1
|
Ohta H, Nozawa T, Nakano T, Morimoto Y, Ishizuka T. Nonlinear age-related differences in probabilistic learning in mice: A 5-armed bandit task study. Neurobiol Aging 2024; 142:8-16. [PMID: 39029360 DOI: 10.1016/j.neurobiolaging.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/21/2024]
Abstract
This study explores the impact of aging on reinforcement learning in mice, focusing on changes in learning rates and behavioral strategies. A 5-armed bandit task (5-ABT) and a computational Q-learning model were used to evaluate the positive and negative learning rates and the inverse temperature across three age groups (3, 12, and 18 months). Results showed a significant decline in the negative learning rate of 18-month-old mice, which was not observed for the positive learning rate. This suggests that older mice maintain the ability to learn from successful experiences while decreasing the ability to learn from negative outcomes. We also observed a significant age-dependent variation in inverse temperature, reflecting a shift in action selection policy. Middle-aged mice (12 months) exhibited higher inverse temperature, indicating a higher reliance on previous rewarding experiences and reduced exploratory behaviors, when compared to both younger and older mice. This study provides new insights into aging research by demonstrating that there are age-related differences in specific components of reinforcement learning, which exhibit a non-linear pattern.
Collapse
Affiliation(s)
- Hiroyuki Ohta
- Department of Pharmacology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.
| | - Takashi Nozawa
- Mejiro University, 4-31-1 Naka-Ochiai, Shinjuku, Tokyo 161-8539, Japan
| | - Takashi Nakano
- Department of Computational Biology, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi 470-1192, Japan; International Center for Brain Science (ICBS), Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi 470-1192, Japan
| | - Yuji Morimoto
- Department of Physiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Toshiaki Ishizuka
- Department of Pharmacology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| |
Collapse
|
2
|
Gómez-Pascual A, Glikman DM, Ng HX, Tomkins JE, Lu L, Xu Y, Ashbrook DG, Kaczorowski C, Kempermann G, Killmar J, Mozhui K, Ohlenschläger O, Aebersold R, Ingram DK, Williams EG, Williams RW, Overall RW, Jucker M, de Bakker DEM. The Pgb1 locus controls glycogen aggregation in astrocytes of the aged hippocampus without impacting cognitive function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.22.567373. [PMID: 38045339 PMCID: PMC10690248 DOI: 10.1101/2023.11.22.567373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
In aged humans and mice, aggregates of hypobranched glycogen molecules called polyglucosan bodies (PGBs) accumulate in hippocampal astrocytes. PGBs are known to drive cognitive decline in neurological diseases but remain largely unstudied in the context of typical brain aging. Here, we show that PGBs arise in autophagy-dysregulated astrocytes of the aged C57BL/6J mouse hippocampus. To map the genetic cause of age-related PGB accumulation, we quantified PGB burden in 32 fully sequenced BXD-recombinant inbred mouse strains, which display a 400-fold variation in hippocampal PGB burden at 16-18 months of age. A major modifier locus was mapped to chromosome 1 at 72-75 Mb, which we defined as the Pgb1 locus. To evaluate candidate genes and downstream mechanisms by which Pgb1 controls the aggregation of glycogen, extensive hippocampal transcriptomic and proteomic datasets were produced for aged mice of the BXD family. We utilized these datasets to identify Smarcal1 and Usp37 as potential regulators of PGB accumulation. To assess the effect of PGB burden on age-related cognitive decline, we performed phenome-wide association scans, transcriptomic analyses as well as conditioned fear memory and Y-maze testing. Importantly, we did not find any evidence suggesting a negative impact of PGBs on cognition. Taken together, our study demonstrates that the Pgb1 locus controls glycogen aggregation in astrocytes of the aged hippocampus without affecting age-related cognitive decline.
Collapse
Affiliation(s)
- A Gómez-Pascual
- Department of Information and Communications Engineering, University of Murcia, Murcia, Spain
| | | | - H X Ng
- Department of Cognitive Science University of California, San Diego, USA
| | - J E Tomkins
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - L Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Center, Memphis, TN, USA
| | - Y Xu
- Department of Cellular Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - D G Ashbrook
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Center, Memphis, TN, USA
| | | | - G Kempermann
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - J Killmar
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Center, Memphis, TN, USA
| | - K Mozhui
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Center, Memphis, TN, USA
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Center, Memphis, TN, USA
| | - O Ohlenschläger
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - R Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich. Zurich, Switzerland
| | - D K Ingram
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - E G Williams
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | - R W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Center, Memphis, TN, USA
| | - R W Overall
- Humboldt University of Berlin, Berlin, Germany
| | - M Jucker
- Department of Cellular Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - D E M de Bakker
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| |
Collapse
|
3
|
Brown RE. Measuring the replicability of our own research. J Neurosci Methods 2024; 406:110111. [PMID: 38521128 DOI: 10.1016/j.jneumeth.2024.110111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
In the study of transgenic mouse models of neurodevelopmental and neurodegenerative disorders, we use batteries of tests to measure deficits in behaviour and from the results of these tests, we make inferences about the mental states of the mice that we interpret as deficits in "learning", "memory", "anxiety", "depression", etc. This paper discusses the problems of determining whether a particular transgenic mouse is a valid mouse model of disease X, the problem of background strains, and the question of whether our behavioural tests are measuring what we say they are. The problem of the reliability of results is then discussed: are they replicable between labs and can we replicate our results in our own lab? This involves the study of intra- and inter- experimenter reliability. The variables that influence replicability and the importance of conducting a complete behavioural phenotype: sensory, motor, cognitive and social emotional behaviour are discussed. Then the thorny question of failure to replicate is examined: Is it a curse or a blessing? Finally, the role of failure in research and what it tells us about our research paradigms is examined.
Collapse
Affiliation(s)
- Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
4
|
Zhang Y, Chen H, Zeng M, Guo P, Liu M, Cao B, Wang R, Hao F, Zheng X, Feng W. Futoquinol improves Aβ 25-35-induced memory impairment in mice by inhibiting the activation of p38MAPK through the glycolysis pathway and regulating the composition of the gut microbiota. Phytother Res 2024; 38:1799-1814. [PMID: 38330236 DOI: 10.1002/ptr.8136] [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: 06/21/2023] [Revised: 12/18/2023] [Accepted: 01/13/2024] [Indexed: 02/10/2024]
Abstract
Futoquinol (Fut) is a compound extracted from Piper kadsura that has a nerve cell protection effect. However, it is unclear whether Fut has protective effects in Alzheimer's disease (AD). In this study, we aimed to explore the therapeutic effect of Fut in AD and its underlying mechanism. UPLC-MS/MS method was performed to quantify Fut in the hippocampus of mice brain. The cognition ability, neuronal and mitochondria damage, and levels of Aβ1-42, Aβ1-40, p-Tau, oxidative stress, apoptosis, immune cells, and inflammatory factors were measured in Aβ25-35-induced mice. The content of bacterial meta-geometry was predicted in the microbial composition based on 16S rDNA. The protein levels of HK II, p-p38MAPK, and p38MAPK were detected. PC-12 cells were cultured in vitro, and glucose was added to activate glycolysis to further explore the mechanism of action of Fut intervention in AD. Fut improved the memory and learning ability of Aβ25-35 mice, and reduced neuronal damage and the deposition of Aβ and Tau proteins. Moreover, Fut reduced mitochondrial damage, the levels of oxidative stress, apoptosis, and inflammatory factors. Fut significantly inhibited the expression of HK II and p-p38MAPK proteins. The in vitro experiment showed that p38MAPK was activated and Fut action inhibited after adding 10 mM glucose. Fut might inhibit the activation of p38MAPK through the glycolysis pathway, thereby reducing oxidative stress, apoptosis, and inflammatory factors and improving Aβ25-35-induced memory impairment in mice. These data provide pharmacological rationale for Fut in the treatment of AD.
Collapse
Affiliation(s)
- Yuhan Zhang
- College of pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Hui Chen
- College of pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Mengnan Zeng
- College of pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Pengli Guo
- College of pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Meng Liu
- College of pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Bing Cao
- College of pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Ru Wang
- College of pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Fengxiao Hao
- College of pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Xiaoke Zheng
- College of pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Weisheng Feng
- College of pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| |
Collapse
|
5
|
McDermott KD, Frechou MA, Jordan JT, Martin SS, Gonçalves JT. Delayed formation of neural representations of space in aged mice. Aging Cell 2023; 22:e13924. [PMID: 37491802 PMCID: PMC10497831 DOI: 10.1111/acel.13924] [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: 03/29/2023] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Aging is associated with cognitive deficits, with spatial memory being very susceptible to decline. The hippocampal dentate gyrus (DG) is important for processing spatial information in the brain and is particularly vulnerable to aging, yet its sparse activity has led to difficulties in assessing changes in this area. Using in vivo two-photon calcium imaging, we compared DG neuronal activity and representations of space in young and aged mice walking on an unfamiliar treadmill. We found that calcium activity was significantly higher and less tuned to location in aged mice, resulting in decreased spatial information encoded in the DG. However, with repeated exposure to the same treadmill, both spatial tuning and information levels in aged mice became similar to young mice, while activity remained elevated. Our results show that spatial representations of novel environments are impaired in the aged hippocampus and gradually improve with increased familiarity. Moreover, while the aged DG is hyperexcitable, this does not disrupt neural representations of familiar environments.
Collapse
Affiliation(s)
- Kelsey D. McDermott
- Dominick P. Purpura Department of Neuroscience and Gottesmann Institute for Stem Cell Biology and Regenerative MedicineAlbert Einstein College of MedicineBronxNew YorkUSA
| | - M. Agustina Frechou
- Dominick P. Purpura Department of Neuroscience and Gottesmann Institute for Stem Cell Biology and Regenerative MedicineAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Jake T. Jordan
- Dominick P. Purpura Department of Neuroscience and Gottesmann Institute for Stem Cell Biology and Regenerative MedicineAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Sunaina S. Martin
- Dominick P. Purpura Department of Neuroscience and Gottesmann Institute for Stem Cell Biology and Regenerative MedicineAlbert Einstein College of MedicineBronxNew YorkUSA
| | - J. Tiago Gonçalves
- Dominick P. Purpura Department of Neuroscience and Gottesmann Institute for Stem Cell Biology and Regenerative MedicineAlbert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
6
|
Greene SM, Klein PR, Alcala GA, Bustamante I, Bordas B, Johnson A, Vu V, Uhm SY, Gould GG. Aging to 24 months increased C57BL/6J mouse social sniffing and hippocampal Neto1 levels, and impaired female spatial learning. Behav Processes 2023; 211:104929. [PMID: 37586617 PMCID: PMC11441572 DOI: 10.1016/j.beproc.2023.104929] [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: 02/20/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/18/2023]
Abstract
Understanding how natural aging impacts rodent performance in translational behavior tests is critical to teasing apart impairments due to age-related decline from neurodegenerative disorder modeling. Reduced neuropilin and tolloid-like 1 (NETO1), an accessory protein of ionotropic glutamate receptors involved in synaptic plasticity, was associated with Alzheimer's disease, yet aging effects on Neto1 remain unclear. For these reasons, our goal was to characterize how Neto1 expression corresponded with social, repetitive, and spatial learning behaviors and stress response across the C57BL/6J mouse lifespan. We measured social preferences in three-chamber tests, and motor stereotypies by marble burying. Cognitive flexibility is typically assessed in the Morris water maze (MWM), wherein C57BL/6J mice exhibit deficits with age. However, fatigue or locomotor impairment may confound interpretation of MWM performance. Therefore, we used a less arduous water T-maze (WTM) to compare spatial learning flexibility in 2, 9-15, and 24-month-old male and female mice to test the hypothesis that deficits would emerge with age. In both sexes, 9-15-month-olds made more chamber entries during social preference tests, while 2-month-olds did less social sniffing than aged mice. No age or sex differences emerged in marble burying or serum corticosterone measurements. In 24-month-olds hippocampal Neto1was increased relative to 2-month-olds, and male cognitive flexibility was strong, while spatial learning and reversal learning of 24-month-old females was impaired in WTM irrespective of Neto1 expression. The WTM is a useful alternative assessment for cognitive flexibility deficits in aged mice, and the role of hippocampal Neto1 in promoting social sniffing is of interest.
Collapse
Affiliation(s)
- Susan M Greene
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; University of the Incarnate Word, 4301 Broadway, San Antonio, TX 78209, USA
| | - Preston R Klein
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Gloria-Andrea Alcala
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; University of the Incarnate Word, 4301 Broadway, San Antonio, TX 78209, USA
| | - Isabela Bustamante
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Trinity University, One Trinity Place, San Antonio, TX 78212, USA
| | - Blanka Bordas
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA 24016, USA
| | - Alexia Johnson
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Howard University, Washington D.C. 20059, USA
| | - Vy Vu
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - So Yeon Uhm
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Georgianna G Gould
- Department of Cellular & Integrative Physiology, Center for Biomedical Neuroscience, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
7
|
Brandt A, Kromm F, Hernández-Arriaga A, Martínez Sánchez I, Bozkir HÖ, Staltner R, Baumann A, Camarinha-Silva A, Heijtz RD, Bergheim I. Cognitive Alterations in Old Mice Are Associated with Intestinal Barrier Dysfunction and Induced Toll-like Receptor 2 and 4 Signaling in Different Brain Regions. Cells 2023; 12:2153. [PMID: 37681885 PMCID: PMC10486476 DOI: 10.3390/cells12172153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Emerging evidence implicate the 'microbiota-gut-brain axis' in cognitive aging and neuroinflammation; however, underlying mechanisms still remain to be elucidated. Here, we assessed if potential alterations in intestinal barrier function and microbiota composition as well as levels of two key pattern-recognition receptors namely Toll-like receptor (TLR) 2 and TLR4, in blood and different brain regions, and depending signaling cascades are paralleling aging associated alterations of cognition in healthy aging mice. Cognitive function was assessed in the Y-maze and intestinal and brain tissue and blood were collected in young (4 months old) and old (24 months old) male C57BL/6 mice to determine intestinal microbiota composition by Illumina amplicon sequencing, the concentration of TLR2 and TLR4 ligands in plasma and brain tissue as well as to determine markers of intestinal barrier function, senescence and TLR2 and TLR4 signaling. Cognitive function was significantly impaired in old mice. Also, in old mice, intestinal microbiota composition was significantly altered, while the relative abundance of Gram-negative or Gram-positive bacteria in the small and large intestines at different ages was not altered. Moreover, intestinal barrier function was impaired in small intestine of old mice, and the levels of TLR2 and TLR4 ligands were also significantly higher in both portal and peripheral blood. Furthermore, levels of TLR2 and TLR4 ligands, and downstream markers of TLR signaling were higher in the hippocampal and prefrontal cortex of old mice compared to young animals. Taken together, our results suggest that even in 'healthy' aging, cognitive function is impaired in mice going along with an increased intestinal translocation of TLR ligands and alterations of TLR signaling in several brain regions.
Collapse
Affiliation(s)
- Annette Brandt
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, 1090 Vienna, Austria
| | - Franziska Kromm
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, 1090 Vienna, Austria
| | - Angélica Hernández-Arriaga
- Animal Nutrition Department, Institute of Animal Science, University of Hohenheim, 70593 Stuttgart, Germany
| | - Inés Martínez Sánchez
- Department of Neuroscience, Karolinska Institute, Biomedicum, 17177 Stockholm, Sweden
| | - Haktan Övül Bozkir
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, 1090 Vienna, Austria
| | - Raphaela Staltner
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, 1090 Vienna, Austria
| | - Anja Baumann
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, 1090 Vienna, Austria
| | - Amélia Camarinha-Silva
- Animal Nutrition Department, Institute of Animal Science, University of Hohenheim, 70593 Stuttgart, Germany
| | - Rochellys Diaz Heijtz
- Department of Neuroscience, Karolinska Institute, Biomedicum, 17177 Stockholm, Sweden
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
8
|
Amelchenko EM, Bezriadnov DV, Chekhov OA, Anokhin KV, Lazutkin AA, Enikolopov G. Age-related decline in cognitive flexibility is associated with the levels of hippocampal neurogenesis. Front Neurosci 2023; 17:1232670. [PMID: 37645372 PMCID: PMC10461065 DOI: 10.3389/fnins.2023.1232670] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/28/2023] [Indexed: 08/31/2023] Open
Abstract
Aging is associated with impairments in learning, memory, and cognitive flexibility, as well as a gradual decline in hippocampal neurogenesis. We investigated the performance of 6-and 14-month-old mice (considered mature adult and late middle age, respectively) in learning and memory tasks based on the Morris water maze (MWM) and determined their levels of preceding and current neurogenesis. While both age groups successfully performed in the spatial version of MWM (sMWM), the older mice were less efficient compared to the younger mice when presented with modified versions of the MWM that required a reassessment of the previously acquired experience. This was detected in the reversal version of MWM (rMWM) and was particularly evident in the context discrimination MWM (cdMWM), a novel task that required integrating various distal cues, local cues, and altered contexts and adjusting previously used search strategies. Older mice were impaired in several metrics that characterize rMWM and cdMWM, however, they showed improvement and narrowed the performance gap with the younger mice after additional training. Furthermore, we analyzed the adult-born mature and immature neurons in the hippocampal dentate gyrus and found a significant correlation between neurogenesis levels in individual mice and their performance in the tasks demanding cognitive flexibility. These results provide a detailed description of the age-related changes in learning and memory and underscore the importance of hippocampal neurogenesis in supporting cognitive flexibility.
Collapse
Affiliation(s)
- Evgeny M. Amelchenko
- Center for Developmental Genetics, Stony Brook, NY, United States
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| | | | - Olga A. Chekhov
- Center for Developmental Genetics, Stony Brook, NY, United States
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| | - Konstantin V. Anokhin
- P.K. Anokhin Research Institute of Normal Physiology RAS, Moscow, Russia
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow, Russia
| | - Alexander A. Lazutkin
- Center for Developmental Genetics, Stony Brook, NY, United States
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
- Institute of Higher Nervous Activity and Neurophysiology RAS, Moscow, Russia
| | - Grigori Enikolopov
- Center for Developmental Genetics, Stony Brook, NY, United States
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
9
|
Lyu J, Nagarajan R, Kambali M, Wang M, Rudolph U. Selective inhibition of somatostatin-positive dentate hilar interneurons induces age-related cellular changes and cognitive dysfunction. PNAS NEXUS 2023; 2:pgad134. [PMID: 37168673 PMCID: PMC10165806 DOI: 10.1093/pnasnexus/pgad134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/19/2023] [Accepted: 04/04/2023] [Indexed: 05/13/2023]
Abstract
The cellular basis of age-related impairments of hippocampal function is not fully understood. In order to evaluate the role of somatostatin-positive (Sst+) interneurons in the dentate gyrus (DG) hilus in this process, we chemogenetically inhibited Sst+ interneurons in the DG hilus. Chronic chemogenetic inhibition (CCI) of these neurons resulted in increased c-Fos staining in the DG hilus, a decrease in the percentage of GAD67- and of Sst-expressing interneurons in the DG, and increased microglial activation in DG, CA3, and CA1. Total dendritic length and spine density were reduced in DG and CA1, suggesting reduced dendritic complexity. Behaviorally, the recognition index in an object recognition task and the percentage of spontaneous alternations in the Y-maze were decreased, while in both initial and reversal learning in the Morris water maze, the latencies to find the hidden platform were increased, suggesting cognitive dysfunction. Our findings establish a causal role for a reduced function of Sst+ interneurons in the DG hilus for cognitive decline and suggest that this reduced function may contribute to age-related impairments of learning and memory. Furthermore, our CCI mice may represent a cellularly defined model of hippocampal aging.
Collapse
Affiliation(s)
- Jinrui Lyu
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
- Neuroscience Program, College of Liberal Arts and Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
| | - Rajasekar Nagarajan
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
| | - Maltesh Kambali
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
| | - Muxiao Wang
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
- Neuroscience Program, College of Liberal Arts and Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
| | | |
Collapse
|
10
|
McDermott KD, Frechou MA, Jordan JT, Martin SS, Gonçalves JT. Delayed formation of neural representations of space in aged mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.03.531021. [PMID: 37034736 PMCID: PMC10081265 DOI: 10.1101/2023.03.03.531021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Aging is associated with cognitive deficits, with spatial memory being very susceptible to decline. The hippocampal dentate gyrus (DG) is important for processing spatial information in the brain and is particularly vulnerable to aging, yet its sparse activity has led to difficulties in assessing changes in this area. Using in vivo two-photon calcium imaging, we compared DG neuronal activity and representations of space in young and aged mice walking on an unfamiliar treadmill. We found that calcium activity was significantly higher and less tuned to location in aged mice, resulting in decreased spatial information encoded in the DG. However, with repeated exposure to the same treadmill, both spatial tuning and information levels in aged mice became similar to young mice, while activity remained elevated. Our results show that spatial representations of novel environments are impaired in the aged hippocampus and gradually improve with increased familiarity. Moreover, while the aged DG is hyperexcitable, this does not disrupt neural representations of familiar environments.
Collapse
Affiliation(s)
- Kelsey D. McDermott
- Dominick P. Purpura Department of Neuroscience and Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - M. Agustina Frechou
- Dominick P. Purpura Department of Neuroscience and Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Jake T. Jordan
- Dominick P. Purpura Department of Neuroscience and Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Sunaina S. Martin
- Dominick P. Purpura Department of Neuroscience and Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - J. Tiago Gonçalves
- Dominick P. Purpura Department of Neuroscience and Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
11
|
Castillo-Mariqueo L, Giménez-Llort L. Impact of Behavioral Assessment and Re-Test as Functional Trainings That Modify Survival, Anxiety and Functional Profile (Physical Endurance and Motor Learning) of Old Male and Female 3xTg-AD Mice and NTg Mice with Normal Aging. Biomedicines 2022; 10:973. [PMID: 35625710 PMCID: PMC9138863 DOI: 10.3390/biomedicines10050973] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 02/01/2023] Open
Abstract
Longitudinal approaches for disease-monitoring in old animals face survival and frailty limitations, but also assessment and re-test bias on genotype and sex effects. The present work investigated these effects on 56 variables for behavior, functional profile, and biological status of male and female 3xTg-AD mice and NTg counterparts using two designs: (1) a longitudinal design: naïve 12-month-old mice re-tested four months later; and (2) a cross-sectional design: naïve 16-month-old mice compared to those re-tested. The results confirmed the impact as (1) improvement of survival (NTg rested females), variability of gait (3xTg-AD 16-month-old re-tested and naïve females), physical endurance (3xTg-AD re-tested females), motor learning (3xTg-AD and NTg 16-month-old re-tested females), and geotaxis (3xTg-AD naïve 16-month-old males); but (2) worse anxiety (3xTg-AD 16-month-old re-tested males), HPA axis (3xTg-AD 16-month-old re-tested and naïve females) and sarcopenia (3xTg-AD 16-month-old naïve females). Males showed more functional correlations than females. The functional profile, biological status, and their correlation are discussed as relevant elements for AD-pathology. Therefore, repetition of behavioral batteries could be considered training by itself, with some variables sensitive to genotype, sex, and re-test. In the AD-genotype, females achieved the best performance in physical endurance and motor learning, while males showed a deterioration in most studied variables.
Collapse
Affiliation(s)
- Lidia Castillo-Mariqueo
- 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
| | - Lydia 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
|
12
|
Wang N, Tan Y, Zhou Q, Mao R, Yang Y. The impairment of the hippocampal neuro-vascular unit precedes changes in spatial cognition in naturally aged rats. Neurosci Lett 2022; 776:136580. [DOI: 10.1016/j.neulet.2022.136580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 11/30/2022]
|
13
|
Li Z, Chen X, Vong JSL, Zhao L, Huang J, Yan LYC, Ip B, Wing YK, Lai HM, Mok VCT, Ko H. Systemic GLP-1R agonist treatment reverses mouse glial and neurovascular cell transcriptomic aging signatures in a genome-wide manner. Commun Biol 2021; 4:656. [PMID: 34079050 PMCID: PMC8172568 DOI: 10.1038/s42003-021-02208-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/11/2021] [Indexed: 11/09/2022] Open
Abstract
Pharmacological reversal of brain aging is a long-sought yet challenging strategy for the prevention and treatment of age-related neurodegeneration, due to the diverse cell types and complex cellular pathways impacted by the aging process. Here, we report the genome-wide reversal of transcriptomic aging signatures in multiple major brain cell types, including glial and mural cells, by systemic glucagon-like peptide-1 receptor (GLP-1R) agonist (GLP-1RA) treatment. The age-related expression changes reversed by GLP-1RA encompass both shared and cell type-specific functional pathways that are implicated in aging and neurodegeneration. Concomitantly, Alzheimer's disease (AD)-associated transcriptomic signature in microglia that arises from aging is reduced. These results show the feasibility of reversing brain aging by pharmacological means, provide mechanistic insights into the neurological benefits of GLP-1RAs, and imply that GLP-1R agonism may be a generally applicable pharmacological intervention for patients at risk of age-related neurodegeneration.
Collapse
Affiliation(s)
- Zhongqi Li
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xinyi Chen
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Joaquim S L Vong
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Lei Zhao
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Junzhe Huang
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Leo Y C Yan
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Bonaventure Ip
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yun Kwok Wing
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hei-Ming Lai
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Vincent C T Mok
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Chow Yuk Ho Technology Centre for Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | - Ho Ko
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Chow Yuk Ho Technology Centre for Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Peter Hung Pain Research Institute, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
14
|
Liu W, Cao Y, Lin Y, Tan KS, Zhao H, Guo H, Tan W. Enhancement of Fear Extinction Memory and Resistance to Age-Related Cognitive Decline in Butyrylcholinesterase Knockout Mice and ( R)-Bambuterol Treated Mice. BIOLOGY 2021; 10:biology10050404. [PMID: 34062954 PMCID: PMC8147965 DOI: 10.3390/biology10050404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/24/2021] [Accepted: 05/01/2021] [Indexed: 12/24/2022]
Abstract
Simple Summary Fear extinction is the driving mechanism to reduce the fear response, and it is the basis of exposure-based cognitive-behavioral therapy. Butyrylcholinesterase (BChE) seems to be involved in regulating cognitive function, and its relationship with fear extinction memory has not been reported. BChE knockout mice and wild-type mice with administration of (R)-bambuterol, a BChE inhibitor, were used in this study. In addition to immunohistochemistry and metabolite analysis using mass spectrometry imaging, the influence of age on the conditioned fear test, Morris water maze experiment, and open field test were carefully evaluated. Our results showed that BChE inhibition accelerates the fear extinction memory in mice and delays the cognitive decline in the early stages of aging. Abstract Butyrylcholinesterase (BChE) is detected in plaques preferentially in Alzheimer’s disease (AD) and may be associated with stress disorders. However, the physiological function of BChE in the central nervous system remains to be further investigated. BChE knockout (KO) mice and wild-type (WT) mice with orally or intranasal administration of (R)-bambuterol were used to explore the effect of BChE on behavior changes. (R)-bambuterol is a specific and reversible inhibitor of BChE. The behavior changes were evaluated and compared among 3–10 month old mice. Our finding showed that BChE KO and (R)-bambuterol administration enhanced episodic memory, including fear conditioning memory and fear extinction memory in fear conditioning and fear extinction test. BChE KO and (R)-bambuterol administered mice rescued age-related spatial memory and general activity in the water maze test and open field test. The brain metabolomics were imaged using a desorption electrospray ionization mass spectrometry imaging (DESI-MSI). The image of DESI-MS demonstrated that glutamine content increased in the brain of BChE KO mice. In conclusion, this study found that inhibition of BChE ameliorated episodic and spatial memories. This study also suggested that (R)-bambuterol as a BChE inhibitor has the potential application in the treatment of post-traumatic stress disorder (PTSD) and early cognitive decline.
Collapse
Affiliation(s)
- Weiwei Liu
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; (W.L.); (Y.L.); (H.Z.); (H.G.)
| | - Yan Cao
- YZ Health-tech Inc., Hengqin District, Zhuhai 519000, China; (Y.C.); (K.S.T.)
- School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yue Lin
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; (W.L.); (Y.L.); (H.Z.); (H.G.)
| | - Keai Sinn Tan
- YZ Health-tech Inc., Hengqin District, Zhuhai 519000, China; (Y.C.); (K.S.T.)
- School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Haishan Zhao
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; (W.L.); (Y.L.); (H.Z.); (H.G.)
| | - Haihua Guo
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; (W.L.); (Y.L.); (H.Z.); (H.G.)
| | - Wen Tan
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China; (W.L.); (Y.L.); (H.Z.); (H.G.)
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Correspondence:
| |
Collapse
|