1
|
Han YY, Li K, Hu JY, Wu JC, Li X, Liu DX, Li CH. Gender Differences in Dendritic Damage, Gut Microbiota Dysbiosis, and Cognitive Impairment During Aging Processes. CNS Neurosci Ther 2024; 30:e70164. [PMID: 39723486 DOI: 10.1111/cns.70164] [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: 03/28/2024] [Revised: 08/20/2024] [Accepted: 10/20/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Cognitive impairment is a common and feared characteristic of aging processes, and one key mechanism of cognition is hippocampal synaptic structure. Previous studies have reported that gut microbiota dysbiosis occurred in neurodegenerative diseases and other brain disorders with cognitive impairment. However, it is not clear how gender differences affect cognitive impairment in aging processes and whether they affect synaptic structure and gut microbiota. Here, we studied the gender differences in cognitive ability, dendritic morphology, and gut microbiota of adult, middle-, and old-aged rats. METHODS The cognitive ability of rats using was assessed by the Y-maze SAB test, the light/dark discrimination test, and the MWM test. Dendritic morphology was investegated by Golgi staining. Microbiota composition, diversity and richness were analyzed by 16S rRNA gene sequencing. RESULTS The results showed that the cognitive ability of old-aged rats was decreased than adult and middle-aged rats in the spontaneous alternation behavior test, the light/dark discrimination test in Y-maze, and the MWM test; males have better cognitive ability than the females for middle-aged rats. The neuronal dendritic structures of CA1, CA3, and DG regions of the hippocampus were damaged to different degrees during aging, and the spine loss of females was more than that of males in CA1 and CA3 of middle-aged rats. In addition, the microbial diversity of gut microbiota was significantly decreased in old-aged male rats; the distribution and composition of microbiota communities were different between male and female rats at different ages. CONCLUSION These findings revealed that cognitive impairment in aged rats might result from dendritic damage in the hippocampus and gut microbiota dysbiosis, which provides direct evidence that gender differences in dendritic damage and gut microbiota dysbiosis might associate with cognitive impairment in naturally aged rats.
Collapse
Affiliation(s)
- Yuan-Yuan Han
- Department of Radiology, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
- School of Life Science, South China Normal University, Guangzhou, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kang Li
- School of Life Science, South China Normal University, Guangzhou, China
| | - Jing-Yu Hu
- School of Life Science, South China Normal University, Guangzhou, China
| | - Ji-Chao Wu
- School of Life Science, South China Normal University, Guangzhou, China
| | - Xiao Li
- School of Life Science, South China Normal University, Guangzhou, China
| | - De-Xiang Liu
- Department of Radiology, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chu-Hua Li
- Department of Radiology, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
- School of Life Science, South China Normal University, Guangzhou, China
| |
Collapse
|
2
|
Santoro B, Srinivas KV, Reyes I, Tian C, Masurkar AV. Cell type-specific impact of aging and Alzheimer disease on hippocampal CA1 perforant path input. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609952. [PMID: 39253428 PMCID: PMC11383042 DOI: 10.1101/2024.08.27.609952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
The perforant path (PP) carries direct inputs from entorhinal cortex to CA1 pyramidal neurons (PNs), with an impact dependent on PN position across transverse (CA1a-CA1c) and radial (superficial/deep) axes. It remains unclear how aging and Alzheimer disease (AD) affect PP input, despite its critical role in memory and early AD. Applying ex vivo recordings and two-photon microscopy in slices from mice up to 30 months old, we interrogated PP responses across PN subpopulations and compared them to Schaffer collateral and intrinsic excitability changes. We found that aging uniquely impacts PP excitatory responses, abolishing transverse and radial differences via a mechanism independent of presynaptic and membrane excitability change. This is amplified in aged 3xTg-AD mice, with further weakening of PP inputs to CA1a superficial PNs associated with distal dendritic spine loss. This demonstrates a unique feature of aging-related circuit dysfunction, with mechanistic implications related to memory impairment and synaptic vulnerability.
Collapse
|
3
|
Go J, Maeng SY, Chang DH, Park HY, Min KS, Kim JE, Choi YK, Noh JR, Ro H, Kim BC, Kim KS, Lee CH. Agathobaculum butyriciproducens improves ageing-associated cognitive impairment in mice. Life Sci 2024; 339:122413. [PMID: 38219919 DOI: 10.1016/j.lfs.2024.122413] [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: 10/06/2023] [Revised: 12/10/2023] [Accepted: 01/03/2024] [Indexed: 01/16/2024]
Abstract
AIMS The gut microbiota is increasingly recognised as a pivotal regulator of immune system homeostasis and brain health. Recent research has implicated the gut microbiota in age-related cognitive impairment and dementia. Agathobaculum butyriciproducens SR79 T (SR79), which was identified in the human gut, has been reported to be beneficial in addressing cognitive deficits and pathophysiologies in a mouse model of Alzheimer's disease. However, it remains unknown whether SR79 affects age-dependent cognitive impairment. MAIN METHOD To explore the effects of SR79 on cognitive function during ageing, we administered SR79 to aged mice. Ageing-associated behavioural alterations were examined using the open field test (OFT), tail suspension test (TST), novel object recognition test (NORT), Y-maze alternation test (Y-maze), and Morris water maze test (MWM). We investigated the mechanisms of action in the gut and brain using molecular and histological analyses. KEY FINDINGS Administration of SR79 improved age-related cognitive impairment without altering general locomotor activity or depressive behaviour in aged mice. Furthermore, SR79 increased mature dendritic spines in the pyramidal cells of layer III and phosphorylation of CaMKIIα in the cortex of aged mice. Age-related activation of astrocytes in the cortex of layers III-V of the aged brain was reduced following SR79 administration. Additionally, SR79 markedly increased IL-10 production and Foxp3 and Muc2 mRNA expression in the colons of aged mice. SIGNIFICANCE These findings suggest that treatment with SR79 may be a beneficial microbial-based approach for enhancing cognitive function during ageing.
Collapse
Affiliation(s)
- Jun Go
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - So-Young Maeng
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea; College of Biosciences & Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Dong-Ho Chang
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hye-Yeon Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Kyeong-Seon Min
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea; Department of Biomaterials Science, College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Ju-Eun Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Young-Keun Choi
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Jung-Ran Noh
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Hyunju Ro
- College of Biosciences & Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Byoung-Chan Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea; HealthBiome, Inc., Daejeon, Republic of Korea
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea; Department of Functional Genomics, KRIBB School, University of Science and Technology (UST), 217 Gajeong-ro, Youseong-gu, Daejeon, Republic of Korea.
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea; Department of Functional Genomics, KRIBB School, University of Science and Technology (UST), 217 Gajeong-ro, Youseong-gu, Daejeon, Republic of Korea.
| |
Collapse
|
4
|
Yarmohammadi-Samani P, Vatanparast J. Sex-specific dendritic morphology of hippocampal pyramidal neurons in the adolescent and young adult rats. Int J Dev Neurosci 2024; 84:47-63. [PMID: 37933732 DOI: 10.1002/jdn.10307] [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: 06/17/2023] [Revised: 09/02/2023] [Accepted: 10/16/2023] [Indexed: 11/08/2023] Open
Abstract
CA1 and CA3 pyramidal neurons are the major sources of hippocampal efferents. The structural features of these neurons are presumed to be involved in various normal/abnormal cognitive and emotional outcomes by influencing the pattern of synaptic inputs and neuronal signal processing. Although many studies have described hippocampal structure differences between males and females, these reports mainly focused on gross anatomical features in adult or aged models, and such distinctions on neuronal morphology and dendritic spine density during adolescence, a period of high vulnerability to neurodevelopmental disorders, have received much less attention. In this work, we analyzed dendritic architecture and density of spines in CA1 and CA3 neurons of male and female rats in early adolescence (postnatal day, PND 40) and compared them with those in late adolescence/young adulthood (PND 60). On PND 40, CA1 neurons of male rats showed more Sholl intersections and spine density in apical and basal dendrites compared to those in females. The Sholl intersections in basal dendrites of CA3 neurons were also more in males, whereas the number of apical dendrite intersections was not significantly different between sexes. In male rats, there was a notable decrease in the number of branch and terminal points in the basal dendrite of CA1 neurons of young adults when compared to their sex-matched adolescent rats. On the other hand, CA1 neurons in young adult females also showed more Sholl intersections in apical and basal dendrites compared to adolescent females. Meanwhile, the total cable length, the number of branches, and terminal points of apical dendrites in CA3 neurons also exhibited a significant reduction in young adult male rats compared to their sex-matched adolescents. In young adult rats, both apical and basal dendrites of CA3 neurons in males showed fewer intersections with Sholl circles, but there were no significant differences in dendritic spine density or count estimation between males and females. On the other hand, young adult female rats had more Sholl intersections and dendritic spine count on the basal dendrites of CA3 neurons compared to adolescent females. Although no significant sex- and age-dependent difference in neuronal density was detected in CA1 and CA3 subareas, CA3 pyramidal neurons of both male and female rats showed reduced soma area compared to adolescent rats. Our findings show that the sex differences in the dendritic structure of CA1 and CA3 neurons vary by age and also by the compartments of dendritic arbors. Such variations in the morphology of hippocampal pyramidal neurons may take part as a basis for normal cognitive and affective differences between the sexes, as well as distinct sensitivity to interfering factors and the prevalence of neuropsychological diseases.
Collapse
Affiliation(s)
| | - Jafar Vatanparast
- Department of Biology, School of Science, Shiraz University, Shiraz, Iran
| |
Collapse
|
5
|
Kim SR, Eom Y, Lee SH. Comprehensive analysis of sex differences in the function and ultrastructure of hippocampal presynaptic terminals. Neurochem Int 2023; 169:105570. [PMID: 37451344 DOI: 10.1016/j.neuint.2023.105570] [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: 01/25/2023] [Revised: 05/08/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Sex differences in the brain, encompassing variations in specific brain structures, size, cognitive function, and synaptic connections, have been identified across numerous species. While previous research has explored sex differences in postsynaptic structures, synaptic plasticity, and hippocampus-dependent functions, the hippocampal presynaptic terminals remain largely uninvestigated. The hippocampus is a critical structure responsible for multiple brain functions. This study examined presynaptic differences in cultured hippocampal neurons derived from male and female mice using a combination of biochemical assays, functional analyses measuring exocytosis and endocytosis of synaptic vesicle proteins, ultrastructural analyses via electron microscopy, and presynaptic Ca2+-specific optical probes. Our findings revealed that female neurons exhibited a higher number of synaptic vesicles at presynaptic terminals compared to male neurons. However, no significant differences were observed in presynaptic protein expression, presynaptic terminal ultrastructure, synaptic vesicle exocytosis and endocytosis, or presynaptic Ca2+ alterations between male and female neurons.
Collapse
Affiliation(s)
- Sung Rae Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea; Brain Research Core Facilities of Korea Brain Research Institute (KBRI), Daegu 41068, Republic of Korea.
| | - Yunkyung Eom
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| |
Collapse
|
6
|
Cai Y, Song W, Li J, Jing Y, Liang C, Zhang L, Zhang X, Zhang W, Liu B, An Y, Li J, Tang B, Pei S, Wu X, Liu Y, Zhuang CL, Ying Y, Dou X, Chen Y, Xiao FH, Li D, Yang R, Zhao Y, Wang Y, Wang L, Li Y, Ma S, Wang S, Song X, Ren J, Zhang L, Wang J, Zhang W, Xie Z, Qu J, Wang J, Xiao Y, Tian Y, Wang G, Hu P, Ye J, Sun Y, Mao Z, Kong QP, Liu Q, Zou W, Tian XL, Xiao ZX, Liu Y, Liu JP, Song M, Han JDJ, Liu GH. The landscape of aging. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2354-2454. [PMID: 36066811 PMCID: PMC9446657 DOI: 10.1007/s11427-022-2161-3] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
Aging is characterized by a progressive deterioration of physiological integrity, leading to impaired functional ability and ultimately increased susceptibility to death. It is a major risk factor for chronic human diseases, including cardiovascular disease, diabetes, neurological degeneration, and cancer. Therefore, the growing emphasis on "healthy aging" raises a series of important questions in life and social sciences. In recent years, there has been unprecedented progress in aging research, particularly the discovery that the rate of aging is at least partly controlled by evolutionarily conserved genetic pathways and biological processes. In an attempt to bring full-fledged understanding to both the aging process and age-associated diseases, we review the descriptive, conceptual, and interventive aspects of the landscape of aging composed of a number of layers at the cellular, tissue, organ, organ system, and organismal levels.
Collapse
Affiliation(s)
- Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Wei Song
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Jing
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chuqian Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Liyuan Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Xia Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenhui Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Beibei Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Yongpan An
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Baixue Tang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Siyu Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xueying Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuxuan Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Cheng-Le Zhuang
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Xuefeng Dou
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fu-Hui Xiao
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Dingfeng Li
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ya Zhao
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China
| | - Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Lihui Wang
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China
| | - Yujing Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- The Fifth People's Hospital of Chongqing, Chongqing, 400062, China.
| | - Xiaoyuan Song
- MOE Key Laboratory of Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Liang Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Jun Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China.
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Ye Tian
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Gelin Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China.
| | - Ping Hu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, 510005, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, 98195, USA.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Qiang Liu
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China.
| | - Zhi-Xiong Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China.
| | - Yong Liu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China.
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China.
- Department of Immunology and Pathology, Monash University Faculty of Medicine, Prahran, Victoria, 3181, Australia.
- Hudson Institute of Medical Research, and Monash University Department of Molecular and Translational Science, Clayton, Victoria, 3168, Australia.
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing, 100871, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
7
|
Sotoudeh N, Namavar MR. Optimisation of ketamine‐xylazine anaesthetic dose and its association with changes in the dendritic spine of CA1 hippocampus in the young and old male and female Wistar rats. Vet Med Sci 2022; 8:2545-2552. [DOI: 10.1002/vms3.936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Narges Sotoudeh
- Department of Anatomical Sciences School of Medicine, Shiraz University of Medical Sciences Shiraz Iran
- Histomorphometry and Stereology Research Center Shiraz University of Medical Sciences Shiraz Iran
| | - Mohammad Reza Namavar
- Department of Anatomical Sciences School of Medicine, Shiraz University of Medical Sciences Shiraz Iran
- Clinical Neurology Research Center Shiraz University of Medical Sciences Shiraz Iran
| |
Collapse
|
8
|
Juraska JM. Changes in sex differences in neuroanatomical structure and cognitive behavior across the life span. Learn Mem 2022; 29:340-348. [PMID: 36206396 PMCID: PMC9488018 DOI: 10.1101/lm.053499.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022]
Abstract
Sex differences occur in the structure and function of the rat cerebral cortex and hippocampus, which can change from the juvenile period through old age. Although the evidence is incomplete, it appears that in at least some portions of the cortex these differences develop due to the rise of ovarian hormones at puberty and are potentially not dependent on the perinatal rise in testosterone, which is essential for sexual differentiation of the hypothalamus and sexual behavior. During aging of female rats, the presence of continued ovarian hormone secretion after cessation of the estrous cycle also influences sex differences in neuroanatomical structure and cognitive behavior, resulting in nullification or reversal of sex differences seen in younger adults. Sex differences can be altered by experience in a stimulating environment during the juvenile/adolescent period, and sex differences in performance even can be affected by the parameters of a task. Thus, broad generalizations about differences such as "spatial ability" are to be avoided. It is clear that to understand how the brain produces behavior, sex and hormones have to be taken into account.
Collapse
Affiliation(s)
- Janice M Juraska
- Department of Psychology, Program in Neuroscience, University of Illinois at Urbana-Champaign, Champaign, Illinois 61820, USA
| |
Collapse
|
9
|
Huffels CFM, van Dijk RE, Karst H, Meye FJ, Hol EM, Middeldorp J. Systemic Injection of Aged Blood Plasma in Adult C57BL/6 Mice Induces Neurophysiological Impairments in the Hippocampal CA1. J Alzheimers Dis 2022; 89:283-297. [PMID: 35871343 DOI: 10.3233/jad-220337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Aging is characterized by systemic alterations and forms an important risk factor for Alzheimer's disease. Recently, it has been indicated that blood-borne factors present in the systemic milieu contribute to the aging process. Exposing young mice to aged blood plasma results in impaired neurogenesis and synaptic plasticity in the dentate gyrus, as well as impaired cognition. Vice versa, treating aged mice with young blood plasma rescues impairments associated with aging. OBJECTIVE Whether blood-borne factors are sufficient to drive impairments outside the dentate gyrus, how they impact neurophysiology, and how the functional outcome compares to impairments found in mouse models for AD is still unclear. METHODS Here, we treated adult mice with blood plasma from aged mice and assessed neurophysiological parameters in the hippocampal CA1. RESULTS Mice treated with aged blood plasma show significantly impaired levels of long-term potentiation (LTP), similar to those present in APP/PS1 mice. These impaired levels of LTP in plasma-treated mice are associated with alterations in basic properties of glutamatergic transmission and the enhanced activity of voltage-gated Ca2 + channels. CONCLUSION Together, the data presented in this study show that blood-borne factors are sufficient to drive neurophysiological impairments in the hippocampal CA1.
Collapse
Affiliation(s)
- Christiaan F M Huffels
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Roland E van Dijk
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Henk Karst
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Frank J Meye
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Jinte Middeldorp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.,Department of Neurobiology & Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| |
Collapse
|
10
|
Weerasinghe-Mudiyanselage PDE, Ang MJ, Kang S, Kim JS, Moon C. Structural Plasticity of the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:3349. [PMID: 35328770 PMCID: PMC8955928 DOI: 10.3390/ijms23063349] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/10/2022] Open
Abstract
Neuroplasticity is the capacity of neural networks in the brain to alter through development and rearrangement. It can be classified as structural and functional plasticity. The hippocampus is more susceptible to neuroplasticity as compared to other brain regions. Structural modifications in the hippocampus underpin several neurodegenerative diseases that exhibit cognitive and emotional dysregulation. This article reviews the findings of several preclinical and clinical studies about the role of structural plasticity in the hippocampus in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. In this study, literature was surveyed using Google Scholar, PubMed, Web of Science, and Scopus, to review the mechanisms that underlie the alterations in the structural plasticity of the hippocampus in neurodegenerative diseases. This review summarizes the role of structural plasticity in the hippocampus for the etiopathogenesis of neurodegenerative diseases and identifies the current focus and gaps in knowledge about hippocampal dysfunctions. Ultimately, this information will be useful to propel future mechanistic and therapeutic research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Poornima D. E. Weerasinghe-Mudiyanselage
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Mary Jasmin Ang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
- College of Veterinary Medicine, University of the Philippines Los Baños, Los Baños 4031, Philippines
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| |
Collapse
|
11
|
Khajehdehi M, Khalaj-Kondori M, Baradaran B. Molecular evidences on anti-inflammatory, anticancer, and memory-boosting effects of frankincense. Phytother Res 2022; 36:1194-1215. [PMID: 35142408 DOI: 10.1002/ptr.7399] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 12/16/2022]
Abstract
Chemical diversity of natural products with drug-like features has attracted much attention from medicine to develop more safe and effective drugs. Their anti-inflammatory, antitumor, analgesic, and other therapeutic properties are sometimes more successful than chemical drugs in controlling disease due to fewer drug resistance and side effects and being more tolerable in a long time. Frankincense, the oleo gum resin extracted from the Boswellia species, contains some of these chemicals. The anti-inflammatory effect of its main ingredient, boswellic acid, has been traditionally used to treat many diseases, mainly those target memory functions. In this review, we have accumulated research evidence from the beneficial effect of Frankincense consumption in memory improvement and the prevention of inflammation and cancer. Besides, we have discussed the molecular pathways mediating the therapeutic effects of this natural supplement.
Collapse
Affiliation(s)
- Mina Khajehdehi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Gramuntell Y, Klimczak P, Coviello S, Perez-Rando M, Nacher J. Effects of Aging on the Structure and Expression of NMDA Receptors of Somatostatin Expressing Neurons in the Mouse Hippocampus. Front Aging Neurosci 2022; 13:782737. [PMID: 35002680 PMCID: PMC8733323 DOI: 10.3389/fnagi.2021.782737] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
Changes in the physiology, neurochemistry and structure of neurons, particularly of their dendritic spines, are thought to be crucial players in age-related cognitive decline. One of the most studied brain structures affected by aging is the hippocampus, known to be involved in different essential cognitive processes. While the aging-associated quantitative changes in dendritic spines of hippocampal pyramidal cells have already been studied, the relationship between aging and the structural dynamics of hippocampal interneurons remains relatively unknown. Spines are not a frequent feature in cortical inhibitory neurons, but these postsynaptic structures are abundant in a subpopulation of somatostatin expressing interneurons, particularly in oriens-lacunosum moleculare (O-LM) cells in the hippocampal CA1. Previous studies from our laboratory have shown that the spines of these interneurons are highly plastic and influenced by NMDA receptor manipulation. Thus, in the present study, we have investigated the impact of aging on this interneuronal subpopulation. The analyses were performed in 3−, 9−, and 16-month-old GIN mice, a strain in which somatostatin positive interneurons express GFP. We studied the changes in the density of dendritic spines, en passant boutons, and the expression of NMDA receptors (GluN1 and GluN2B) using confocal microscopy and image analysis. We observed a significant decrease in dendritic spine density in 9-month-old animals when compared with 3-month-old animals. We also observed a decrease in the expression of the GluN2B subunit in O-LM cells, but not of that of GluN1, during aging. These results will constitute the basis for more advanced studies of the structure and connectivity of interneurons during aging and their contribution to cognitive decline.
Collapse
Affiliation(s)
- Yaiza Gramuntell
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Patrycja Klimczak
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain.,Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Simona Coviello
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Marta Perez-Rando
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain.,Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Juan Nacher
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain.,Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| |
Collapse
|
13
|
Verbruggen L, Ates G, Lara O, De Munck J, Villers A, De Pauw L, Ottestad-Hansen S, Kobayashi S, Beckers P, Janssen P, Sato H, Zhou Y, Hermans E, Njemini R, Arckens L, Danbolt NC, De Bundel D, Aerts JL, Barbé K, Guillaume B, Ris L, Bentea E, Massie A. Lifespan extension with preservation of hippocampal function in aged system x c--deficient male mice. Mol Psychiatry 2022; 27:2355-2368. [PMID: 35181756 PMCID: PMC9126817 DOI: 10.1038/s41380-022-01470-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/11/2022] [Accepted: 02/02/2022] [Indexed: 11/25/2022]
Abstract
The cystine/glutamate antiporter system xc- has been identified as the major source of extracellular glutamate in several brain regions as well as a modulator of neuroinflammation, and genetic deletion of its specific subunit xCT (xCT-/-) is protective in mouse models for age-related neurological disorders. However, the previously observed oxidative shift in the plasma cystine/cysteine ratio of adult xCT-/- mice led to the hypothesis that system xc- deletion would negatively affect life- and healthspan. Still, till now the role of system xc- in physiological aging remains unexplored. We therefore studied the effect of xCT deletion on the aging process of mice, with a particular focus on the immune system, hippocampal function, and cognitive aging. We observed that male xCT-/- mice have an extended lifespan, despite an even more increased plasma cystine/cysteine ratio in aged compared to adult mice. This oxidative shift does not negatively impact the general health status of the mice. On the contrary, the age-related priming of the innate immune system, that manifested as increased LPS-induced cytokine levels and hypothermia in xCT+/+ mice, was attenuated in xCT-/- mice. While this was associated with only a very moderate shift towards a more anti-inflammatory state of the aged hippocampus, we observed changes in the hippocampal metabolome that were associated with a preserved hippocampal function and the retention of hippocampus-dependent memory in male aged xCT-/- mice. Targeting system xc- is thus not only a promising strategy to prevent cognitive decline, but also to promote healthy aging.
Collapse
Affiliation(s)
- Lise Verbruggen
- grid.8767.e0000 0001 2290 8069Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Gamze Ates
- grid.8767.e0000 0001 2290 8069Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Olaya Lara
- grid.8767.e0000 0001 2290 8069Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Jolien De Munck
- grid.8767.e0000 0001 2290 8069Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Agnès Villers
- grid.8364.90000 0001 2184 581XDepartment of Neurosciences, Université de Mons (UMONS), Mons, Belgium
| | - Laura De Pauw
- grid.8767.e0000 0001 2290 8069Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Sigrid Ottestad-Hansen
- grid.5510.10000 0004 1936 8921Neurotransporter Group, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Sho Kobayashi
- grid.268394.20000 0001 0674 7277Department of Food, Life and Environmental Science, Faculty of Agriculture, Yamagata University, Yamagata, Japan
| | - Pauline Beckers
- grid.7942.80000 0001 2294 713XInstitute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Pauline Janssen
- grid.8767.e0000 0001 2290 8069Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Hideyo Sato
- grid.260975.f0000 0001 0671 5144Department of Medical Technology, Niigata University, Niigata, Japan
| | - Yun Zhou
- grid.5510.10000 0004 1936 8921Neurotransporter Group, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Emmanuel Hermans
- grid.7942.80000 0001 2294 713XInstitute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Rose Njemini
- grid.8767.e0000 0001 2290 8069Frailty in Ageing research Department, VUB, Brussels, Belgium
| | - Lutgarde Arckens
- grid.5596.f0000 0001 0668 7884Laboratory of Neuroplasticity and Neuroproteomics, and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
| | - Niels C. Danbolt
- grid.5510.10000 0004 1936 8921Neurotransporter Group, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Dimitri De Bundel
- grid.8767.e0000 0001 2290 8069Pharmaceutical Chemistry, Drug Analysis and Drug Information, C4N, VUB, Brussels, Belgium
| | - Joeri L. Aerts
- grid.8767.e0000 0001 2290 8069Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Kurt Barbé
- grid.8767.e0000 0001 2290 8069The Biostatistics and Medical Informatics Department, VUB, Brussels, Belgium
| | | | - Laurence Ris
- grid.8364.90000 0001 2184 581XDepartment of Neurosciences, Université de Mons (UMONS), Mons, Belgium
| | - Eduard Bentea
- grid.8767.e0000 0001 2290 8069Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Ann Massie
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| |
Collapse
|
14
|
Kim W, Chung C. Brain-wide cellular mapping of acute stress-induced activation in male and female mice. FASEB J 2021; 35:e22041. [PMID: 34780680 DOI: 10.1096/fj.202101287r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/14/2021] [Accepted: 10/29/2021] [Indexed: 11/11/2022]
Abstract
Mood disorders are more prevalent and often reported to be more severe in women; however, little is known about the underlying mechanisms of this sexual prevalence. To gain insight into the functional differences in female brains in response to stress, we systemically compared brain activation in male and female C57BL/6N mice after acute stress exposure. We measured c-Fos expression levels in 18 brain areas related to stress responses after a 3-h long restraint stress and found that activation was sexually dimorphic in several brain areas, including the nucleus accumbens, ventral tegmental area, nucleus reuniens, and medial part of the lateral habenula. Moreover, stress-activated a substantial number of cells in the medial prefrontal cortex, amygdala, and lateral part of the lateral habenula; however, the levels of activation were comparable in males and females, suggesting that the core stress responding machineries are largely shared. Pearson correlation analysis revealed several interesting connections between the analyzed areas that are implicated in stress responses and depression. Overall, stress strengthened intra-circuitries in the hippocampus, amygdala, and prefrontal cortex in female mice, whereas more longer-range connections were highlighted in stressed male mice. Our study provides a highly valuable neuroanatomical framework for investigating the circuit mechanism underlying the higher vulnerability to depression in women.
Collapse
Affiliation(s)
- Woonhee Kim
- Department of Biological Sciences, Konkuk University, Seoul, South Korea
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul, South Korea
| |
Collapse
|
15
|
Alshammari GM, Al-Qahtani WH, Alshuniaber MA, Yagoub AEA, Al-Khalifah AS, Al-Harbi LN, Alhussain MH, AlSedairy SA, Yahya MA. Quercetin improves the impairment in memory function and attenuates hippocampal damage in cadmium chloride-intoxicated male rats by suppressing acetylcholinesterase and concomitant activation of SIRT1 signaling. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
16
|
Cuestas Torres DM, Cardenas FP. Synaptic plasticity in Alzheimer's disease and healthy aging. Rev Neurosci 2021; 31:245-268. [PMID: 32250284 DOI: 10.1515/revneuro-2019-0058] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
The strength and efficiency of synaptic connections are affected by the environment or the experience of the individual. This property, called synaptic plasticity, is directly related to memory and learning processes and has been modeled at the cellular level. These types of cellular memory and learning models include specific stimulation protocols that generate a long-term strengthening of the synapses, called long-term potentiation, or a weakening of the said long-term synapses, called long-term depression. Although, for decades, researchers have believed that the main cause of the cognitive deficit that characterizes Alzheimer's disease (AD) and aging was the loss of neurons, the hypothesis of an imbalance in the cellular and molecular mechanisms of synaptic plasticity underlying this deficit is currently widely accepted. An understanding of the molecular and cellular changes underlying the process of synaptic plasticity during the development of AD and aging will direct future studies to specific targets, resulting in the development of much more efficient and specific therapeutic strategies. In this review, we classify, discuss, and describe the main findings related to changes in the neurophysiological mechanisms of synaptic plasticity in excitatory synapses underlying AD and aging. In addition, we suggest possible mechanisms in which aging can become a high-risk factor for the development of AD and how its development could be prevented or slowed.
Collapse
Affiliation(s)
- Diana Marcela Cuestas Torres
- Departamento de Psicología and Departamento de Biología, Laboratorio de Neurociencia y Comportamiento, Universidad de los Andes, Cra 1 N° 18A-12, CP 111711, Bogotá, Colombia
| | - Fernando P Cardenas
- Departamento de Psicología, Laboratorio de Neurociencia y Comportamiento, Universidad de los Andes, Cra 1 N° 18A-12, CP 111711, Bogotá, Colombia
| |
Collapse
|
17
|
Francavilla R, Guet-McCreight A, Amalyan S, Hui CW, Topolnik D, Michaud F, Marino B, Tremblay MÈ, Skinner FK, Topolnik L. Alterations in Intrinsic and Synaptic Properties of Hippocampal CA1 VIP Interneurons During Aging. Front Cell Neurosci 2020; 14:554405. [PMID: 33173468 PMCID: PMC7591401 DOI: 10.3389/fncel.2020.554405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/10/2020] [Indexed: 12/21/2022] Open
Abstract
Learning and memory deficits are hallmarks of the aging brain, with cortical neuronal circuits representing the main target in cognitive deterioration. While GABAergic inhibitory and disinhibitory circuits are critical in supporting cognitive processes, their roles in age-related cognitive decline remain largely unknown. Here, we examined the morphological and physiological properties of the hippocampal CA1 vasoactive intestinal peptide/calretinin-expressing (VIP+/CR+) type 3 interneuron-specific (I-S3) cells across mouse lifespan. Our data showed that while the number and morphological features of I-S3 cells remained unchanged, their firing and synaptic properties were significantly altered in old animals. In particular, the action potential duration and the level of steady-state depolarization were significantly increased in old animals in parallel with a significant decrease in the maximal firing frequency. Reducing the fast-delayed rectifier potassium or transient sodium conductances in I-S3 cell computational models could reproduce the age-related changes in I-S3 cell firing properties. However, experimental data revealed no difference in the activation properties of the Kv3.1 and A-type potassium currents, indicating that transient sodium together with other ion conductances may be responsible for the observed phenomena. Furthermore, I-S3 cells in aged mice received a stronger inhibitory drive due to concomitant increase in the amplitude and frequency of spontaneous inhibitory currents. These age-associated changes in the I-S3 cell properties occurred in parallel with an increased inhibition of their target interneurons and were associated with spatial memory deficits and increased anxiety. Taken together, these data indicate that VIP+/CR+ interneurons responsible for local circuit disinhibition survive during aging but exhibit significantly altered physiological properties, which may result in the increased inhibition of hippocampal interneurons and distorted mnemonic functions.
Collapse
Affiliation(s)
- Ruggiero Francavilla
- Department of Biochemistry, Microbiology and Bioinformatics, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada
- Neuroscience Axis, Centre Hospitalier Universitaire (CHU) de Québec Research Center – Université Laval, Québec, QC, Canada
| | - Alexandre Guet-McCreight
- Krembil Research Institute, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Sona Amalyan
- Department of Biochemistry, Microbiology and Bioinformatics, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada
- Neuroscience Axis, Centre Hospitalier Universitaire (CHU) de Québec Research Center – Université Laval, Québec, QC, Canada
| | - Chin Wai Hui
- Neuroscience Axis, Centre Hospitalier Universitaire (CHU) de Québec Research Center – Université Laval, Québec, QC, Canada
| | - Dimitry Topolnik
- Neuroscience Axis, Centre Hospitalier Universitaire (CHU) de Québec Research Center – Université Laval, Québec, QC, Canada
| | - Félix Michaud
- Department of Biochemistry, Microbiology and Bioinformatics, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada
- Neuroscience Axis, Centre Hospitalier Universitaire (CHU) de Québec Research Center – Université Laval, Québec, QC, Canada
| | - Beatrice Marino
- Department of Biochemistry, Microbiology and Bioinformatics, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada
- Neuroscience Axis, Centre Hospitalier Universitaire (CHU) de Québec Research Center – Université Laval, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Neuroscience Axis, Centre Hospitalier Universitaire (CHU) de Québec Research Center – Université Laval, Québec, QC, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Frances K. Skinner
- Krembil Research Institute, University Health Network, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Departments of Medicine (Neurology) and Physiology, University of Toronto, Toronto, ON, Canada
| | - Lisa Topolnik
- Department of Biochemistry, Microbiology and Bioinformatics, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada
- Neuroscience Axis, Centre Hospitalier Universitaire (CHU) de Québec Research Center – Université Laval, Québec, QC, Canada
| |
Collapse
|
18
|
Flores G, Flores-Gómez GD, Díaz A, Penagos-Corzo JC, Iannitti T, Morales-Medina JC. Natural products present neurotrophic properties in neurons of the limbic system in aging rodents. Synapse 2020; 75:e22185. [PMID: 32779216 DOI: 10.1002/syn.22185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/21/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022]
Abstract
Aging is a complex process that can lead to neurodegeneration and, consequently, several pathologies, including dementia. Physiological aging leads to changes in several body organs, including those of the central nervous system (CNS). Morphological changes in the CNS and particularly the brain result in motor and cognitive deficits affecting learning and memory and the circadian cycle. Characterizing neural modifications is critical to designing new therapies to target aging and associated pathologies. In this review, we compared aging to the changes occurring within the brain and particularly the limbic system. Then, we focused on key natural compounds, apamin, cerebrolysin, Curcuma longa, resveratrol, and N-PEP-12, which have shown neurotrophic effects particularly in the limbic system. Finally, we drew our conclusions delineating future perspectives for the development of novel natural therapeutics to ameliorate aging-related processes.
Collapse
Affiliation(s)
- Gonzalo Flores
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Gabriel Daniel Flores-Gómez
- Departamento de Ciencias de la Salud, Licenciatura en Medicina, Universidad de las Américas Puebla, Puebla, México
| | - Alfonso Díaz
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | | | - Tommaso Iannitti
- Charles River Discovery Research Services UK Limited part of the Charles River Group, Bristol, UK
| | - Julio César Morales-Medina
- Centro de Investigación en Reproducción Animal, CINVESTAV- Universidad Autónoma de Tlaxcala, Tlaxcala, México
| |
Collapse
|
19
|
Hillerer KM, Slattery DA, Pletzer B. Neurobiological mechanisms underlying sex-related differences in stress-related disorders: Effects of neuroactive steroids on the hippocampus. Front Neuroendocrinol 2019; 55:100796. [PMID: 31580837 PMCID: PMC7115954 DOI: 10.1016/j.yfrne.2019.100796] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
Men and women differ in their vulnerability to a variety of stress-related illnesses, but the underlying neurobiological mechanisms are not well understood. This is likely due to a comparative dearth of neurobiological studies that assess male and female rodents at the same time, while human neuroimaging studies often don't model sex as a variable of interest. These sex differences are often attributed to the actions of sex hormones, i.e. estrogens, progestogens and androgens. In this review, we summarize the results on sex hormone actions in the hippocampus and seek to bridge the gap between animal models and findings in humans. However, while effects of sex hormones on the hippocampus are largely consistent in animals and humans, methodological differences challenge the comparability of animal and human studies on stress effects. We summarise our current understanding of the neurobiological mechanisms that underlie sex-related differences in behavior and discuss implications for stress-related illnesses.
Collapse
Affiliation(s)
- Katharina M Hillerer
- Department of Obstetrics and Gynaecology, Salzburger Landeskrankenhaus (SALK), Paracelsus Medical University (PMU), Clinical Research Center Salzburg (CRCS), Salzburg, Austria.
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Belinda Pletzer
- Department of Psychology, University of Salzburg, Salzburg, Austria; Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| |
Collapse
|
20
|
Das SC, Chen D, Callor WB, Christensen E, Coon H, Williams ME. DiI-mediated analysis of presynaptic and postsynaptic structures in human postmortem brain tissue. J Comp Neurol 2019; 527:3087-3098. [PMID: 31152449 DOI: 10.1002/cne.24722] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/20/2022]
Abstract
Most cognitive and psychiatric disorders are thought to be disorders of the synapse, yet the precise synapse defects remain unknown. Because synapses are highly specialized anatomical structures, defects in synapse formation and function can often be observed as changes in microscale neuroanatomy. Unfortunately, few methods are available for accurate analysis of synaptic structures in human postmortem tissues. Here, we present a methodological pipeline for assessing presynaptic and postsynaptic structures in human postmortem tissue that is accurate, rapid, and relatively inexpensive. Our method uses small tissue blocks from postmortem human brains, immersion fixation, lipophilic dye (DiI) labeling, and confocal microscopy. As proof of principle, we analyzed presynaptic and postsynaptic structures from hippocampi of 13 individuals aged 4 months to 71 years. Our results indicate that postsynaptic CA1 dendritic spine shape and density do not change in adults, while presynaptic DG mossy fiber boutons undergo significant structural rearrangements with normal aging. This suggests that mossy fiber synapses, which play a major role in learning and memory, may remain dynamic throughout life. Importantly, we find that human CA1 spine densities observed using this method on tissue that is up to 28 h postmortem is comparable to prior studies using tissue with much shorter postmortem intervals. Thus, the ease of our protocol and suitability on tissue with longer postmortem intervals should facilitate higher-powered studies of human presynaptic and postsynaptic structures in healthy and diseased states.
Collapse
Affiliation(s)
- Sujan C Das
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, Utah
| | - Danli Chen
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, Utah
| | | | - Eric Christensen
- Utah State Office of Medical Examiner, Utah Department of Health, Salt Lake City, Utah
| | - Hilary Coon
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, Utah
| | - Megan E Williams
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
21
|
Chen FJ, Liu B, Wu Q, Liu J, Xu YY, Zhou SY, Shi JS. Icariin Delays Brain Aging in Senescence-Accelerated Mouse Prone 8 (SAMP8) Model via Inhibiting Autophagy. J Pharmacol Exp Ther 2019; 369:121-128. [PMID: 30837279 DOI: 10.1124/jpet.118.253310] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/06/2019] [Indexed: 11/22/2022] Open
Abstract
Icariin (ICA), a major flavonoid extracted from the Chinese tonic herb Epimedium, exerts beneficial effects in a variety of age-dependent diseases, such as Alzheimer's disease (AD). However, the antiaging mechanisms remain unclear. The senescence-accelerated mouse-prone 8 (SAMP8) model has been used to study age-related neurodegenerative changes associated with aging and the pathogenesis of AD. Hence, the current study was designed to examine the effect of ICA on age-related cognitive decline in SAMP8 mice and explore the role of autophagy in the ICA-mediated neuroprotection. SAMP8 mice were administered with ICA starting at 5 months of age, and the treatment lasted for 3 consecutive months. Morris water maze was used to evaluate cognitive function. The senescence-associated β-galactosidase staining was used to determine the number of senescence cells. The neuronal morphologic changes were examined via Nissl staining. The hippocampal neuronal ultrastructure was examined by transmission electron microscopy. The expression of autophagy protein was examined by Western blot. ICA-treated SAMP8 mice exhibited a robust improvement in spatial learning and memory function. Meanwhile, ICA reduced the number of senescence cells in the brains of SAMP8 mice, inhibited neuronal loss, and reversed neuronal structural changes in the hippocampi of SAMP8 mice. Moreover, ICA treatment also decreased the formation of autophagosomes in the hippocampus of SAMP8 mice, and reduced the expression of autophagy-related proteins LC3-II and p62. These results demonstrate that ICA possesses the ability to delay brain aging in SAMP8 mice, and the mechanisms are possibly mediated through the regulation of autophagy.
Collapse
Affiliation(s)
- Fa-Ju Chen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, People's Republic of China
| | - Bo Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, People's Republic of China
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, People's Republic of China
| | - Jie Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, People's Republic of China
| | - Yun-Yan Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, People's Republic of China
| | - Shao-Yu Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, People's Republic of China
| | - Jing-Shan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, People's Republic of China
| |
Collapse
|
22
|
Structural and molecular correlates of cognitive aging in the rat. Sci Rep 2019; 9:2005. [PMID: 30765864 PMCID: PMC6376121 DOI: 10.1038/s41598-019-39645-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 06/01/2018] [Indexed: 12/12/2022] Open
Abstract
Aging is associated with cognitive decline. Herein, we studied a large cohort of old age and young adult male rats and confirmed that, as a group, old rats display poorer spatial learning and behavioral flexibility than younger adults. Surprisingly, when animals were clustered as good and bad performers, our data revealed that while in younger animals better cognitive performance was associated with longer dendritic trees and increased levels of synaptic markers in the hippocampus and prefrontal cortex, the opposite was found in the older group, in which better performance was associated with shorter dendrites and lower levels of synaptic markers. Additionally, in old, but not young individuals, worse performance correlated with increased levels of BDNF and the autophagy substrate p62, but decreased levels of the autophagy complex protein LC3. In summary, while for younger individuals “bigger is better”, “smaller is better” is a more appropriate aphorism for older subjects.
Collapse
|
23
|
Farooqi NAI, Scotti M, Yu A, Lew J, Monnier P, Botteron KN, Campbell BC, Booij L, Herba CM, Séguin JR, Castellanos-Ryan N, McCracken JT, Nguyen TV. Sex-specific contribution of DHEA-cortisol ratio to prefrontal-hippocampal structural development, cognitive abilities and personality traits. J Neuroendocrinol 2019; 31:e12682. [PMID: 30597689 PMCID: PMC6394408 DOI: 10.1111/jne.12682] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 11/29/2018] [Accepted: 12/28/2018] [Indexed: 01/06/2023]
Abstract
Although dehydroepiandrosterone (DHEA) may exert neuroprotective effects in the developing brain, prolonged or excessive elevations in cortisol may exert neurotoxic effects. The ratio between DHEA and cortisol (DC ratio) has been linked to internalising and externalising disorders, as well as cognitive performance, supporting the clinical relevance of this hormonal ratio during development. However, the brain mechanisms by which these effects may be mediated have not yet been identified. Furthermore, although there is evidence that the effects of cortisol in the central nervous system may be sexually dimorphic in humans, the opposite is true for DHEA, with human studies showing no sex-specific associations in cortical thickness, cortico-amygdalar or cortico-hippocampal structural covariance. Therefore, it remains unclear whether sex moderates the developmental associations between DC ratio, brain structure, cognition and behaviour. In the present study, we examined the associations between DC ratio, structural covariance of the hippocampus with whole-brain cortical thickness, and measures of personality, behaviour and cognition in a longitudinal sample of typically developing children, adolescents and young adults aged 6-22 years (N = 225 participants [F = 128]; 355 scans [F = 208]), using mixed effects models that accounted for both within- and between-subject variances. We found sex-specific interactions between DC ratio and anterior cingulate cortex-hippocampal structural covariance, with higher DC ratios being associated with a more negative covariance between these structures in girls, and a more positive covariance in boys. Furthermore, the negative prefrontal-hippocampal structural covariance found in girls was associated with higher verbal memory and mathematical ability, whereas the positive covariance found in boys was associated with lower cooperativeness and reward dependence personality traits. These findings support the notion that the ratio between DHEA and cortisol levels may contribute, at least in part, to the development of sex differences in cognitive abilities, as well as risk for internalising/externalising disorders, via an alteration in prefrontal-hippocampal structure during the transition from childhood to adulthood.
Collapse
Affiliation(s)
- Nasr A. I. Farooqi
- Department of Psychiatry, McGill University, Montreal, QC,
Canada, H3A1A1
| | - Martina Scotti
- Department of Psychiatry, McGill University, Montreal, QC,
Canada, H3A1A1
| | - Ally Yu
- Department of Psychology, McGill University, Montreal, QC,
Canada, H4A 3J1
| | - Jimin Lew
- Department of Psychology, McGill University, Montreal, QC,
Canada, H4A 3J1
| | - Patricia Monnier
- Department of Obstetrics-Gynecology, McGill University
Health Center, Montreal, QC, Canada, H4A 3J1
- Research Institute of the McGill University Health Center,
Montreal, QC, Canada, H4A 3J1
| | - Kelly N Botteron
- Department of Psychiatry, Washington University School of
Medicine, St. Louis, MO, USA, 63110
- Brain Development Cooperative Group
| | - Benjamin C. Campbell
- Department of Anthropology, University of
Wisconsin-Milwaukee, Milwaukee, WI, USA, 53211
| | - Linda Booij
- Department of Psychiatry, McGill University, Montreal, QC,
Canada, H3A1A1
- Department of Psychology, Concordia University, Montreal,
QC, Canada, H4B 1R6
- CHU Sainte Justine Hospital Research Centre, University of
Montreal, Montreal, QC, Canada, H3T1C5
| | - Catherine M. Herba
- CHU Sainte Justine Hospital Research Centre, University of
Montreal, Montreal, QC, Canada, H3T1C5
- Department of Psychology, Université du
Québec à Montréal, Montreal, QC, Canada
| | - Jean R. Séguin
- CHU Sainte Justine Hospital Research Centre, University of
Montreal, Montreal, QC, Canada, H3T1C5
- Department of Psychiatry and Addiction, University of
Montreal, Montreal, QC, Canada, H3T1C5
| | - Natalie Castellanos-Ryan
- CHU Sainte Justine Hospital Research Centre, University of
Montreal, Montreal, QC, Canada, H3T1C5
- School of Psychoeducation, University of Montreal,
Montreal QC, Canada, H2V 2S9
| | - James T McCracken
- Brain Development Cooperative Group
- Department of Child and Adolescent Psychiatry, University
of California in Los Angeles, Los Angeles, CA, USA, 90024
| | - Tuong-Vi Nguyen
- Department of Psychiatry, McGill University, Montreal, QC,
Canada, H3A1A1
- Department of Obstetrics-Gynecology, McGill University
Health Center, Montreal, QC, Canada, H4A 3J1
- Research Institute of the McGill University Health Center,
Montreal, QC, Canada, H4A 3J1
| |
Collapse
|
24
|
Metzler-Baddeley C, Mole JP, Sims R, Fasano F, Evans J, Jones DK, Aggleton JP, Baddeley RJ. Fornix white matter glia damage causes hippocampal gray matter damage during age-dependent limbic decline. Sci Rep 2019; 9:1060. [PMID: 30705365 PMCID: PMC6355929 DOI: 10.1038/s41598-018-37658-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
Aging leads to gray and white matter decline but their causation remains unclear. We explored two classes of models of age and dementia risk related brain changes. The first class of models emphasises the importance of gray matter: age and risk-related processes cause neurodegeneration and this causes damage in associated white matter tracts. The second class of models reverses the direction of causation: aging and risk factors cause white matter damage and this leads to gray matter damage. We compared these models with linear mediation analysis and quantitative MRI indices (from diffusion, quantitative magnetization transfer and relaxometry imaging) of tissue properties in two limbic structures implicated in age-related memory decline: the hippocampus and the fornix in 166 asymptomatic individuals (aged 38–71 years). Aging was associated with apparent glia but not neurite density damage in the fornix and the hippocampus. Mediation analysis supported white matter damage causing gray matter decline; controlling for fornix glia damage, the correlations between age and hippocampal damage disappear, but not vice versa. Fornix and hippocampal differences were both associated with reductions in episodic memory performance. These results suggest that fornix white matter glia damage may cause hippocampal gray matter damage during age-dependent limbic decline.
Collapse
Affiliation(s)
- Claudia Metzler-Baddeley
- Cardiff University Brain Research Imaging Centre (CUBRIC), Maindy Road, Cathays, Cardiff, CF24 4HQ, UK.
| | - Jilu P Mole
- Cardiff University Brain Research Imaging Centre (CUBRIC), Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Rebecca Sims
- Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Fabrizio Fasano
- Cardiff University Brain Research Imaging Centre (CUBRIC), Maindy Road, Cathays, Cardiff, CF24 4HQ, UK.,Siemens Healthcare, Head Office, Sir William Siemens Square, Surrey, GU16 8QD, UK
| | - John Evans
- Cardiff University Brain Research Imaging Centre (CUBRIC), Maindy Road, Cathays, Cardiff, CF24 4HQ, UK
| | - Derek K Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), Maindy Road, Cathays, Cardiff, CF24 4HQ, UK.,School of Psychology, Faculty of Health Sciences, Australian Catholic University, Melbourne, Victoria, 3065, Australia
| | - John P Aggleton
- School of Psychology, Cardiff University, Tower Building, 70 Park Place, Cardiff, CF10 3AT, UK
| | - Roland J Baddeley
- Experimental Psychology, University of Bristol, 12a Priory Road, Bristol, BS8 1TU, UK
| |
Collapse
|
25
|
Keith RE, Azcarate JM, Keith MJ, Hung CW, Badakhsh MF, Dumas TC. Direct Intracellular Signaling by the Carboxy terminus of NMDA Receptor GluN2 Subunits Regulates Dendritic Morphology in Hippocampal CA1 Pyramidal Neurons. Neuroscience 2019; 396:138-153. [PMID: 30471357 PMCID: PMC6311441 DOI: 10.1016/j.neuroscience.2018.11.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/16/2018] [Accepted: 11/14/2018] [Indexed: 12/22/2022]
Abstract
N-methyl-d-aspartate receptors (NMDARs) are glutamatergic receptors that take part in excitatory synaptic transmission and drive functional and structural neuronal plasticity, including activity-dependent changes in dendritic morphology. Forebrain NMDARs contribute to neuronal plasticity in at least two ways: through calcium-mediated processes or via direct intracellular postsynaptic signaling. Both properties are regulated by the GluN2 subunits. However, the separate contributions of these properties to the regulation of dendritic morphology are unknown. We created transgenic mice that express chimeric GluN2 subunits and examined the impact on pyramidal cell dendritic morphology in hippocampal region CA1. Golgi-Cox impregnation and transgenic expression of green fluorescent protein were employed to visualize dendritic arbors. In adult mice with a predominantly native GluN2A background, overexpression of the GluN2B carboxy terminus increased the total path of the dendritic arbor without affecting branch number or tortuosity. Overexpressing the amino terminus and transmembrane domains of GluN2B had little effect. It may be inferred from these results that NMDAR-dependent intracellular signaling regulates dendritic morphology of hippocampal pyramidal cells more so than calcium conductance dynamics. The findings add to the understanding of NMDAR-mediated signaling in hippocampal neurons and support re-investigation of the molecular underpinnings of NMDAR involvement in postnatal dendrite maturation.
Collapse
Affiliation(s)
- Rachel E Keith
- Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA 22030, United States; Krasnow Institute of Advanced Study, George Mason University, Fairfax, VA 22030, United States
| | - Jessica M Azcarate
- Krasnow Institute of Advanced Study, George Mason University, Fairfax, VA 22030, United States
| | - Matthew J Keith
- Krasnow Institute of Advanced Study, George Mason University, Fairfax, VA 22030, United States
| | - Carey W Hung
- Krasnow Institute of Advanced Study, George Mason University, Fairfax, VA 22030, United States
| | - Maryam F Badakhsh
- Krasnow Institute of Advanced Study, George Mason University, Fairfax, VA 22030, United States
| | - Theodore C Dumas
- Psychology Department, George Mason University, Fairfax, VA 22030, United States; Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA 22030, United States; Krasnow Institute of Advanced Study, George Mason University, Fairfax, VA 22030, United States.
| |
Collapse
|
26
|
Loss of thin spines and small synapses contributes to defective hippocampal function in aged mice. Neurobiol Aging 2018; 71:91-104. [PMID: 30118927 DOI: 10.1016/j.neurobiolaging.2018.07.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/07/2018] [Accepted: 07/14/2018] [Indexed: 12/23/2022]
Abstract
Aging is a normal physiological process associated with impairments in cognitive function, including learning and memory. Here, the underlying synaptic mechanisms by which aging leads to the decline of spatial learning and memory function were investigated in 25-month-old aged mice versus 2-month-old young mice. Deficits of spatial learning and memory, as well as selective loss of thin spines, but not mushroom-type spines on apical dendrites of CA1 pyramidal cells were found in aged mice. Specifically, loss of thin spines in aged mice with memory deficits was primarily found on dendritic segments located in the Schaffer pathway, and the density of thin spines significantly correlated with spatial memory performance. The loss of thin spines was evidenced by a decrease in small synapses that express diminutive amounts of postsynaptic density protein-95 and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit GluR1. Furthermore, mushroom-type spines and GluR1-expressed large synapses were not affected in aged mice with impaired memory. Taken together, these data suggest that the selective loss of those highly plastic thin spines with sparse postsynaptic density protein-95 and GluR1 receptors may significantly contribute to cognitive deficits in aged individuals.
Collapse
|
27
|
Santarelli AJ, Khan AM, Poulos AM. Contextual fear retrieval-induced Fos expression across early development in the rat: An analysis using established nervous system nomenclature ontology. Neurobiol Learn Mem 2018; 155:42-49. [PMID: 29807127 DOI: 10.1016/j.nlm.2018.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/07/2018] [Accepted: 05/19/2018] [Indexed: 02/07/2023]
Abstract
The neural circuits underlying the acquisition, retention and retrieval of contextual fear conditioning have been well characterized in the adult animal. A growing body of work in younger rodents indicates that context-mediated fear expression may vary across development. However, it remains unclear how this expression may be defined across the full range of key developmental ages. Nor is it fully clear whether the structure of the adult context fear network generalizes to earlier ages. In this study, we compared context fear retrieval-induced behavior and neuroanatomically constrained immediate early-gene expression across infant (P19), early and late juvenile (P24 and P35), and adult (P90) male Long-Evans rats. We focused our analysis on neuroanatomically defined subregions and nuclei of the basolateral complex of the amygdala (BLA complex), dorsal and ventral portions of the hippocampus and the subregions of the medial prefrontal cortex as defined by the nomenclature of the Swanson (2004) adult rat brain atlas. Relative to controls and across all ages tested, there were greater numbers of Fos immunoreactive (Fos-ir) neurons in the posterior part of the basolateral amygdalar nuclei (BLAp) following context fear retrieval that correlated statistically with the expression of freezing. However, Fos-ir within regions having known connections with the BLA complex was differentially constrained by developmental age: early juvenile, but not adult rats exhibited an increase of context fear-dependent Fos-ir neurons in prelimbic and infralimbic areas, while adult, but not juvenile rats displayed increases in Fos-ir neurons within the ventral CA1 hippocampus. These results suggest that juvenile and adult rodents may recruit developmentally unique pathways in the acquisition and retrieval of contextual fear. This study extends prior work by providing a broader set of developmental ages and a rigorously defined neuroanatomical ontology within which the contextual fear network can be studied further.
Collapse
Affiliation(s)
- Anthony J Santarelli
- Department of Psychology, Center for Neuroscience, State University of New York, University at Albany, Albany, NY 12222, USA
| | - Arshad M Khan
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Andrew M Poulos
- Department of Psychology, Center for Neuroscience, State University of New York, University at Albany, Albany, NY 12222, USA.
| |
Collapse
|
28
|
Bettio LEB, Rajendran L, Gil-Mohapel J. The effects of aging in the hippocampus and cognitive decline. Neurosci Biobehav Rev 2017; 79:66-86. [PMID: 28476525 DOI: 10.1016/j.neubiorev.2017.04.030] [Citation(s) in RCA: 385] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/15/2017] [Accepted: 04/10/2017] [Indexed: 02/06/2023]
Abstract
Aging is a natural process that is associated with cognitive decline as well as functional and social impairments. One structure of particular interest when considering aging and cognitive decline is the hippocampus, a brain region known to play an important role in learning and memory consolidation as well as in affective behaviours and mood regulation, and where both functional and structural plasticity (e.g., neurogenesis) occur well into adulthood. Neurobiological alterations seen in the aging hippocampus including increased oxidative stress and neuroinflammation, altered intracellular signalling and gene expression, as well as reduced neurogenesis and synaptic plasticity, are thought to be associated with age-related cognitive decline. Non-invasive strategies such as caloric restriction, physical exercise, and environmental enrichment have been shown to counteract many of the age-induced alterations in hippocampal signalling, structure, and function. Thus, such approaches may have therapeutic value in counteracting the deleterious effects of aging and protecting the brain against age-associated neurodegenerative processes.
Collapse
Affiliation(s)
- Luis E B Bettio
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Luckshi Rajendran
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joana Gil-Mohapel
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; UBC Island Medical program, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
29
|
Fan X, Wheatley EG, Villeda SA. Mechanisms of Hippocampal Aging and the Potential for Rejuvenation. Annu Rev Neurosci 2017; 40:251-272. [PMID: 28441118 DOI: 10.1146/annurev-neuro-072116-031357] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The past two decades have seen remarkable progress in our understanding of the multifactorial drivers of hippocampal aging and cognitive decline. Recent findings have also raised the possibility of functional rejuvenation in the aged hippocampus. In this review, we aim to synthesize the mechanisms that drive hippocampal aging and evaluate critically the potential for rejuvenation. We discuss the functional changes in synaptic plasticity and regenerative potential of the aged hippocampus, followed by mechanisms of microglia aging, and assess the cross talk between these proaging processes. We then examine proyouth interventions that demonstrate significant promise in reversing age-related impairments in the hippocampus and, finally, attempt to look ahead toward novel therapeutics for brain aging.
Collapse
Affiliation(s)
- Xuelai Fan
- Department of Anatomy, University of California, San Francisco, California 94143; , , .,The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California 94143
| | - Elizabeth G Wheatley
- Department of Anatomy, University of California, San Francisco, California 94143; , , .,The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California 94143.,Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, California 94143
| | - Saul A Villeda
- Department of Anatomy, University of California, San Francisco, California 94143; , , .,The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California 94143.,Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, California 94143
| |
Collapse
|
30
|
Penney J, Mendell A, Zeng M, Tran K, Lymer J, Turner PV, Choleris E, MacLusky N, Lu R. LUMAN/CREB3 is a key regulator of glucocorticoid-mediated stress responses. Mol Cell Endocrinol 2017; 439:95-104. [PMID: 27789393 DOI: 10.1016/j.mce.2016.10.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 10/21/2016] [Accepted: 10/21/2016] [Indexed: 01/28/2023]
Abstract
Altered glucocorticoid sensitivity is believed to contribute to a number of human diseases, including inflammatory and autoimmune conditions as well as disorders characterized by abnormal hypothalamic-pituitary-adrenal axis (HPA) function. LUMAN (or CREB3), originally identified through its interaction with a cell cycle regulator HCFC1, is an endoplasmic reticulum membrane-bound transcription factor that is involved in the unfolded protein response. Here we demonstrate that LUMAN changes the glucocorticoid response by modulating the expression of the glucocorticoid receptor leading to an overall increase in GR activity. Luman-deficient mice exhibited a blunted stress response characterized by low levels of both anxiety and depressive-like behaviour in addition to low circulating corticosterone levels. These mice also have reduced dendritic branching in the CA3 region of the hippocampus, consistent with increased GR responses. These findings are consistent with the notion that elevated GR activities are the primary cause of the observed phenotype in these LUMAN-deficient mice. We thus postulate that LUMAN is a key regulator of GR-mediated signaling and modulates HPA axis reactivity.
Collapse
Affiliation(s)
- Jenna Penney
- Department of Molecular and Cellular Biology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Ari Mendell
- Department of Biomedical Sciences, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Minghua Zeng
- Department of Molecular and Cellular Biology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Khoa Tran
- Department of Biomedical Sciences, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Jennifer Lymer
- Department of Psychology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Patricia V Turner
- Department of Pathobiology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Elena Choleris
- Department of Psychology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Neil MacLusky
- Department of Biomedical Sciences, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada
| | - Ray Lu
- Department of Molecular and Cellular Biology, University of Guelph, 488 Gordon Street, Guelph, Ontario, N1G 2W1, Canada.
| |
Collapse
|
31
|
Wellman CL. Visualizing Changes in Neuronal Dendritic Morphology in Response to Stress and Pharmacological Challenge. CURRENT PROTOCOLS IN NEUROSCIENCE 2017; 78:8.38.1-8.38.18. [PMID: 28046203 DOI: 10.1002/cpns.18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
This unit outlines a protocol for Golgi staining, which has been used extensively to reliably and quantitatively assess alterations in dendritic arborization and spine density as a result of a variety of factors, including chronic administration of glucocorticoids, chronic stress, and pharmacological manipulations. The method stains neurons in their entirety, allowing for sophisticated analyses of branch lengths and numbers as well as patterns of dendritic branching. Advantages of the technique include its usefulness in multisite collaborations and its utility in visualizing neurons in multiple regions within the same brain. Given that it typically labels approximately one in one hundred neurons, many neurons per region of interest can be sampled per animal, greatly increasing the ability to obtain a representative sample of neurons. Limitations include its time-consuming nature, the hazardous chemicals employed, and the inability to use the stain to identify discrete subpopulations of neurons based on their projections, activation, or protein expression. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Cara L Wellman
- Department of Psychological and Brain Sciences, Center for the Integrative Study of Animal Behavior, and Program in Neuroscience, Indiana University, Bloomington, Indiana
| |
Collapse
|
32
|
Koebele SV, Bimonte-Nelson HA. The endocrine-brain-aging triad where many paths meet: female reproductive hormone changes at midlife and their influence on circuits important for learning and memory. Exp Gerontol 2016; 94:14-23. [PMID: 27979770 DOI: 10.1016/j.exger.2016.12.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 01/15/2023]
Abstract
Female mammals undergo natural fluctuations in sex steroid hormone levels throughout life. These fluctuations span from early development, to cyclic changes associated with the menstrual or estrous cycle and pregnancy, to marked hormone flux during perimenopause, and a final decline at reproductive senescence. While the transition to reproductive senescence is not yet fully understood, the vast majority of mammals experience this spontaneous, natural phenomenon with age, which has broad implications for long-lived species. Indeed, this post-reproductive life stage, and its transition, involves significant and enduring physiological changes, including considerably altered sex steroid hormone and gonadotropin profiles that impact multiple body systems, including the brain. The endocrine-brain-aging triad is especially noteworthy, as many paths meet and interact. Many of the brain regions affected by aging are also sensitive to changes in ovarian hormone levels, and aging and reproductive senescence are both associated with changes in memory performance. This review explores how menopause is related to cognitive aging, and discusses some of the key neural systems and molecular factors altered with age and reproductive hormone level changes, with an emphasis on brain regions important for learning and memory.
Collapse
Affiliation(s)
- Stephanie V Koebele
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States
| | - Heather A Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, AZ 85287, United States; Arizona Alzheimer's Consortium, Phoenix, AZ 85006, United States.
| |
Collapse
|
33
|
Koss WA, Frick KM. Sex differences in hippocampal function. J Neurosci Res 2016; 95:539-562. [DOI: 10.1002/jnr.23864] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/28/2016] [Accepted: 07/11/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Wendy A. Koss
- Department of Psychology; University of Wisconsin-Milwaukee; Milwaukee Wisconsin
| | - Karyn M. Frick
- Department of Psychology; University of Wisconsin-Milwaukee; Milwaukee Wisconsin
| |
Collapse
|
34
|
Hatch RJ, Leinenga G, Götz J. Scanning Ultrasound (SUS) Causes No Changes to Neuronal Excitability and Prevents Age-Related Reductions in Hippocampal CA1 Dendritic Structure in Wild-Type Mice. PLoS One 2016; 11:e0164278. [PMID: 27727310 PMCID: PMC5058555 DOI: 10.1371/journal.pone.0164278] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/22/2016] [Indexed: 01/09/2023] Open
Abstract
Scanning ultrasound (SUS) is a noninvasive approach that has recently been shown to ameliorate histopathological changes and restore memory functions in an Alzheimer's disease mouse model. Although no overt neuronal damage was reported, the short- and long-term effects of SUS on neuronal excitability and dendritic tree morphology had not been investigated. To address this, we performed patch-clamp recordings from hippocampal CA1 pyramidal neurons in wild-type mice 2 and 24 hours after a single SUS treatment, and one week and 3 months after six weekly SUS treatments, including sham treatments as controls. In both treatment regimes, no changes in CA1 neuronal excitability were observed in SUS-treated neurons when compared to sham-treated neurons at any time-point. For the multiple treatment groups, we also determined the dendritic morphology and spine densities of the neurons from which we had recorded. The apical trees of sham-treated neurons were reduced at the 3 month time-point when compared to one week; however, surprisingly, no longitudinal change was detected in the apical dendritic trees of SUS-treated neurons. In contrast, the length and complexity of the basal dendritic trees were not affected by SUS treatment at either time-point. The apical dendritic spine densities were reduced, independent of the treatment group, at 3 months compared to one week. Collectively, these data suggest that ultrasound can be employed to prevent an age-associated loss of dendritic structure without impairing neuronal excitability.
Collapse
Affiliation(s)
- Robert John Hatch
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia Campus, Brisbane, QLD 4072, Australia
| | - Gerhard Leinenga
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia Campus, Brisbane, QLD 4072, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, St Lucia Campus, Brisbane, QLD 4072, Australia
- * E-mail:
| |
Collapse
|
35
|
Chronic Ampakine Treatments Stimulate Dendritic Growth and Promote Learning in Middle-Aged Rats. J Neurosci 2016; 36:1636-46. [PMID: 26843645 DOI: 10.1523/jneurosci.3157-15.2016] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Positive allosteric modulators of AMPA-type glutamate receptors (ampakines) have been shown to rescue synaptic plasticity and reduce neuropathology in rodent models of cognitive disorders. Here we tested whether chronic ampakine treatment offsets age-related dendritic retraction in middle-aged (MA) rats. Starting at 10 months of age, rats were housed in an enriched environment and given daily treatment with a short half-life ampakine or vehicle for 3 months. Dendritic branching and spine measures were collected from 3D reconstructions of Lucifer yellow-filled CA1 pyramidal cells. There was a substantial loss of secondary branches, relative to enriched 2.5-month-old rats, in apical and basal dendritic fields of vehicle-treated, but not ampakine-treated, 13-month-old rats. Baseline synaptic responses in CA1 were only subtly different between the two MA groups, but long-term potentiation was greater in ampakine-treated rats. Unsupervised learning of a complex environment was used to assess treatment effects on behavior. Vehicle- and drug-treated rats behaved similarly during a first 30 min session in the novel environment but differed markedly on subsequent measures of long-term memory. Markov sequence analysis uncovered a clear increase in the predictability of serial movements between behavioral sessions 2 and 3 in the ampakine, but not vehicle, group. These results show that a surprising degree of dendritic retraction occurs by middle age and that this can be mostly offset by pharmacological treatments without evidence for unwanted side effects. The functional consequences of rescue were prominent with regard to memory but also extended to self-organization of behavior. SIGNIFICANCE STATEMENT Brain aging is characterized by a progressive loss of dendritic arbors and the emergence of impairments to learning-related synaptic plasticity. The present studies show that dendritic losses are evident by middle age despite housing in an enriched environment and can be mostly reversed by long-term, oral administration of a positive allosteric modulator of AMPA-type glutamate receptors. Dendritic recovery was accompanied by improvements to both synaptic plasticity and the encoding of long-term memory of a novel, complex environment. Because the short half-life compound had no evident negative effects, the results suggest a plausible strategy for treating age-related neuronal deterioration.
Collapse
|
36
|
Raber J, Allen AR, Weber S, Chakraborti A, Sharma S, Fike JR. Effect of behavioral testing on spine density of basal dendrites in the CA1 region of the hippocampus modulated by (56)Fe irradiation. Behav Brain Res 2016; 302:263-8. [PMID: 26801826 DOI: 10.1016/j.bbr.2016.01.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/13/2016] [Accepted: 01/16/2016] [Indexed: 01/11/2023]
Abstract
A unique feature of the space radiation environment is the presence of high-energy charged particles, including (56)Fe ions, which can present a significant hazard to space flight crews during and following a mission. (56)Fe irradiation-induced cognitive changes often involve alterations in hippocampal function. These alterations might involve changes in spine morphology and density. In addition to irradiation, performing a cognitive task can also affect spine morphology. Therefore, it is often hard to determine whether changes in spine morphology and density are due to an environmental challenge or group differences in performance on cognitive tests. In this study, we tested the hypothesis that the ability of exploratory behavior to increase specific measures of hippocampal spine morphology and density is affected by (56)Fe irradiation. In sham-irradiated mice, exploratory behavior increased basal spine density in the CA1 region of the hippocampus and the enclosed blade of the dentate gyrus. These effects were not seen in irradiated mice. In addition, following exploratory behavior, there was a trend toward a decrease in the percent stubby spines on apical dendrites in the CA3 region of the hippocampus in (56)Fe-irradiated, but not sham-irradiated, mice. Other hippocampal regions and spine measures affected by (56)Fe irradiation showed comparable radiation effects in behaviorally naïve and cognitively tested mice. Thus, the ability of exploratory behavior to alter spine density and morphology in specific hippocampal regions is affected by (56)Fe irradiation.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States; Departments of Neurology, Radiation Medicine and Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States.
| | - Antiño R Allen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Sydney Weber
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States
| | - Ayanabha Chakraborti
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, CA 94110, United States; The Brain Research Institute at Monash Sunway, Selangor Darul Ehsan, Malaysia
| | - Sourabh Sharma
- Departments of Neurology, Radiation Medicine and Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States
| | - John R Fike
- Departments of Neurology, Radiation Medicine and Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States; Department of Radiation Oncology, University of California, San Francisco, CA 94110, United States
| |
Collapse
|
37
|
Maysinger D, Ji J, Hutter E, Cooper E. Nanoparticle-Based and Bioengineered Probes and Sensors to Detect Physiological and Pathological Biomarkers in Neural Cells. Front Neurosci 2015; 9:480. [PMID: 26733793 PMCID: PMC4683200 DOI: 10.3389/fnins.2015.00480] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/30/2015] [Indexed: 01/11/2023] Open
Abstract
Nanotechnology, a rapidly evolving field, provides simple and practical tools to investigate the nervous system in health and disease. Among these tools are nanoparticle-based probes and sensors that detect biochemical and physiological properties of neurons and glia, and generate signals proportionate to physical, chemical, and/or electrical changes in these cells. In this context, quantum dots (QDs), carbon-based structures (C-dots, grapheme, and nanodiamonds) and gold nanoparticles are the most commonly used nanostructures. They can detect and measure enzymatic activities of proteases (metalloproteinases, caspases), ions, metabolites, and other biomolecules under physiological or pathological conditions in neural cells. Here, we provide some examples of nanoparticle-based and genetically engineered probes and sensors that are used to reveal changes in protease activities and calcium ion concentrations. Although significant progress in developing these tools has been made for probing neural cells, several challenges remain. We review many common hurdles in sensor development, while highlighting certain advances. In the end, we propose some future directions and ideas for developing practical tools for neural cell investigations, based on the maxim "Measure what is measurable, and make measurable what is not so" (Galileo Galilei).
Collapse
Affiliation(s)
- Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University Montreal, QC, Canada
| | - Jeff Ji
- Department of Pharmacology and Therapeutics, McGill University Montreal, QC, Canada
| | - Eliza Hutter
- Department of Pharmacology and Therapeutics, McGill University Montreal, QC, Canada
| | - Elis Cooper
- Department of Physiology, McGill University Montreal, QC, Canada
| |
Collapse
|
38
|
Bullmann T, Seeger G, Stieler J, Hanics J, Reimann K, Kretzschmann TP, Hilbrich I, Holzer M, Alpár A, Arendt T. Tau phosphorylation-associated spine regression does not impair hippocampal-dependent memory in hibernating golden hamsters. Hippocampus 2015; 26:301-18. [DOI: 10.1002/hipo.22522] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 08/12/2015] [Accepted: 08/31/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Torsten Bullmann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
- Frey Initiative Research Unit, RIKEN Quantitative Biology Center; Japan
| | - Gudrun Seeger
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Jens Stieler
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - János Hanics
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences; Budapest Hungary
- Department of Anatomy; Semmelweis University; Budapest Hungary
| | - Katja Reimann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Tanja Petra Kretzschmann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Isabel Hilbrich
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Max Holzer
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Alán Alpár
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences; Budapest Hungary
- Department of Anatomy; Semmelweis University; Budapest Hungary
| | - Thomas Arendt
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| |
Collapse
|
39
|
Seliger SL, Wendell CR, Waldstein SR, Ferrucci L, Zonderman AB. Renal function and long-term decline in cognitive function: the Baltimore Longitudinal Study of Aging. Am J Nephrol 2015. [PMID: 26201453 DOI: 10.1159/000430922] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Renal disease has been associated with greater risk of dementia and greater cognitive impairment. However, the relationship of lower renal function with long-term decline in specific domains of cognitive function remains unclear among community-dwelling, non-demented individuals. METHODS Stroke- and dementia-free participants (n = 2,116) were enrolled in the Baltimore Longitudinal Study of Aging, a community-based, prospective, longitudinal study. Renal function was estimated by the inverse of serum creatinine adjusted for age, sex and race and (in sensitivity analyses) estimated glomerular filtration rate (eGFR) using the MDRD formula. Outcome measures were changes in scores on 6 cognitive tests encompassing a range of cognitive functions, measured at 2-year intervals. Mixed-effects regression models examined the longitudinal relations of renal function with cognitive functions after adjusting for demographics, comorbidity and other potential confounders. RESULTS Mean age at initial testing was 53.9 years (SD 17.1), and 94 participants (4.4%) had an eGFR <60 ml/min/1.73 m(2) and 18.5% had at least one comorbidity. With increasing age, longitudinal increases in creatinine concentrations were associated with more rapid decline in performance on several cognitive measures, including the learning slope of the California Verbal Learning Test, a test of verbal learning (p < 0.01), and the Benton Visual Retention Test, a test of visual memory (p < 0.01). Associations were similar for changes in eGFRMDRD, which was also associated with the rate of decline in verbal memory. CONCLUSION In a community-based adult population, declines in renal function independently associated with greater long-term declines in visual memory and verbal memory and learning.
Collapse
Affiliation(s)
- Stephen L Seliger
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Md., USA
| | | | | | | | | |
Collapse
|
40
|
Bowman RE, Luine V, Diaz Weinstein S, Khandaker H, DeWolf S, Frankfurt M. Bisphenol-A exposure during adolescence leads to enduring alterations in cognition and dendritic spine density in adult male and female rats. Horm Behav 2015; 69:89-97. [PMID: 25554518 PMCID: PMC6116732 DOI: 10.1016/j.yhbeh.2014.12.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 12/16/2014] [Accepted: 12/20/2014] [Indexed: 01/01/2023]
Abstract
We have previously demonstrated that adolescent exposure of rats to bisphenol-A (BPA), an environmental endocrine disrupter, increases anxiety, impairs spatial memory, and decreases dendritic spine density in the CA1 region of the hippocampus (CA1) and medial prefrontal cortex (mPFC) when measured in adolescents in both sexes. The present study examined whether the behavioral and morphological alterations following BPA exposure during adolescent development are maintained into adulthood. Male and female, adolescent rats received BPA, 40μg/kg/bodyweight, or control treatments for one week. In adulthood, subjects were tested for anxiety and locomotor activity, spatial memory, non-spatial visual memory, and sucrose preference. Additionally, stress-induced serum corticosterone levels and dendritic spine density in the mPFC and CA1 were measured. BPA-treated males, but not females, had decreased arm visits on the elevated plus maze, but there was no effect on anxiety. Non-spatial memory, object recognition, was also decreased in BPA treated males, but not in females. BPA exposure did not alter spatial memory, object placement, but decreased exploration during the tasks in both sexes. No significant group differences in sucrose preference or serum corticosterone levels in response to a stress challenge were found. However, BPA exposure, regardless of sex, significantly decreased spine density of both apical and basal dendrites on pyramidal cells in CA1 but had no effect in the mPFC. Current data are discussed in relation to BPA dependent changes, which were present during adolescence and did, or did not, endure into adulthood. Overall, adolescent BPA exposure, below the current reference safe daily limit set by the U.S.E.P.A., leads to alterations in some behaviors and neuronal morphology that endure into adulthood.
Collapse
Affiliation(s)
- Rachel E Bowman
- Department of Psychology, Sacred Heart University, Fairfield, CT 06825, USA.
| | - Victoria Luine
- Department of Psychology, Hunter College, CUNY, New York, NY 10065, USA
| | | | - Hameda Khandaker
- Department of Psychology, Hunter College, CUNY, New York, NY 10065, USA
| | - Sarah DeWolf
- Department of Psychology, Sacred Heart University, Fairfield, CT 06825, USA
| | - Maya Frankfurt
- Department of Science Education, Hofstra North Shore-LIJ School of Medicine, Hempstead, NY 11549, USA
| |
Collapse
|
41
|
Pietranera L, Brocca ME, Roig P, Lima A, Garcia-Segura LM, De Nicola AF. Estrogens are neuroprotective factors for hypertensive encephalopathy. J Steroid Biochem Mol Biol 2015; 146:15-25. [PMID: 24736028 DOI: 10.1016/j.jsbmb.2014.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 10/25/2022]
Abstract
Estrogens are neuroprotective factors for brain diseases, including hypertensive encephalopathy. In particular, the hippocampus is highly damaged by high blood pressure, with several hippocampus functions being altered in humans and animal models of hypertension. Working with a genetic model of primary hypertension, the spontaneously hypertensive rat (SHR), we have shown that SHR present decreased dentate gyrus neurogenesis, astrogliosis, low expression of brain derived neurotrophic factor (BDNF), decreased number of neurons in the hilus of the dentate gyrus, increased basal levels of the estrogen-synthesizing enzyme aromatase, and atrophic dendritic arbor with low spine density in the CA1 region compared to normotensive Wistar Kyoto (WKY) ratsl. Changes also occur in the hypothalamus of SHR, with increased expression of the hypertensinogenic peptide arginine vasopressin (AVP) and its V1b receptor. Following chronic estradiol treatment, SHR show decreased blood pressure, enhanced hippocampus neurogenesis, decreased the reactive astrogliosis, increased BDNF mRNA and protein expression in the dentate gyrus, increased neuronal number in the hilus of the dentate gyrus, further increased the hyperexpression of aromatase and replaced spine number with remodeling of the dendritic arbor of the CA1 region. We have detected by qPCR the estradiol receptors ERα and ERβ in hippocampus from both SHR and WKY rats, suggesting direct effects of estradiol on brain cells. We hypothesize that a combination of exogenously given estrogens plus those locally synthesized by estradiol-stimulated aromatase may better alleviate the hippocampal and hypothalamic encephalopathy of SHR. This article is part of a Special Issue entitled "Sex steroids and brain disorders".
Collapse
Affiliation(s)
- Luciana Pietranera
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina; Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425 Buenos Aires, Argentina
| | - Maria Elvira Brocca
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Paulina Roig
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Analia Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas, Avenida Doctor Arce 37, E-28002 Madrid, Spain
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina; Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425 Buenos Aires, Argentina.
| |
Collapse
|
42
|
Dhupia N, Rathour RK, Narayanan R. Dendritic atrophy constricts functional maps in resonance and impedance properties of hippocampal model neurons. Front Cell Neurosci 2015; 8:456. [PMID: 25628537 PMCID: PMC4289900 DOI: 10.3389/fncel.2014.00456] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 12/16/2014] [Indexed: 11/23/2022] Open
Abstract
A gradient in the density of hyperpolarization-activated cyclic-nucleotide gated (HCN) channels is necessary for the emergence of several functional maps within hippocampal pyramidal neurons. Here, we systematically analyzed the impact of dendritic atrophy on nine such functional maps, related to input resistance and local/transfer impedance properties, using conductance-based models of hippocampal pyramidal neurons. We introduced progressive dendritic atrophy in a CA1 pyramidal neuron reconstruction through a pruning algorithm, measured all functional maps in each pruned reconstruction, and arrived at functional forms for the dependence of underlying measurements on dendritic length. We found that, across frequencies, atrophied neurons responded with higher efficiency to incoming inputs, and the transfer of signals across the dendritic tree was more effective in an atrophied reconstruction. Importantly, despite the presence of identical HCN-channel density gradients, spatial gradients in input resistance, local/transfer resonance frequencies and impedance profiles were significantly constricted in reconstructions with dendritic atrophy, where these physiological measurements across dendritic locations converged to similar values. These results revealed that, in atrophied dendritic structures, the presence of an ion channel density gradient alone was insufficient to sustain homologous functional maps along the same neuronal topograph. We assessed the biophysical basis for these conclusions and found that this atrophy-induced constriction of functional maps was mediated by an enhanced spatial spread of the influence of an HCN-channel cluster in atrophied trees. These results demonstrated that the influence fields of ion channel conductances need to be localized for channel gradients to express themselves as homologous functional maps, suggesting that ion channel gradients are necessary but not sufficient for the emergence of functional maps within single neurons.
Collapse
Affiliation(s)
- Neha Dhupia
- Cellular Neurophysiology Laboratory, Indian Institute of Science Bangalore, India ; Centre for Converging Technologies, University of Rajasthan Jaipur, India
| | - Rahul K Rathour
- Cellular Neurophysiology Laboratory, Indian Institute of Science Bangalore, India
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Indian Institute of Science Bangalore, India
| |
Collapse
|
43
|
Ménard C, Gaudreau P, Quirion R. Signaling pathways relevant to cognition-enhancing drug targets. Handb Exp Pharmacol 2015; 228:59-98. [PMID: 25977080 DOI: 10.1007/978-3-319-16522-6_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aging is generally associated with a certain cognitive decline. However, individual differences exist. While age-related memory deficits can be observed in humans and rodents in the absence of pathological conditions, some individuals maintain intact cognitive functions up to an advanced age. The mechanisms underlying learning and memory processes involve the recruitment of multiple signaling pathways and gene expression, leading to adaptative neuronal plasticity and long-lasting changes in brain circuitry. This chapter summarizes the current understanding of how these signaling cascades could be modulated by cognition-enhancing agents favoring memory formation and successful aging. It focuses on data obtained in rodents, particularly in the rat as it is the most common animal model studied in this field. First, we will discuss the role of the excitatory neurotransmitter glutamate and its receptors, downstream signaling effectors [e.g., calcium/calmodulin-dependent protein kinase II (CaMKII), protein kinase C (PKC), extracellular signal-regulated kinases (ERK), mammalian target of rapamycin (mTOR), cAMP response element-binding protein (CREB)], associated immediate early gene (e.g., Homer 1a, Arc and Zif268), and growth factors [insulin-like growth factors (IGFs) and brain-derived neurotrophic factor (BDNF)] in synaptic plasticity and memory formation. Second, the impact of the cholinergic system and related modulators on memory will be briefly reviewed. Finally, since dynorphin neuropeptides have recently been associated with memory impairments in aging, it is proposed as an attractive target to develop novel cognition-enhancing agents.
Collapse
Affiliation(s)
- Caroline Ménard
- Douglas Mental Health University Institute, McGill University, Perry Pavilion, 6875 LaSalle Boulevard, Montreal, QC, Canada, H4H 1R3
| | | | | |
Collapse
|
44
|
Fortress AM, Frick KM. Epigenetic regulation of estrogen-dependent memory. Front Neuroendocrinol 2014; 35:530-49. [PMID: 24878494 PMCID: PMC4174980 DOI: 10.1016/j.yfrne.2014.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 02/09/2023]
Abstract
Hippocampal memory formation is highly regulated by post-translational histone modifications and DNA methylation. Accordingly, these epigenetic processes play a major role in the effects of modulatory factors, such as sex steroid hormones, on hippocampal memory. Our laboratory recently demonstrated that the ability of the potent estrogen 17β-estradiol (E2) to enhance hippocampal-dependent novel object recognition memory in ovariectomized female mice requires ERK-dependent histone H3 acetylation and DNA methylation in the dorsal hippocampus. Although these data provide valuable insight into the chromatin modifications that mediate the memory-enhancing effects of E2, epigenetic regulation of gene expression is enormously complex. Therefore, more research is needed to fully understand how E2 and other hormones employ epigenetic alterations to shape behavior. This review discusses the epigenetic alterations shown thus far to regulate hippocampal memory, briefly reviews the effects of E2 on hippocampal function, and describes in detail our work on epigenetic regulation of estrogenic memory enhancement.
Collapse
Affiliation(s)
- Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
45
|
Kim A, Zamora-Martinez ER, Edwards S, Mandyam CD. Structural reorganization of pyramidal neurons in the medial prefrontal cortex of alcohol dependent rats is associated with altered glial plasticity. Brain Struct Funct 2014; 220:1705-20. [PMID: 24667898 PMCID: PMC4177030 DOI: 10.1007/s00429-014-0755-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 03/10/2014] [Indexed: 12/22/2022]
Abstract
In rodents, chronic intermittent ethanol vapor exposure (CIE) produces alcohol dependence, alters the activity of pyramidal neurons and decreases the number of glial progenitors in the medial prefrontal cortex (mPFC). Adult male Wistar rats were exposed to CIE and were injected with mitotic markers to label and phenotype proliferating cells to test the hypothesis that CIE produces concurrent alterations in the structure of pyramidal neurons and the cell cycle kinetics and developmental stages of glial progenitors in the mPFC. Medial prefrontal cortical tissue was processed for Golgi-Cox staining, immunohistochemistry and Western blotting analysis. CIE increased dendritic arborization and spine densities within basal and apical dendrites of pyramidal neurons via aberrant reorganization of actin cytoskeleton-associated molecules. CIE concomitantly increased the expression of total NR2B subunits without affecting phosphorylation of NR2B at Tyr-1472 or levels of PSD-95. CIE reduced the length of S-phase of the cell cycle of glial progenitors and reduced proliferation and differentiation of progenitors into bHLH transcription factor Olig2-expressing premyelinating oligodendrocyte progenitor cells (OPCs). CIE also produced a corresponding hyperphosphorylation of Olig2, and reduced expression of myelin basic protein. Our findings demonstrate that CIE-induced alterations in OPCs and myelin-related proteins are associated with profound alterations in the structure of pyramidal neurons. In sum, our results not only provide evidence that alcohol dependence leads to pathological changes in the mPFC, which may in part define a cellular basis for cognitive impairments associated with alcoholism, but also show dependence-associated morphological changes in the PFC at the single neuron level.
Collapse
Affiliation(s)
- Airee Kim
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Eva R. Zamora-Martinez
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Scott Edwards
- Department of Physiology, Alcohol & Drug Abuse Center of Excellence, LSU Health Sciences Center, New Orleans, LA, USA
| | - Chitra D. Mandyam
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Skaggs School of Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
46
|
Effect of Boswellia serrata gum resin on the morphology of hippocampal CA1 pyramidal cells in aged rat. Anat Sci Int 2014; 90:47-53. [DOI: 10.1007/s12565-014-0228-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 01/16/2014] [Indexed: 12/29/2022]
|
47
|
Brain SERT Expression of Male Rats Is Reduced by Aging and Increased by Testosterone Restitution. NEUROSCIENCE JOURNAL 2013; 2013:201909. [PMID: 26317087 PMCID: PMC4437264 DOI: 10.1155/2013/201909] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 11/23/2013] [Indexed: 11/28/2022]
Abstract
In preclinical and clinical studies aging has been associated with a deteriorated response to antidepressant treatment. We hypothesize that such impairment is explained by an age-related decrease in brain serotonin transporter (SERT) expression associated with low testosterone (T) levels. The objectives of this study were to establish (1) if brain SERT expression is reduced by aging and (2) if the SERT expression in middle-aged rats is increased by T-restitution. Intact young rats (3–5 months) and gonad-intact middle-aged rats with or without T-restitution were used. The identification of the brain SERT expression was done by immunofluorescence in prefrontal cortex, lateral septum, hippocampus, and raphe nuclei. An age-dependent reduction of SERT expression was observed in all brain regions examined, while T-restitution recovered the SERT expression only in the dorsal raphe of middle-aged rats. This last action seems relevant since dorsal raphe plays an important role in the antidepressant action of selective serotonin reuptake inhibitors. All data suggest that this mechanism accounts for the T-replacement usefulness to improve the response to antidepressants in the aged population.
Collapse
|
48
|
Kato A, Hojo Y, Higo S, Komatsuzaki Y, Murakami G, Yoshino H, Uebayashi M, Kawato S. Female hippocampal estrogens have a significant correlation with cyclic fluctuation of hippocampal spines. Front Neural Circuits 2013; 7:149. [PMID: 24151456 PMCID: PMC3798982 DOI: 10.3389/fncir.2013.00149] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 09/03/2013] [Indexed: 01/07/2023] Open
Abstract
Synaptic plasticity of the female hippocampus may cyclically fluctuate across the estrous cycle. The spine density fluctuation had been explained by fluctuation of plasma estradiol (E2) and progesterone (PROG), with the assumption that these steroids penetrate into the hippocampus. Recently, however, we demonstrated that male hippocampal levels of sex steroids are much higher than those in plasma, suggesting a weak contribution of plasma steroids to the spine density. By combination of mass-spectrometric analysis with HPLC-purification and picolinoyl-derivatization of hippocampal sex steroids, we determined the accurate concentration of E2 and PROG at four stages of plasma estrous cycle including Proestrus (Pro), Estrus (Est), Diestrus 1 (D1), and Diestrus 2 (D2). Hippocampal levels of E2 and PROG showed cyclic fluctuation with a peak at Pro for E2 and at D1 for PROG, having a positive correlation with the plasma estrous cycle. All these sex steroid levels are much higher in the hippocampus than in plasma. Even after ovariectomy a significant levels of E2 and PROG were observed in the hippocampus. The total spine density showed higher values at Pro and D1, and lower values at Est and D2, having a good correlation with the peak levels of hippocampal E2 or PROG. We also examined fluctuation of the head diameter of spines. Interestingly, mRNA expression level of steroidogenic enzymes (P450arom and 17β-HSD, etc.) and sex-steroid receptors did not significantly change across the estrous cycle. Therefore, the fluctuation of total hippocampal PROG (equal to sum of hippocampus-synthesized PROG and plasma PROG) may be originated from the contribution of cyclic change in plasma PROG, which can induce the fluctuation of total hippocampal E2, since steroid conversion activity of hippocampus might be nearly the same across the estrus cycle.
Collapse
Affiliation(s)
- Asami Kato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Markham JA, Mullins SE, Koenig JI. Periadolescent maturation of the prefrontal cortex is sex-specific and is disrupted by prenatal stress. J Comp Neurol 2013; 521:1828-43. [PMID: 23172080 DOI: 10.1002/cne.23262] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 09/01/2012] [Accepted: 11/06/2012] [Indexed: 12/19/2022]
Abstract
The prefrontal cortex (PFC) undergoes dramatic, sex-specific maturation during adolescence. Adolescence is a vulnerable window for developing mental illnesses that show significant sexual dimorphisms. Gestational stress is associated with increased risk for both schizophrenia, which is more common among men, and cognitive deficits. We have shown that male, but not female, rats exposed to prenatal stress develop postpubertal deficits in cognitive behaviors supported by the prefrontal cortex. Here we tested the hypothesis that repeated variable prenatal stress during the third week of rat gestation disrupts periadolescent development of prefrontal neurons in a sex-specific fashion. Using Golgi-Cox stained tissue, we compared dendritic arborization and spine density of prelimbic layer III neurons in prenatally stressed and control animals at juvenile (day 20), prepubertal (day 30), postpubertal (day 56), and adult (day 90) ages (N = 115). Dendritic ramification followed a sex-specific pattern that was disrupted during adolescence in prenatally stressed males, but not in females. In contrast, the impact of prenatal stress on the female PFC was not evident until adulthood. Prenatal stress also caused reductions in brain and body weights, and the latter effect was more pronounced among males. Additionally, there was a trend toward reduced testosterone levels for adult prenatally stressed males. Our findings indicate that, similarly to humans, the rat PFC undergoes sex-specific development during adolescence and furthermore that this process is disrupted by prenatal stress. These findings may be relevant to both the development of normal sex differences in cognition as well as differential male-female vulnerability to psychiatric conditions.
Collapse
Affiliation(s)
- Julie A Markham
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland-Baltimore School of Medicine, Baltimore, Maryland 21228, USA.
| | | | | |
Collapse
|
50
|
Brocca ME, Pietranera L, Beauquis J, De Nicola AF. Estradiol increases dendritic length and spine density in CA1 neurons of the hippocampus of spontaneously hypertensive rats: a Golgi impregnation study. Exp Neurol 2013; 247:158-64. [PMID: 23628746 DOI: 10.1016/j.expneurol.2013.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 03/20/2013] [Accepted: 04/12/2013] [Indexed: 02/06/2023]
Abstract
Increased neuronal vulnerability has been described in the brain of spontaneously hypertensive rats (SHR), models of primary hypertension. Previous data indicate that estradiol treatment corrects several dysfunctions of the hippocampus and hypothalamus of SHR. Considering this evidence we analyzed the dendritic arborization and spine density of the CA1 subfield in SHR and Wistar-Kyoto (WKY) normotensive rats with and without estradiol treatment. Five month old male SHR and WKY rats received single estradiol or cholesterol pellets (sham treatment) for 2 weeks. A substantial rise of circulating estradiol (>25 fold) and testicular atrophy was present in all estradiol-receiving rats. In both SHR and WKY rats, estradiol decreased blood pressure by ~20 mm Hg; however, a moderate hypertension persisted in SHR (164 mm Hg). Using a modified Golgi impregnation technique, apical and basal dendrites of the CA1 subfield were subjected to Sholl analysis. Spine density was also statistically analyzed. Apical dendritic length was significantly lower in SHR compared to WKY rats (p<0.01), whereas estradiol treatment increased dendritic length in the SHR group only (SHR vs SHR+estradiol; p<0.01). Apical dendritic length plotted against the shell distances 20-100, 120-200 and 220-300 μm, revealed that changes were more pronounced in the range 120-200 μm between SHR vs. WKY rats (p<0.05) and SHR vs. SHR+estradiol (p<0.05). Instead, basal dendrites were not significantly modified by hypertension or steroid treatment. Spine density of apical dendrites was lower in SHR than WKY (p<0.05) and was up-regulated in the SHR+estradiol group compared to the SHR group (p<0.001). Similar changes were obtained for basal dendritic spines. These data suggest that changes of neuronal processes in SHR are plastic events restorable by estradiol treatment. In conjunction with previous results, the present data reveal new targets of estradiol neuroprotection in the brain of hypertensive rats.
Collapse
Affiliation(s)
- María Elvira Brocca
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Argentina
| | | | | | | |
Collapse
|