects of caffeine on most behavioral variables, especially those related to neophobia and other anxiety-like behaviors, emotionality, and cognitive flexibility. The 3xTg-AD and NTg mice were differently influenced by caffeine. Overall, the increase of neophobia and other anxiety-related behaviors resulted in an exacerbation of BPSD-like profile in 3xTg-AD mice. Learning and memory, strongly influenced by anxiety in 3xTg-AD mice, got little benefit from caffeine, only shown after a detailed analysis of navigation strategies. The worsened pattern in NTg mice and the use of search strategies in 3xTg-AD mice make both groups more similar. Circadian motor activity showed genotype differences, which were found to be enhanced by caffeine. Selective effects of caffeine on NTg were found in the modulation of behaviors related to emotional profile and risk assessment. Caffeine normalized splenomegaly of 3xTg-AD mice, a physical indicator of their impaired peripheral immune system, and trended to increase their corticosterone levels. Our observations of adverse caffeine effects in an Alzheimer's disease model together with previous clinical observations suggest that an exacerbation of BPSD-like symptoms may partly interfere with the beneficial cognitive effects of caffeine. These results are relevant when coffee-derived new potential treatments for dementia are to be devised and tested.
Collapse
Affiliation(s)
- Raquel Baeta-Corral
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Björn Johansson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden
- Department of Geriatrics, Karolinska University Hospital, Solna, Sweden
| | - Lydia Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Hoeijmakers L, Lesuis SL, Krugers H, Lucassen PJ, Korosi A. A preclinical perspective on the enhanced vulnerability to Alzheimer's disease after early-life stress. Neurobiol Stress 2018; 8:172-185. [PMID: 29888312 PMCID: PMC5991337 DOI: 10.1016/j.ynstr.2018.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 12/13/2022] Open
Abstract
Stress experienced early in life (ES), in the form of childhood maltreatment, maternal neglect or trauma, enhances the risk for cognitive decline in later life. Several epidemiological studies have now shown that environmental and adult life style factors influence AD incidence or age-of-onset and early-life environmental conditions have attracted attention in this respect. There is now emerging interest in understanding whether ES impacts the risk to develop age-related neurodegenerative disorders, and their severity, such as in Alzheimer's disease (AD), which is characterized by cognitive decline and extensive (hippocampal) neuropathology. While this might be relevant for the identification of individuals at risk and preventive strategies, this topic and its possible underlying mechanisms have been poorly studied to date. In this review, we discuss the role of ES in modulating AD risk and progression, primarily from a preclinical perspective. We focus on the possible involvement of stress-related, neuro-inflammatory and metabolic factors in mediating ES-induced effects on later neuropathology and the associated impairments in neuroplasticity. The available studies suggest that the age of onset and progression of AD-related neuropathology and cognitive decline can be affected by ES, and may aggravate the progression of AD neuropathology. These relevant changes in AD pathology after ES exposure in animal models call for future clinical studies to elucidate whether stress exposure during the early-life period in humans modulates later vulnerability for AD.
Collapse
Affiliation(s)
| | | | | | | | - Aniko Korosi
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Morello M, Landel V, Lacassagne E, Baranger K, Annweiler C, Féron F, Millet P. Vitamin D Improves Neurogenesis and Cognition in a Mouse Model of Alzheimer's Disease. Mol Neurobiol 2018; 55:6463-6479. [PMID: 29318446 PMCID: PMC6061182 DOI: 10.1007/s12035-017-0839-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022]
Abstract
The impairment of hippocampal neurogenesis at the early stages of Alzheimer’s disease (AD) is believed to support early cognitive decline. Converging studies sustain the idea that vitamin D might be linked to the pathophysiology of AD and to hippocampal neurogenesis. Nothing being known about the effects of vitamin D on hippocampal neurogenesis in AD, we assessed them in a mouse model of AD. In a previous study, we observed that dietary vitamin D supplementation in female AD-like mice reduced cognitive decline only when delivered during the symptomatic phase. With these data in hand, we wondered whether the consequences of vitamin D administration on hippocampal neurogenesis are stage-dependent. Male wild-type and transgenic AD-like mice (5XFAD model) were fed with a diet containing either no vitamin D (0VD) or a normal dose of vitamin D (NVD) or a high dose of vitamin D (HVD), from month 1 to month 6 (preventive arm) or from month 4 to month 9 (curative arm). Working memory was assessed using the Y-maze, while amyloid burden, astrocytosis, and neurogenesis were quantified using immunohistochemistry. In parallel, the effects of vitamin D on proliferation and differentiation were assayed on primary cultures of murine neural progenitor cells. Improved working memory and neurogenesis were observed when high vitamin D supplementation was administered during the early phases of the disease, while a normal dose of vitamin D increased neurogenesis during the late phases. Conversely, an early hypovitaminosis D increased the number of amyloid plaques in AD mice while a late hypovitaminosis D impaired neurogenesis in AD and WT mice. The observed in vivo vitamin D-associated increased neurogenesis was partially substantiated by an augmented in vitro proliferation but not an increased differentiation of neural progenitors into neurons. Finally, a sexual dimorphism was observed. Vitamin D supplementation improved the working memory of males and females, when delivered during the pre-symptomatic and symptomatic phases, respectively. Our study establishes that (i) neurogenesis is improved by vitamin D in a male mouse model of AD, in a time-dependent manner, and (ii) cognition is enhanced in a gender-associated way. Additional pre-clinical studies are required to further understand the gender- and time-specific mechanisms of action of vitamin D in AD. This may lead to an adaptation of vitamin D supplementation in relation to patient’s gender and age as well as to the stage of the disease.
Collapse
Affiliation(s)
- Maria Morello
- Aix Marseille Univ, CNRS, NICN, Marseille, France.,Clinical Biochemistry, Department of Experimental Medicine and Surgery, Faculty of Medicine, University Hospital of Tor Vergata, Rome, Italy.,Division of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention Faculty of Medicine, University of Tor Vergata, Rome, Italy
| | | | | | | | - Cedric Annweiler
- Department of Neurosciences and Aging, Division of Geriatric Medicine, Angers University Hospital, Angers University Memory Clinic, Research Center on Autonomy and Longevity, UPRES EA 4638, University of Angers, UNAM, Angers, France.,Robarts Research Institute, Department of Medical Biophysics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|
33
|
Lesuis SL, Maurin H, Borghgraef P, Lucassen PJ, Leuven FV, Krugers HJ. Positive and negative early life experiences differentially modulate long term survival and amyloid protein levels in a mouse model of Alzheimer's disease. Oncotarget 2016; 7:39118-39135. [PMID: 27259247 PMCID: PMC5129918 DOI: 10.18632/oncotarget.9776] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 05/12/2016] [Indexed: 11/25/2022] Open
Abstract
Stress has been implicated as a risk factor for the severity and progression of sporadic Alzheimer's disease (AD). Early life experiences determine stress responsivity in later life, and modulate age-dependent cognitive decline. Therefore, we examined whether early life experiences influence AD outcome in a bigenic mouse model which progressively develops combined tau and amyloid pathology (biAT mice).Mice were subjected to either early life stress (ELS) or to 'positive' early handling (EH) postnatally (from day 2 to 9). In biAT mice, ELS significantly compromised long term survival, in contrast to EH which increased life expectancy. In 4 month old mice, ELS-reared biAT mice displayed increased hippocampal Aβ levels, while these levels were reduced in EH-reared biAT mice. No effects of ELS or EH were observed on the brain levels of APP, protein tau, or PSD-95. Dendritic morphology was moderately affected after ELS and EH in the amygdala and medial prefrontal cortex, while object recognition memory and open field performance were not affected. We conclude that despite the strong transgenic background, early life experiences significantly modulate the life expectancy of biAT mice. Parallel changes in hippocampal Aβ levels were evident, without affecting cognition of young adult biAT mice.
Collapse
Affiliation(s)
- Sylvie L. Lesuis
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Herve Maurin
- Experimental Genetics Group - LEGTEGG, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Peter Borghgraef
- Experimental Genetics Group - LEGTEGG, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Paul J. Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Fred Van Leuven
- Experimental Genetics Group - LEGTEGG, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Harm J. Krugers
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Activity of muscarinic, galanin and cannabinoid receptors in the prodromal and advanced stages in the triple transgenic mice model of Alzheimer's disease. Neuroscience 2016; 329:284-93. [PMID: 27223629 DOI: 10.1016/j.neuroscience.2016.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 12/31/2022]
Abstract
Neurochemical alterations in Alzheimer's disease (AD) include cholinergic neuronal loss in the nucleus basalis of Meynert (nbM) and a decrease in densities of the M2 muscarinic receptor subtype in areas related to learning and memory. Neuromodulators present in the cholinergic pathways, such as neuropeptides and neurolipids, control these cognitive processes and have become targets of research in order to understand and treat the pathophysiological and clinical stages of the disease. This is the case of the endocannabinoid and galaninergic systems, which have been found to be up-regulated in AD, and could therefore have a neuroprotective role. In the present study, the functional coupling of Gi/o protein-coupled receptors to GalR1, and the CB1 receptor subtype for endocannabinoids were analyzed in the 3xTg-AD mice model of AD. In addition, the activity mediated by Gi/o protein-coupled M2/4 muscarinic receptor subtypes was also analyzed in brain areas involved in anxiety and cognition. Thus, male mice were studied at 4 and 15months of age (prodromal and advanced stages, respectively) and compared to age-matched non-transgenic (NTg) mice (adult and old, respectively). In 4-month-old 3xTg-AD mice, the [(35)S]GTPγS binding stimulated by galanin was significantly increased in the hypothalamus, but a decrease of functional M2/4 receptors was observed in the posterior amygdala. The CB1 cannabinoid receptor activity was up-regulated in the anterior thalamus at that age. In 15-month-old 3xTg-AD mice, muscarinic receptor activity was found to be increased in motor cortex, while CB1 activity was decreased in nbM. No changes were found in GalR1-mediated activity at this age. Our results provide further evidence of the relevance of limbic areas in the prodromal stage of AD, the profile of which is characterized by anxiety. The up-regulation of galaninergic and endocannabinoid systems support the hypothesis of their neuroprotective roles, and these are established prior to the onset of clear clinical cognitive symptoms of the disease.
Collapse
|
35
|
Bagci E, Aydin E, Mihasan M, Maniu C, Hritcu L. Anxiolytic and antidepressant-like effects ofFerulago angulataessential oil in the scopolamine rat model of Alzheimer's disease. FLAVOUR FRAG J 2015. [DOI: 10.1002/ffj.3289] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Eyup Bagci
- Department of Biology, Faculty of Science; Firat University; 23119 Elazig Turkey
| | - Emel Aydin
- Department of Biology, Faculty of Science; Firat University; 23119 Elazig Turkey
| | - Marius Mihasan
- Department of Biology; Alexandru Ioan Cuza University; Bd. Carol I, No.11 Iasi 700506 Romania
| | - Calin Maniu
- Department of Biology; Alexandru Ioan Cuza University; Bd. Carol I, No.11 Iasi 700506 Romania
| | - Lucian Hritcu
- Department of Biology; Alexandru Ioan Cuza University; Bd. Carol I, No.11 Iasi 700506 Romania
| |
Collapse
|
36
|
Torres-Lista V, Giménez-Llort L. Early postnatal handling and environmental enrichment improve the behavioral responses of 17-month-old 3xTg-AD and non-transgenic mice in the Forced Swim Test in a gender-dependent manner. Behav Processes 2015; 120:120-7. [PMID: 26431900 DOI: 10.1016/j.beproc.2015.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/14/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022]
Abstract
Forced Swimming Test (FST) models behavioural despair in animals by loss of motivation to respond or the refusal to escape. The present study was aimed at characterizing genetic (genotype and gender) and environmental factors (age/stage of disease and rearing conditions: C, standard; H, early postnatal handling; EE, environmental enrichment consisting in physical exercise as well as social and object enrichment) that may modulate the poor behavioural and cognitive flexibility response we have recently described in 12-month-old male 3xTg-AD mice in the FST. The comprehensive analysis of the ethogram shown in the FST considered the intervals of the test (0-2 and 2-6min), all the elicited behavioural responses (immobility, swimming and climbing) and their features (total duration and frequency of episodes). The long persistence of behaviours found in 17-month-old (late-stages of disease) 3xTg-AD mice was comparable to that recently described in males at 12 months of age (beginning of advanced stages) but also suggested increased age-dependent frailty in both genotypes. The poor behavioral flexibility of 3xTg-AD mice to elicit the behavioural despair shown by the NTg mice, was also found in the female gender. Finally, the present work demonstrates that early-life interventions were able to improve the time and frequency of episodes of immobility, being more evident in the female gender of both old NTg and 3xTg-AD mice. Ontogenic modulation by early-postnatal handling resulted in a more effective long-term improvement of the elicited behaviours in the FST than that achieved by environmental enrichment. The results talk in favor of the beneficence of early-life interventions on ageing in both healthy and disease conditions.
Collapse
Affiliation(s)
- Virginia Torres-Lista
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain; Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lydia Giménez-Llort
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain; Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
37
|
Río-Ȧlamos C, Oliveras I, Cañete T, Blázquez G, Martínez-Membrives E, Tobeña A, Fernández-Teruel A. Neonatal handling decreases unconditioned anxiety, conditioned fear, and improves two-way avoidance acquisition: a study with the inbred Roman high (RHA-I)- and low-avoidance (RLA-I) rats of both sexes. Front Behav Neurosci 2015. [PMID: 26217201 PMCID: PMC4498386 DOI: 10.3389/fnbeh.2015.00174] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The present study evaluated the long-lasting effects of neonatal handling (NH; administered during the first 21 days of life) on unlearned and learned anxiety-related responses in inbred Roman High- (RHA-I) and Low-avoidance (RLA-I) rats. To this aim, untreated and neonatally-handled RHA-I and RLA-I rats of both sexes were tested in the following tests/tasks: a novel object exploration (NOE) test, the elevated zero maze (ZM) test, a “baseline acoustic startle” (BAS) test, a “context-conditioned fear” (CCF) test and the acquisition of two-way active—shuttle box—avoidance (SHAV). RLA-I rats showed higher unconditioned (novel object exploration test -“NOE”-, elevated zero maze test -“ZM”-, BAS), and conditioned (CCF, SHAV) anxiety. NH increased exploration of the novel object in the NOE test as well as exploration of the open sections of the ZM test in both rat strains and sexes, although the effects were relatively more marked in the (high anxious) RLA-I strain and in females. NH did not affect BAS, but reduced CCF in both strains and sexes, and improved shuttle box avoidance acquisition especially in RLA-I (and particularly in females) and in female RHA-I rats. These are completely novel findings, which indicate that even some genetically-based anxiety/fear-related phenotypes can be significantly modulated by previous environmental experiences such as the NH manipulation.
Collapse
Affiliation(s)
- Cristóbal Río-Ȧlamos
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, School of Medicine, Institute of Neurosciences, Autonomous University of Barcelona Barcelona, Spain
| | - Ignasi Oliveras
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, School of Medicine, Institute of Neurosciences, Autonomous University of Barcelona Barcelona, Spain
| | - Toni Cañete
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, School of Medicine, Institute of Neurosciences, Autonomous University of Barcelona Barcelona, Spain
| | - Gloria Blázquez
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, School of Medicine, Institute of Neurosciences, Autonomous University of Barcelona Barcelona, Spain
| | - Esther Martínez-Membrives
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, School of Medicine, Institute of Neurosciences, Autonomous University of Barcelona Barcelona, Spain
| | - Adolf Tobeña
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, School of Medicine, Institute of Neurosciences, Autonomous University of Barcelona Barcelona, Spain
| | - Alberto Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, School of Medicine, Institute of Neurosciences, Autonomous University of Barcelona Barcelona, Spain
| |
Collapse
|