1
|
Lee SY, Lenef JD, Delgado Cornejo DO, Ortiz-Ortiz AM, Ma T, Arthur TS, Roberts CA, Dasgupta NP. Tuning the selectivity of bimetallic Cu electrocatalysts for CO 2 reduction using atomic layer deposition. Chem Commun (Camb) 2024. [PMID: 39688273 DOI: 10.1039/d4cc04820b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Cu-Zn bimetallic catalysts were synthesized on 3-D gas diffusion electrodes using atomic layer deposition (ALD) techniques. Electrochemical CO2 reduction was evaluated, and a significant variation in the product selectivity was observed compared to unmodified Cu catalysts. As low as a single ALD cycle of ZnO resulted in a reduction of C2H4 production and shift towards CO selectivity, which is attributed to changes in the chemical state of the surface. Our findings demonstrate the impact of atomically-precise surface modifications on electrocatalyst selectivity.
Collapse
Affiliation(s)
- Si Young Lee
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Julia D Lenef
- Department of Materials Science & Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Alondra M Ortiz-Ortiz
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Tao Ma
- Michigan Center for Materials Characterization, University of Michigan, Ann Arbor, MI 48109, USA
| | - Timothy S Arthur
- Toyota Research Institute of North America, Ann Arbor, MI, 48105, USA.
| | - Charles A Roberts
- Toyota Research Institute of North America, Ann Arbor, MI, 48105, USA.
| | - Neil P Dasgupta
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
- Department of Materials Science & Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
2
|
Castillon CCM, Otsuka S, Armstrong JN, Contractor A. Subregional activity in the dentate gyrus is amplified during elevated cognitive demands. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.31.621367. [PMID: 39554113 PMCID: PMC11565968 DOI: 10.1101/2024.10.31.621367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Neural activity in the dentate gyrus (DG) is required for the detection and discrimination of novelty, context and patterns, amongst other cognitive processes. Prior work has demonstrated that there are differences in the activation of granule neurons in the supra and infrapyramidal blades of the DG during a range of hippocampal dependent tasks. Here we used an automated touch screen pattern separation task combined to temporally controlled tagging of active neurons to determine how performance in a cognitively demanding task affected patterns of neural activity in the DG. We found an increase in the blade-biased activity of suprapyramidal mature granule cells (mGCs) during the performance of a high cognitive demand segment of the task, with a further characteristic distribution of active neurons along the apex to blade, and hilar to molecular layer axes. Chemogenetic inhibition of adult-born granule cells (abDGCs) beyond a critical window of their maturation significantly impaired performance of mice when cognitive demand was high, but not when it was low. abDGC inhibition also elevated the total activity of mGCs and disturbed the patterned distribution of active mGCs even in mice that eventually succeeded in the task. Conversely chemogenetic inhibition of mGCs reduced success in the high cognitive demand portion of this task and decreased the global number of active GCs without affecting the patterned distribution of active cells. These findings demonstrate how a high cognitive demand pattern separation task preferentially activates mGCs in subregions of the DG and are consistent with an inhibitory role for abDGCs on the dentate circuit which in part governs the spatially organized patterns of activity of mGCs.
Collapse
|
3
|
Rabiller G, Ip Z, Zarrabian S, Zhang H, Sato Y, Yazdan-Shahmorad A, Liu J. Type-2 Diabetes Alters Hippocampal Neural Oscillations and Disrupts Synchrony between the Hippocampus and Cortex. Aging Dis 2024; 15:2255-2270. [PMID: 38029397 PMCID: PMC11346393 DOI: 10.14336/ad.2023.1106] [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: 09/06/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) increases the risk of neurological diseases, yet how brain oscillations change as age and T2DM interact is not well characterized. To delineate the age and diabetic effect on neurophysiology, we recorded local field potentials with multichannel electrodes spanning the somatosensory cortex and hippocampus (HPC) under urethane anesthesia in diabetic and normoglycemic control mice, at 200 and 400 days of age. We analyzed the signal power of brain oscillations, brain state, sharp wave associate ripples (SPW-Rs), and functional connectivity between the cortex and HPC. We found that while both age and T2DM were correlated with a breakdown in long-range functional connectivity and reduced neurogenesis in the dentate gyrus and subventricular zone, T2DM further slowed brain oscillations and reduced theta-gamma coupling. Age and T2DM also prolonged the duration of SPW-Rs and increased gamma power during SPW-R phase. Our results have identified potential electrophysiological substrates of hippocampal changes associated with T2DM and age. The perturbed brain oscillation features and diminished neurogenesis may underlie T2DM-accelerated cognitive impairment.
Collapse
Affiliation(s)
- Gratianne Rabiller
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- San Francisco VA medical Center, San Francisco, CA, USA
| | - Zachary Ip
- Departments of Bioengineering, University of Washington, Seattle, WA, USA
| | - Shahram Zarrabian
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- San Francisco VA medical Center, San Francisco, CA, USA
| | - Hongxia Zhang
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- San Francisco VA medical Center, San Francisco, CA, USA
| | - Yoshimichi Sato
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- San Francisco VA medical Center, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Azadeh Yazdan-Shahmorad
- Departments of Bioengineering, University of Washington, Seattle, WA, USA
- Electrical and Computer Engineering, University of Washington, Seattle, WA, USA
| | - Jialing Liu
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
- San Francisco VA medical Center, San Francisco, CA, USA
| |
Collapse
|
4
|
Wang Z, Yang K, Sun X. Effect of adult hippocampal neurogenesis on pattern separation and its applications. Cogn Neurodyn 2024; 18:1-14. [PMID: 39568526 PMCID: PMC11564429 DOI: 10.1007/s11571-024-10110-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 11/22/2024] Open
Abstract
Adult hippocampal neurogenesis (AHN) is considered essential in memory formation. The dentate gyrus neural network containing newborn dentate gyrus granule cells at the critical period (4-6 weeks) have been widely discussed in neurophysiological and behavioral experiments. However, how newborn dentate gyrus granule cells at this critical period influence pattern separation of dentate gyrus remains unclear. To address this issue, we propose a biologically related dentate gyrus neural network model with AHN. By Leveraging this model, we find pattern separation is enhanced at the medium level of neurogenesis (5% of mature granule cells). This is because the sparse firing of mature granule cells is increased. We can understand this change from the following two aspects. On one hand, newborn granule cells compete with mature granule cells for inputs from the entorhinal cortex, thereby weakening the firing of mature granule cells. On the other hand, newborn granule cells effectively enhance the feedback inhibition level of the network by promoting the firing of interneurons (Mossy cells and Basket cells) and then indirectly regulating the sparse firing of mature granule cells. To verify the validity of the model for pattern separation, we apply the proposed model to a similar concept separation task and reveal that our model outperforms the original model counterparts in this task.
Collapse
Affiliation(s)
- Zengbin Wang
- School of Science, Beijing University of Posts and Telecommunications, #10 Xitucheng Road, Beijing, 100876 People's Republic of China
| | - Kai Yang
- School of Science, Beijing University of Posts and Telecommunications, #10 Xitucheng Road, Beijing, 100876 People's Republic of China
| | - Xiaojuan Sun
- School of Science, Beijing University of Posts and Telecommunications, #10 Xitucheng Road, Beijing, 100876 People's Republic of China
| |
Collapse
|
5
|
Chen H, Mo P, Zhu J, Xu X, Cheng Z, Yang F, Xu Z, Liu J, Wang L. Anionic Coordination Control in Building Cu-Based Electrocatalytic Materials for CO 2 Reduction Reaction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400661. [PMID: 38597688 DOI: 10.1002/smll.202400661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/22/2024] [Indexed: 04/11/2024]
Abstract
Renewable energy-driven conversion of CO2 to value-added fuels and chemicals via electrochemical CO2 reduction reaction (CO2RR) technology is regarded as a promising strategy with substantial environmental and economic benefits to achieve carbon neutrality. Because of its sluggish kinetics and complex reaction paths, developing robust catalytic materials with exceptional selectivity to the targeted products is one of the core issues, especially for extensively concerned Cu-based materials. Manipulating Cu species by anionic coordination is identified as an effective way to improve electrocatalytic performance, in terms of modulating active sites and regulating structural reconstruction. This review elaborates on recent discoveries and progress of Cu-based CO2RR catalytic materials enhanced by anionic coordination control, regarding reaction paths, functional mechanisms, and roles of different non-metallic anions in catalysis. Finally, the review concludes with some personal insights and provides challenges and perspectives on the utilization of this strategy to build desirable electrocatalysts.
Collapse
Affiliation(s)
- Hanxia Chen
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing, 102206, P. R. China
| | - Pengpeng Mo
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing, 102206, P. R. China
| | - Junpeng Zhu
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing, 102206, P. R. China
| | - Xiaoxue Xu
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing, 102206, P. R. China
| | - Zhixiang Cheng
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing, 102206, P. R. China
| | - Feng Yang
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing, 102206, P. R. China
| | - Zhongfei Xu
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing, 102206, P. R. China
| | - Juzhe Liu
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing, 102206, P. R. China
| | - Lidong Wang
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing, 102206, P. R. China
| |
Collapse
|
6
|
Iqbal MA, Bilen M, Liu Y, Jabre V, Fong BC, Chakroun I, Paul S, Chen J, Wade S, Kanaan M, Harper M, Khacho M, Slack RS. The integrated stress response promotes neural stem cell survival under conditions of mitochondrial dysfunction in neurodegeneration. Aging Cell 2024; 23:e14165. [PMID: 38757355 PMCID: PMC11258489 DOI: 10.1111/acel.14165] [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: 08/29/2023] [Revised: 02/27/2024] [Accepted: 03/18/2024] [Indexed: 05/18/2024] Open
Abstract
Impaired mitochondrial function is a hallmark of aging and a major contributor to neurodegenerative diseases. We have shown that disrupted mitochondrial dynamics typically found in aging alters the fate of neural stem cells (NSCs) leading to impairments in learning and memory. At present, little is known regarding the mechanisms by which neural stem and progenitor cells survive and adapt to mitochondrial dysfunction. Using Opa1-inducible knockout as a model of aging and neurodegeneration, we identify a decline in neurogenesis due to impaired stem cell activation and progenitor proliferation, which can be rescued by the mitigation of oxidative stress through hypoxia. Through sc-RNA-seq, we identify the ATF4 pathway as a critical mechanism underlying cellular adaptation to metabolic stress. ATF4 knockdown in Opa1-deficient NSCs accelerates cell death, while the increased expression of ATF4 enhances proliferation and survival. Using a Slc7a11 mutant, an ATF4 target, we show that ATF4-mediated glutathione production plays a critical role in maintaining NSC survival and function under stress conditions. Together, we show that the activation of the integrated stress response (ISR) pathway enables NSCs to adapt to metabolic stress due to mitochondrial dysfunction and metabolic stress and may serve as a therapeutic target to enhance NSC survival and function in aging and neurodegeneration.
Collapse
Affiliation(s)
- Mohamed Ariff Iqbal
- Department of Cellular and Molecular MedicineUniversity of Ottawa Brain and Mind Research InstituteUniversity of OttawaOttawaOntarioCanada
| | - Maria Bilen
- Department of Cellular and Molecular MedicineUniversity of Ottawa Brain and Mind Research InstituteUniversity of OttawaOttawaOntarioCanada
| | - Yubing Liu
- Department of Cellular and Molecular MedicineUniversity of Ottawa Brain and Mind Research InstituteUniversity of OttawaOttawaOntarioCanada
| | - Vanessa Jabre
- Department of Cellular and Molecular MedicineUniversity of Ottawa Brain and Mind Research InstituteUniversity of OttawaOttawaOntarioCanada
| | - Bensun C. Fong
- Department of Cellular and Molecular MedicineUniversity of Ottawa Brain and Mind Research InstituteUniversity of OttawaOttawaOntarioCanada
| | - Imane Chakroun
- Department of Cellular and Molecular MedicineUniversity of Ottawa Brain and Mind Research InstituteUniversity of OttawaOttawaOntarioCanada
| | - Smitha Paul
- Department of Cellular and Molecular MedicineUniversity of Ottawa Brain and Mind Research InstituteUniversity of OttawaOttawaOntarioCanada
| | - Jingwei Chen
- Department of Cellular and Molecular MedicineUniversity of Ottawa Brain and Mind Research InstituteUniversity of OttawaOttawaOntarioCanada
| | - Steven Wade
- Department of Biochemistry, Microbiology and Immunology, Center for Neuromuscular Disease (CNMD), Ottawa Institute of Systems Biology (OISB), Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Michel Kanaan
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology (OISB), Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Mary‐Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology (OISB), Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Mireille Khacho
- Department of Cellular and Molecular MedicineUniversity of Ottawa Brain and Mind Research InstituteUniversity of OttawaOttawaOntarioCanada
- Department of Biochemistry, Microbiology and Immunology, Center for Neuromuscular Disease (CNMD), Ottawa Institute of Systems Biology (OISB), Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Ruth S. Slack
- Department of Cellular and Molecular MedicineUniversity of Ottawa Brain and Mind Research InstituteUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
7
|
Jiao J, Kang X, Yang J, Jia S, Peng Y, Liu S, Chen C, Xing X, He M, Wu H, Han B. Steering the Reaction Pathway of CO 2 Electroreduction by Tuning the Coordination Number of Copper Catalysts. J Am Chem Soc 2024; 146:15917-15925. [PMID: 38805725 DOI: 10.1021/jacs.4c02607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Cu-based catalysts are optimal for the electroreduction of CO2 to generate hydrocarbon products. However, controlling product distribution remains a challenging topic. The theoretical investigations have revealed that the coordination number (CN) of Cu considerably influences the adsorption energy of *CO intermediates, thereby affecting the reaction pathway. Cu catalysts with different CNs were fabricated by reducing CuO precursors via cyclic voltammetry (Cyc-Cu), potentiostatic electrolysis (Pot-Cu), and pulsed electrolysis (Pul-Cu), respectively. High-CN Cu catalysts predominantly generate C2+ products, while low-CN Cu favors CH4 production. For instance, over the high-CN Pot-Cu, C2+ is the main product, with the Faradaic efficiency (FE) reaching 82.5% and a partial current density (j) of 514.3 mA cm-2. Conversely, the low-CN Pul(3)-Cu favors the production of CH4, achieving the highest FECH4 value of 56.7% with a jCH4 value of 234.4 mA cm-2. In situ X-ray absorption spectroscopy and Raman spectroscopy studies further confirm the different *CO adsorptions over Cu catalysts with different CN, thereby directing the reaction pathway of the CO2RR.
Collapse
Affiliation(s)
- Jiapeng Jiao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, State Key Laboratory of Petroleum Molecular and Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- Institute of Eco-Chongming, Chongming District, Shanghai 202162, China
| | - Xinchen Kang
- Beijing National Laboratory for Molecular Sciences, CAS Laboratory of Colloid and Interface and Thermodynamics, CAS Research/Education Centre for Excellence in Molecular Sciences, Centre for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiahao Yang
- Beijing National Laboratory for Molecular Sciences, CAS Laboratory of Colloid and Interface and Thermodynamics, CAS Research/Education Centre for Excellence in Molecular Sciences, Centre for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuaiqiang Jia
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, State Key Laboratory of Petroleum Molecular and Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- Institute of Eco-Chongming, Chongming District, Shanghai 202162, China
| | - Yaguang Peng
- Beijing National Laboratory for Molecular Sciences, CAS Laboratory of Colloid and Interface and Thermodynamics, CAS Research/Education Centre for Excellence in Molecular Sciences, Centre for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Shiqiang Liu
- Beijing National Laboratory for Molecular Sciences, CAS Laboratory of Colloid and Interface and Thermodynamics, CAS Research/Education Centre for Excellence in Molecular Sciences, Centre for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Chunjun Chen
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, State Key Laboratory of Petroleum Molecular and Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- Institute of Eco-Chongming, Chongming District, Shanghai 202162, China
| | - Xueqing Xing
- Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Mingyuan He
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, State Key Laboratory of Petroleum Molecular and Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- Institute of Eco-Chongming, Chongming District, Shanghai 202162, China
| | - Haihong Wu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, State Key Laboratory of Petroleum Molecular and Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- Institute of Eco-Chongming, Chongming District, Shanghai 202162, China
| | - Buxing Han
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, State Key Laboratory of Petroleum Molecular and Process Engineering, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- Institute of Eco-Chongming, Chongming District, Shanghai 202162, China
- Beijing National Laboratory for Molecular Sciences, CAS Laboratory of Colloid and Interface and Thermodynamics, CAS Research/Education Centre for Excellence in Molecular Sciences, Centre for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
8
|
Ventura S, Duncan S, Ainge JA. Increased flexibility of CA3 memory representations following environmental enrichment. Curr Biol 2024; 34:2011-2019.e7. [PMID: 38636511 DOI: 10.1016/j.cub.2024.03.054] [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: 11/17/2023] [Revised: 02/16/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024]
Abstract
Environmental enrichment (EE) improves memory, particularly the ability to discriminate similar past experiences.1,2,3,4,5,6 The hippocampus supports this ability via pattern separation, the encoding of similar events using dissimilar memory representations.7 This is carried out in the dentate gyrus (DG) and CA3 subfields.8,9,10,11,12 Upregulation of adult neurogenesis in the DG improves memory through enhanced pattern separation.1,2,3,4,5,6,11,13,14,15,16 Adult-born granule cells (abGCs) in DG are suggested to contribute to pattern separation by driving inhibition in regions such as CA3,13,14,15,16,17,18 leading to sparser, nonoverlapping representations of similar events (although a role for abGCs in driving excitation in the hippocampus has also been reported16). Place cells in the hippocampus contribute to pattern separation by remapping to spatial and contextual alterations to the environment.19,20,21,22,23,24,25,26,27 How spatial responses in CA3 are affected by EE and input from increased numbers of abGCs in DG is, however, unknown. Here, we investigate the neural mechanisms facilitating improved memory following EE using associative recognition memory tasks that model the automatic and integrative nature of episodic memory. We find that EE-dependent improvements in difficult discriminations are related to increased neurogenesis and sparser memory representations across the hippocampus. Additionally, we report for the first time that EE changes how CA3 place cells discriminate similar contexts. CA3 place cells of enriched rats show greater spatial tuning, increased firing rates, and enhanced remapping to contextual changes. These findings point to more precise and flexible CA3 memory representations in enriched rats, which provides a putative mechanism for EE-dependent improvements in fine memory discrimination.
Collapse
Affiliation(s)
- Silvia Ventura
- School of Psychology & Neuroscience, University of St. Andrews, St. Mary's Quad, South Street, St. Andrews, Fife, Scotland KY16 9JP, UK
| | - Stephen Duncan
- School of Psychology & Neuroscience, University of St. Andrews, St. Mary's Quad, South Street, St. Andrews, Fife, Scotland KY16 9JP, UK; School of Psychological & Brain Sciences, Indiana University, 1101 E 10th Street, Bloomington, IN 47405, USA
| | - James A Ainge
- School of Psychology & Neuroscience, University of St. Andrews, St. Mary's Quad, South Street, St. Andrews, Fife, Scotland KY16 9JP, UK.
| |
Collapse
|
9
|
Jeong M, Won J, Lim KS, Jeon CY, Choe Y, Jang JH, Ha CM, Yoon JH, Lee Y, Oh YS. Comparative Anatomy of the Dentate Mossy Cells in Nonhuman Primates: Their Spatial Distributions and Axonal Projections Compared With Mouse Mossy Cells. eNeuro 2024; 11:ENEURO.0151-24.2024. [PMID: 38688719 DOI: 10.1523/eneuro.0151-24.2024] [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: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
Glutamatergic mossy cells (MCs) mediate associational and commissural connectivity, exhibiting significant heterogeneity along the septotemporal axis of the mouse dentate gyrus (DG). However, it remains unclear whether the neuronal features of MCs are conserved across mammals. This study compares the neuroanatomy of MCs in the DG of mice and monkeys. The MC marker, calretinin, distinguishes two subpopulations: septal and temporal. Dual-colored fluorescence labeling is utilized to compare the axonal projection patterns of these subpopulations. In both mice and monkeys, septal and temporal MCs project axons across the longitudinal axis of the ipsilateral DG, indicating conserved associational projections. However, unlike in mice, no MC subpopulations in monkeys make commissural projections to the contralateral DG. In monkeys, temporal MCs send associational fibers exclusively to the inner molecular layer, while septal MCs give rise to wide axonal projections spanning multiple molecular layers, akin to equivalent MC subpopulations in mice. Despite conserved septotemporal heterogeneity, interspecies differences are observed in the topological organization of septal MCs, particularly in the relative axonal density in each molecular layer along the septotemporal axis of the DG. In summary, this comparative analysis sheds light on both conserved and divergent features of MCs in the DG of mice and monkeys. These findings have implications for understanding functional differentiation along the septotemporal axis of the DG and contribute to our knowledge of the anatomical evolution of the DG circuit in mammals.
Collapse
Affiliation(s)
- Minseok Jeong
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jinyoung Won
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Republic of Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Republic of Korea
| | - Chang-Yeop Jeon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Republic of Korea
| | - Youngshik Choe
- Developmental Disorders & Rare Diseases Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Jin-Hyeok Jang
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Chang Man Ha
- Research Division and Brain Research Core Facilities, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Jong Hyuk Yoon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Yongjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Yong-Seok Oh
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| |
Collapse
|
10
|
Lazarov O, Gupta M, Kumar P, Morrissey Z, Phan T. Memory circuits in dementia: The engram, hippocampal neurogenesis and Alzheimer's disease. Prog Neurobiol 2024; 236:102601. [PMID: 38570083 PMCID: PMC11221328 DOI: 10.1016/j.pneurobio.2024.102601] [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: 08/10/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Here, we provide an in-depth consideration of our current understanding of engrams, spanning from molecular to network levels, and hippocampal neurogenesis, in health and Alzheimer's disease (AD). This review highlights novel findings in these emerging research fields and future research directions for novel therapeutic avenues for memory failure in dementia. Engrams, memory in AD, and hippocampal neurogenesis have each been extensively studied. The integration of these topics, however, has been relatively less deliberated, and is the focus of this review. We primarily focus on the dentate gyrus (DG) of the hippocampus, which is a key area of episodic memory formation. Episodic memory is significantly impaired in AD, and is also the site of adult hippocampal neurogenesis. Advancements in technology, especially opto- and chemogenetics, have made sophisticated manipulations of engram cells possible. Furthermore, innovative methods have emerged for monitoring neurons, even specific neuronal populations, in vivo while animals engage in tasks, such as calcium imaging. In vivo calcium imaging contributes to a more comprehensive understanding of engram cells. Critically, studies of the engram in the DG using these technologies have shown the important contribution of hippocampal neurogenesis for memory in both health and AD. Together, the discussion of these topics provides a holistic perspective that motivates questions for future research.
Collapse
Affiliation(s)
- Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Pavan Kumar
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zachery Morrissey
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Trongha Phan
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
11
|
Bickle JG, Li Y, Millette A, Dixon R, Wu S, Arias EC, Luna VM, Anacker C. 5-HT 1A Receptors on Dentate Gyrus Granule Cells Confer Stress Resilience. Biol Psychiatry 2024; 95:800-809. [PMID: 37863245 PMCID: PMC10978305 DOI: 10.1016/j.biopsych.2023.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/19/2023] [Accepted: 10/09/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Hyperactivity of granule cells in the ventral dentate gyrus (vDG) promotes vulnerability to chronic stress. However, which receptors in the vDG could be targeted to inhibit this hyperactivity and confer stress resilience is not known. The serotonin 1A receptor (5-HT1AR) is a Gi protein-coupled inhibitory receptor that has been implicated in stress adaptation, anxiety, depression, and antidepressant responses. 5-HT1ARs are highly expressed in the DG, but their potential to promote stress resilience by regulating granule cell activity has never been examined. METHODS We exposed male and female mice expressing 5-HT1ARs only in DG granule cells to 10 days of chronic social defeat stress (CSDS) and treated them with the 5-HT1AR agonist 8-OH-DPAT every day 30 minutes before each defeat throughout the CSDS paradigm. We then used whole-cell current clamp recordings, immunohistochemistry for the immediate early gene cFos, corticosterone immunoassays, and behavioral testing to determine how activating 5-HT1ARs on granule cells affects DG activity, neuroendocrine stress responses, and avoidance behavior. RESULTS We found that activating 5-HT1ARs hyperpolarized DG granule cells and reduced cFos+ granule cells in the vDG following CSDS, indicating that 5-HT1AR activation rescued stress-induced vDG hyperactivity. Moreover, 5-HT1AR activation dampened corticosterone responses to CSDS and prevented the development of stress-induced avoidance in the social interaction test and in the open field test. CONCLUSIONS Our findings show that activating 5-HT1ARs on DG granule cells can prevent stress-induced neuronal hyperactivity of the vDG and confer resilience to chronic stress.
Collapse
Affiliation(s)
- John Gregory Bickle
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Yifei Li
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Amira Millette
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Rushell Dixon
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Serena Wu
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Elena Carazo Arias
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York
| | - Victor Mari Luna
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Christoph Anacker
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University and Research Foundation for Mental Hygiene, Inc., New York State Psychiatric Institute, New York, New York; Columbia University Institute for Developmental Sciences, Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Columbia University Stem Cell Initiative, Columbia University Irving Medical Center, New York, New York.
| |
Collapse
|
12
|
Santiago AN, Castello-Saval J, Nguyen P, Chung HM, Luna VM, Hen R, Chang WL. Effects of electroconvulsive shock on the function, circuitry, and transcriptome of dentate gyrus granule neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.583011. [PMID: 38496461 PMCID: PMC10942314 DOI: 10.1101/2024.03.01.583011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Therapeutic use of electroconvulsive shock (ECS) is 75% effective for the remission of treatment-resistant depression. Like other more common forms of antidepressant treatment such as fluoxetine, ECS has been shown to increase neurogenesis in the hippocampal dentate gyrus of rodent models. Yet the question of how ECS-induced neurogenesis supports improvement of depressive symptoms remains unknown. Here, we show that ECS-induced neurogenesis is necessary to improve depressive-like behavior of mice exposed to chronic corticosterone (Cort). We then use slice electrophysiology to show that optogenetic stimulation of adult-born neurons produces a greater hyperpolarization in mature granule neurons after ECS vs Sham treatment. We identify that this hyperpolarization requires the activation of metabotropic glutamate receptor 2 (mGluR2). Consistent with this finding, we observe reduced expression of the immediate early gene cFos in the granule cell layer of ECS vs Sham subjects. We then show that mGluR2 knockdown specifically in ventral granule neurons blunts the antidepressant-like behavioral effects of ECS. Using single nucleus RNA sequencing, we reveal major transcriptomic shifts in granule neurons after treatment with ECS+Cort or fluoxetine+Cort vs Cort alone. We identify a population of immature cells which has greater representation in both ECS+Cort and fluoxetine+Cort treated samples vs Cort alone. We also find global differences in ECS-vs fluoxetine-induced transcriptomic shifts. Together, these findings highlight a critical role for immature granule cells and mGluR2 signaling in the antidepressant action of ECS.
Collapse
|
13
|
Santiago RMM, Lopes-Dos-Santos V, Aery Jones EA, Huang Y, Dupret D, Tort ABL. Waveform-based classification of dentate spikes. Sci Rep 2024; 14:2989. [PMID: 38316828 PMCID: PMC10844627 DOI: 10.1038/s41598-024-53075-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/27/2024] [Indexed: 02/07/2024] Open
Abstract
Synchronous excitatory discharges from the entorhinal cortex (EC) to the dentate gyrus (DG) generate fast and prominent patterns in the hilar local field potential (LFP), called dentate spikes (DSs). As sharp-wave ripples in CA1, DSs are more likely to occur in quiet behavioral states, when memory consolidation is thought to take place. However, their functions in mnemonic processes are yet to be elucidated. The classification of DSs into types 1 or 2 is determined by their origin in the lateral or medial EC, as revealed by current source density (CSD) analysis, which requires recordings from linear probes with multiple electrodes spanning the DG layers. To allow the investigation of the functional role of each DS type in recordings obtained from single electrodes and tetrodes, which are abundant in the field, we developed an unsupervised method using Gaussian mixture models to classify such events based on their waveforms. Our classification approach achieved high accuracies (> 80%) when validated in 8 mice with DG laminar profiles. The average CSDs, waveforms, rates, and widths of the DS types obtained through our method closely resembled those derived from the CSD-based classification. As an example of application, we used the technique to analyze single-electrode LFPs from apolipoprotein (apo) E3 and apoE4 knock-in mice. We observed that the latter group, which is a model for Alzheimer's disease, exhibited wider DSs of both types from a young age, with a larger effect size for DS type 2, likely reflecting early pathophysiological alterations in the EC-DG network, such as hyperactivity. In addition to the applicability of the method in expanding the study of DS types, our results show that their waveforms carry information about their origins, suggesting different underlying network dynamics and roles in memory processing.
Collapse
Affiliation(s)
- Rodrigo M M Santiago
- Computational Neurophysiology Lab, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Brazil.
| | - Vítor Lopes-Dos-Santos
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Emily A Aery Jones
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, 94158, USA
| | - David Dupret
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Adriano B L Tort
- Computational Neurophysiology Lab, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Brazil
| |
Collapse
|
14
|
Fuchigami T, Itokazu Y, Yu RK. Ganglioside GD3 regulates neural stem cell quiescence and controls postnatal neurogenesis. Glia 2024; 72:167-183. [PMID: 37667994 PMCID: PMC10840680 DOI: 10.1002/glia.24468] [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: 03/16/2023] [Revised: 08/14/2023] [Accepted: 08/26/2023] [Indexed: 09/06/2023]
Abstract
The postnatal neural stem cell (NSC) pool hosts quiescent and activated radial glia-like NSCs contributing to neurogenesis throughout adulthood. However, the underlying regulatory mechanism during the transition from quiescent NSCs to activated NSCs in the postnatal NSC niche is not fully understood. Lipid metabolism and lipid composition play important roles in regulating NSC fate determination. Biological lipid membranes define the individual cellular shape and help maintain cellular organization and are highly heterogeneous in structure and there exist diverse microdomains (also known as lipid rafts), which are enriched with sugar molecules, such as glycosphingolipids. An often overlooked but key aspect is that the functional activities of proteins and genes are highly dependent on their molecular environments. We previously reported that ganglioside GD3 is the predominant species in NSCs and that the reduced postnatal NSC pools are observed in global GD3-synthase knockout (GD3S-KO) mouse brains. The specific roles of GD3 in determining the stage and cell-lineage determination of NSCs remain unclear, since global GD3S-KO mice cannot distinguish if GD3 regulates postnatal neurogenesis or developmental impacts. Here, we show that inducible GD3 deletion in postnatal radial glia-like NSCs promotes NSC activation, resulting in the loss of the long-term maintenance of the adult NSC pools. The reduced neurogenesis in the subventricular zone (SVZ) and the dentate gyrus (DG) of GD3S-conditional-knockout mice led to the impaired olfactory and memory functions. Thus, our results provide convincing evidence that postnatal GD3 maintains the quiescent state of radial glia-like NSCs in the adult NSC niche.
Collapse
Affiliation(s)
- Takahiro Fuchigami
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yutaka Itokazu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Robert K. Yu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
15
|
Chang WL, Hen R. Adult Neurogenesis, Context Encoding, and Pattern Separation: A Pathway for Treating Overgeneralization. ADVANCES IN NEUROBIOLOGY 2024; 38:163-193. [PMID: 39008016 DOI: 10.1007/978-3-031-62983-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In mammals, the subgranular zone of the dentate gyrus is one of two brain regions (with the subventricular zone of the olfactory bulb) that continues to generate new neurons throughout adulthood, a phenomenon known as adult hippocampal neurogenesis (AHN) (Eriksson et al., Nat Med 4:1313-1317, 1998; García-Verdugo et al., J Neurobiol 36:234-248, 1998). The integration of these new neurons into the dentate gyrus (DG) has implications for memory encoding, with unique firing and wiring properties of immature neurons that affect how the hippocampal network encodes and stores attributes of memory. In this chapter, we will describe the process of AHN and properties of adult-born cells as they integrate into the hippocampal circuit and mature. Then, we will discuss some methodological considerations before we review evidence for the role of AHN in two major processes supporting memory that are performed by the DG. First, we will discuss encoding of contextual information for episodic memories and how this is facilitated by AHN. Second, will discuss pattern separation, a major role of the DG that reduces interference for the formation of new memories. Finally, we will review clinical and translational considerations, suggesting that stimulation of AHN may help decrease overgeneralization-a common endophenotype of mood, anxiety, trauma-related, and age-related disorders.
Collapse
Affiliation(s)
- Wei-Li Chang
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY, USA
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Rene Hen
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY, USA.
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
16
|
Tuncdemir SN, Grosmark AD, Chung H, Luna VM, Lacefield CO, Losonczy A, Hen R. Adult-born granule cells facilitate remapping of spatial and non-spatial representations in the dentate gyrus. Neuron 2023; 111:4024-4039.e7. [PMID: 37820723 PMCID: PMC10841867 DOI: 10.1016/j.neuron.2023.09.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/10/2023] [Accepted: 09/13/2023] [Indexed: 10/13/2023]
Abstract
Adult-born granule cells (abGCs) have been implicated in memory discrimination through a neural computation known as pattern separation. Here, using in vivo Ca2+ imaging, we examined how chronic ablation or acute chemogenetic silencing of abGCs affects the activity of mature granule cells (mGCs). In both cases, we observed altered remapping of mGCs. Rather than broadly modulating the activity of all mGCs, abGCs promote the remapping of place cells' firing fields while increasing rate remapping of mGCs that represent sensory cues. In turn, these remapping deficits are associated with behavioral impairments in animals' ability to correctly identify new goal locations. Thus, abGCs facilitate pattern separation through the formation of non-overlapping representations for identical sensory cues encountered in different locations. In the absence of abGCs, the dentate gyrus shifts to a state that is dominated by cue information, a situation that is consistent with the overgeneralization often observed in anxiety or age-related disorders.
Collapse
Affiliation(s)
- Sebnem N Tuncdemir
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY 10032, USA; Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Andres D Grosmark
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Hannah Chung
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Victor M Luna
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY 10032, USA; Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Clay O Lacefield
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY 10032, USA; Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Attila Losonczy
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Rene Hen
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY 10032, USA; Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA.
| |
Collapse
|
17
|
Santiago RM, Lopes-dos-Santos V, Jones EAA, Huang Y, Dupret D, Tort AB. Waveform-based classification of dentate spikes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563826. [PMID: 37961150 PMCID: PMC10634814 DOI: 10.1101/2023.10.24.563826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Synchronous excitatory discharges from the entorhinal cortex (EC) to the dentate gyrus (DG) generate fast and prominent patterns in the hilar local field potential (LFP), called dentate spikes (DSs). As sharp-wave ripples in CA1, DSs are more likely to occur in quiet behavioral states, when memory consolidation is thought to take place. However, their functions in mnemonic processes are yet to be elucidated. The classification of DSs into types 1 or 2 is determined by their origin in the lateral or medial EC, as revealed by current source density (CSD) analysis, which requires recordings from linear probes with multiple electrodes spanning the DG layers. To allow the investigation of the functional role of each DS type in recordings obtained from single electrodes and tetrodes, which are abundant in the field, we developed an unsupervised method using Gaussian mixture models to classify such events based on their waveforms. Our classification approach achieved high accuracies (> 80%) when validated in 8 mice with DG laminar profiles. The average CSDs, waveforms, rates, and widths of the DS types obtained through our method closely resembled those derived from the CSD-based classification. As an example of application, we used the technique to analyze single-electrode LFPs from apolipoprotein (apo) E3 and apoE4 knock-in mice. We observed that the latter group, which is a model for Alzheimer's disease, exhibited wider DSs of both types from a young age, with a larger effect size for DS type 2, likely reflecting early pathophysiological alterations in the EC-DG network, such as hyperactivity. In addition to the applicability of the method in expanding the study of DS types, our results show that their waveforms carry information about their origins, suggesting different underlying network dynamics and roles in memory processing.
Collapse
Affiliation(s)
- Rodrigo M.M. Santiago
- Computational Neurophysiology Lab, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Brazil
| | - Vítor Lopes-dos-Santos
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Emily A. Aery Jones
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - David Dupret
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Adriano B.L. Tort
- Computational Neurophysiology Lab, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Brazil
| |
Collapse
|
18
|
Berdugo‐Vega G, Dhingra S, Calegari F. Sharpening the blades of the dentate gyrus: how adult-born neurons differentially modulate diverse aspects of hippocampal learning and memory. EMBO J 2023; 42:e113524. [PMID: 37743770 PMCID: PMC11059975 DOI: 10.15252/embj.2023113524] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/19/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
For decades, the mammalian hippocampus has been the focus of cellular, anatomical, behavioral, and computational studies aimed at understanding the fundamental mechanisms underlying cognition. Long recognized as the brain's seat for learning and memory, a wealth of knowledge has been accumulated on how the hippocampus processes sensory input, builds complex associations between objects, events, and space, and stores this information in the form of memories to be retrieved later in life. However, despite major efforts, our understanding of hippocampal cognitive function remains fragmentary, and models trying to explain it are continually revisited. Here, we review the literature across all above-mentioned domains and offer a new perspective by bringing attention to the most distinctive, and generally neglected, feature of the mammalian hippocampal formation, namely, the structural separability of the two blades of the dentate gyrus into "supra-pyramidal" and "infra-pyramidal". Next, we discuss recent reports supporting differential effects of adult neurogenesis in the regulation of mature granule cell activity in these two blades. We propose a model for how differences in connectivity and adult neurogenesis in the two blades can potentially provide a substrate for subtly different cognitive functions.
Collapse
Affiliation(s)
- Gabriel Berdugo‐Vega
- CRTD‐Center for Regenerative Therapies DresdenTechnische Universität DresdenDresdenGermany
- Present address:
Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale Lausanne (EPFL)LausanneSwitzerland
| | - Shonali Dhingra
- CRTD‐Center for Regenerative Therapies DresdenTechnische Universität DresdenDresdenGermany
| | - Federico Calegari
- CRTD‐Center for Regenerative Therapies DresdenTechnische Universität DresdenDresdenGermany
| |
Collapse
|
19
|
Ji Q, Yang Y, Xiong Y, Zhang YJ, Jiang J, Zhou LP, Du XH, Wang CX, Zhu ZR. Blockade of adenosine A 2A receptors reverses early spatial memory defects in the APP/PS1 mouse model of Alzheimer's disease by promoting synaptic plasticity of adult-born granule cells. Alzheimers Res Ther 2023; 15:187. [PMID: 37899431 PMCID: PMC10614339 DOI: 10.1186/s13195-023-01337-z] [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: 04/17/2023] [Accepted: 10/17/2023] [Indexed: 10/31/2023]
Abstract
BACKGROUND The over-activation of adenosine A2A receptors (A2AR) is closely implicated in cognitive impairments of Alzheimer's disease (AD). Growing evidence shows that A2AR blockade possesses neuroprotective effects on AD. Spatial navigation impairment is an early manifestation of cognitive deficits in AD. However, whether A2AR blockade can prevent early impairments in spatial cognitive function and the underlying mechanism is still unclear. METHODS A transgenic APP/PS1 mouse model of AD amyloidosis was used in this study. Behavioral tests were conducted to observe the protective effects of A2AR blockade on early spatial memory deficits in 4-month old APP/PS1 mice. To investigate the underlying synaptic mechanism of the protective effects of A2AR blockade, we further examined long-term potentiation (LTP) and network excitation/inhibition balance of dentate gyrus (DG) region, which is relevant to unique synaptic functions of immature adult-born granule cells (abGCs). Subsequently, the protective effects of A2AR blockade on dendritic morphology and synaptic plasticity of 6-week-old abGCs was investigated using retrovirus infection and electrophysiological recordings. The molecular mechanisms underlying neuroprotective properties of A2AR blockade on the synaptic plasticity of abGCs were further explored using molecular biology methods. RESULTS APP/PS1 mice displayed DG-dependent spatial memory deficits at an early stage. Additionally, impaired LTP and an imbalance in network excitation/inhibition were observed in the DG region of APP/PS1 mice, indicating synaptic structural and functional abnormalities of abGCs. A2AR was found to be upregulated in the hippocampus of the APP/PS1 mouse model of AD. Treatment with the selective A2AR antagonist SCH58261 for three weeks significantly ameliorated spatial memory deficits in APP/PS1 mice and markedly restored LTP and network excitation/inhibition balance in the DG region. Moreover, SCH58261 treatment restored dendritic morphology complexity and enhanced synaptic plasticity of abGCs in APP/PS1 mice. Furthermore, SCH58261 treatment alleviated the impairment of synaptic plasticity in abGCs. It achieved this by remodeling the subunit composition of NMDA receptors and increasing the proportion of NR2B receptors in abGCs of APP/PS1 mice. CONCLUSIONS Blockade of A2AR improves early spatial memory deficits in APP/PS1 mice, possibly by reversing synaptic defects of abGCs. This finding suggests that A2AR blockade could be a potential therapy for AD.
Collapse
Affiliation(s)
- Qi Ji
- Department of Medical Psychology, Army Medical University, Gaotanyan Street 30, Chongqing, 400038, PR China
- College of Basic Medicine, Army Medical University, Chongqing, 400038, China
| | - Yang Yang
- Department of Medical Psychology, Army Medical University, Gaotanyan Street 30, Chongqing, 400038, PR China
- Department of Neurosurgery, The 904Th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, 214044, Jiangsu, China
| | - Yun Xiong
- Department of Medical Psychology, Army Medical University, Gaotanyan Street 30, Chongqing, 400038, PR China
- College of Basic Medicine, Army Medical University, Chongqing, 400038, China
| | - Ying-Jie Zhang
- Department of Medical Psychology, Army Medical University, Gaotanyan Street 30, Chongqing, 400038, PR China
| | - Jun Jiang
- Department of Medical Psychology, Army Medical University, Gaotanyan Street 30, Chongqing, 400038, PR China
| | - Li-Ping Zhou
- Department of Medical Psychology, Army Medical University, Gaotanyan Street 30, Chongqing, 400038, PR China
| | - Xiao-Hui Du
- Department of Medical Psychology, Army Medical University, Gaotanyan Street 30, Chongqing, 400038, PR China
| | - Chun-Xiang Wang
- Department of Medical Psychology, Army Medical University, Gaotanyan Street 30, Chongqing, 400038, PR China
| | - Zhi-Ru Zhu
- Department of Medical Psychology, Army Medical University, Gaotanyan Street 30, Chongqing, 400038, PR China.
| |
Collapse
|
20
|
Lee SY, Kim J, Bak G, Lee E, Kim D, Yoo S, Kim J, Yun H, Hwang YJ. Probing Cation Effects on *CO Intermediates from Electroreduction of CO 2 through Operando Raman Spectroscopy. J Am Chem Soc 2023; 145:23068-23075. [PMID: 37807716 DOI: 10.1021/jacs.3c05799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Cations in an electrolyte modulate microenvironments near the catalyst surface and affect product distribution from an electrochemical CO2 reduction reaction, and thus, their interaction with intermediate states has been tried to be probed. Herein, we directly observed the cation effect on *CO intermediates on the Cu(OH)2-derived catalyst in real time through operando surface-enhanced Raman spectroscopy at high overpotentials (-1.0 VRHE). Atop *CO peaks are composed of low-frequency binding *CO (*COLFB) and high-frequency binding *CO (*COHFB) because of their adsorption sites. These two *CO intermediates are found to have different sensitivities to the cation-induced field, and each *CO is proposed to be suitably stabilized for efficient C-C coupling. The proportions between *COHFB and *COLFB are dependent on the type of alkali cations, and the increases in the *COHFB ratio have a high correlation with selective C2H4 production under K+ and Cs+, indicating that *COHFB is the dominant and fast active species. In addition, as the hydrated cation size decreases, *COLFB is more sensitively red-shifted than *COHFB, which promotes C-C coupling and suppresses C1 products. Through time-resolved operando measurements, dynamic changes between the two *CO species are observed, showing the rapid initial adsorption of *COHFB and subsequently reaching a steady ratio between *COLFB and *COHFB.
Collapse
Affiliation(s)
- Si Young Lee
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| | - Jimin Kim
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| | - Gwangsu Bak
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Eunchong Lee
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Dayeon Kim
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Suhwan Yoo
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jiwon Kim
- Department of Chemical and Biomolecular Engineering, Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Republic of Korea
- Clean Energy Research Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Hyewon Yun
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| | - Yun Jeong Hwang
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| |
Collapse
|
21
|
Schoenfeld TJ, Rhee D, Smith JA, Padmanaban V, Brockett AT, Jacobs HN, Cameron HA. Rewarded Maze Training Increases Approach Behavior in Rats Through Neurogenesis-Dependent Growth of Ventral Hippocampus-Prelimbic Circuits. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:725-733. [PMID: 37881563 PMCID: PMC10593943 DOI: 10.1016/j.bpsgos.2023.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 10/27/2023] Open
Abstract
Background Learning complex navigation routes increases hippocampal volume in humans, but it is not clear whether this growth impacts behaviors outside the learning situation or what cellular mechanisms are involved. Methods We trained rats with pharmacogenetic suppression of adult neurogenesis and littermate controls in 3 mazes over 3 weeks and tested novelty approach behavior several days after maze exposure. We then measured hippocampus and prelimbic cortex volumes using magnetic resonance imaging and assessed neuronal and astrocyte morphology. Finally, we investigated the activation and behavioral role of the ventral CA1 (vCA1)-to-prelimbic pathway using immediate-early genes and DREADDs (designer receptors exclusively activated by designer drugs). Results Maze training led to volume increase of both the vCA1 region of the hippocampus and the prelimbic region of the neocortex compared with rats that followed fixed paths. Growth was also apparent in individual neurons and astrocytes in these 2 regions, and behavioral testing showed increased novelty approach in maze-trained rats in 2 different tests. Suppressing adult neurogenesis prevented the effects on structure and approach behavior after maze training without affecting maze learning itself. The vCA1 neurons projecting to the prelimbic area were more activated by novelty in maze-trained animals, and suppression of this pathway decreased approach behavior. Conclusions Rewarded navigational learning experiences induce volumetric and morphologic growth in the vCA1 and prelimbic cortex and enhance activation of the circuit connecting these 2 regions. Both the structural and behavioral effects of maze training require ongoing adult neurogenesis, suggesting a role for new neurons in experience-driven increases in novelty exploration.
Collapse
Affiliation(s)
- Timothy J. Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, Tennessee
| | - Diane Rhee
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Jesse A. Smith
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Varun Padmanaban
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Adam T. Brockett
- Department of Psychology and Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Hannah N. Jacobs
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Heather A. Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
22
|
Mugnaini M, Trinchero MF, Schinder AF, Piatti VC, Kropff E. Unique potential of immature adult-born neurons for the remodeling of CA3 spatial maps. Cell Rep 2023; 42:113086. [PMID: 37676761 PMCID: PMC11342238 DOI: 10.1016/j.celrep.2023.113086] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/30/2023] [Accepted: 08/15/2023] [Indexed: 09/09/2023] Open
Abstract
Mammalian hippocampal circuits undergo extensive remodeling through adult neurogenesis. While this process has been widely studied, the specific contribution of adult-born granule cells (aGCs) to spatial operations in the hippocampus remains unknown. Here, we show that optogenetic activation of 4-week-old (young) aGCs in free-foraging mice produces a non-reversible reconfiguration of spatial maps in proximal CA3 while rarely evoking neural activity. Stimulation of the same neuronal cohort on subsequent days recruits CA3 neurons with increased efficacy but fails to induce further remapping. In contrast, stimulation of 8-week-old (mature) aGCs can reliably activate CA3 cells but produces no alterations in spatial maps. Our results reveal a unique role of young aGCs in remodeling CA3 representations, a potential that can be depleted and is lost with maturation. This ability could contribute to generate orthogonalized downstream codes supporting pattern separation.
Collapse
Affiliation(s)
- Matías Mugnaini
- Department of Physiology, Molecular and Cellular Biology Dr. Héctor Maldonado, Faculty of Exact and Natural Science, University of Buenos Aires, Buenos Aires C1428EGA, Argentina; Laboratory of Physiology and Algorithms of the Brain, Leloir Institute (IIBBA-CONICET), Buenos Aires C1405BWE, Argentina
| | - Mariela F Trinchero
- Laboratory of Neuronal Plasticity, Leloir Institute (IIBBA-CONICET), Buenos Aires C1405BWE, Argentina
| | - Alejandro F Schinder
- Laboratory of Neuronal Plasticity, Leloir Institute (IIBBA-CONICET), Buenos Aires C1405BWE, Argentina.
| | - Verónica C Piatti
- Laboratory of Neuronal Plasticity, Leloir Institute (IIBBA-CONICET), Buenos Aires C1405BWE, Argentina.
| | - Emilio Kropff
- Laboratory of Physiology and Algorithms of the Brain, Leloir Institute (IIBBA-CONICET), Buenos Aires C1405BWE, Argentina.
| |
Collapse
|
23
|
Xu YJ, Dai SK, Duan CH, Zhang ZH, Liu PP, Liu C, Du HZ, Lu XK, Hu S, Li L, Teng ZQ, Liu CM. ASH2L regulates postnatal neurogenesis through Onecut2-mediated inhibition of TGF-β signaling pathway. Cell Death Differ 2023; 30:1943-1956. [PMID: 37433907 PMCID: PMC10406892 DOI: 10.1038/s41418-023-01189-y] [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: 08/20/2021] [Revised: 06/18/2023] [Accepted: 06/29/2023] [Indexed: 07/13/2023] Open
Abstract
The ability of neural stem/progenitor cells (NSPCs) to proliferate and differentiate is required through different stages of neurogenesis. Disturbance in the regulation of neurogenesis causes many neurological diseases, such as intellectual disability, autism, and schizophrenia. However, the intrinsic mechanisms of this regulation in neurogenesis remain poorly understood. Here, we report that Ash2l (Absent, small or homeotic discs-like 2), one core component of a multimeric histone methyltransferase complex, is essential for NSPC fate determination during postnatal neurogenesis. Deletion of Ash2l in NSPCs impairs their capacity for proliferation and differentiation, leading to simplified dendritic arbors in adult-born hippocampal neurons and deficits in cognitive abilities. RNA sequencing data reveal that Ash2l primarily regulates cell fate specification and neuron commitment. Furthermore, we identified Onecut2, a major downstream target of ASH2L characterized by bivalent histone modifications, and demonstrated that constitutive expression of Onecut2 restores defective proliferation and differentiation of NSPCs in adult Ash2l-deficient mice. Importantly, we identified that Onecut2 modulates TGF-β signaling in NSPCs and that treatment with a TGF-β inhibitor rectifies the phenotype of Ash2l-deficient NSPCs. Collectively, our findings reveal the ASH2L-Onecut2-TGF-β signaling axis that mediates postnatal neurogenesis to maintain proper forebrain function.
Collapse
Affiliation(s)
- Ya-Jie Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Shang-Kun Dai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Chun-Hui Duan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Zi-Han Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Pei-Pei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Cong Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Hong-Zhen Du
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Xu-Kun Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, 215000, Suzhou, China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Zhao-Qian Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China.
- Savaid Medical School, University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
24
|
Borzello M, Ramirez S, Treves A, Lee I, Scharfman H, Stark C, Knierim JJ, Rangel LM. Assessments of dentate gyrus function: discoveries and debates. Nat Rev Neurosci 2023; 24:502-517. [PMID: 37316588 PMCID: PMC10529488 DOI: 10.1038/s41583-023-00710-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/16/2023]
Abstract
There has been considerable speculation regarding the function of the dentate gyrus (DG) - a subregion of the mammalian hippocampus - in learning and memory. In this Perspective article, we compare leading theories of DG function. We note that these theories all critically rely on the generation of distinct patterns of activity in the region to signal differences between experiences and to reduce interference between memories. However, these theories are divided by the roles they attribute to the DG during learning and recall and by the contributions they ascribe to specific inputs or cell types within the DG. These differences influence the information that the DG is thought to impart to downstream structures. We work towards a holistic view of the role of DG in learning and memory by first developing three critical questions to foster a dialogue between the leading theories. We then evaluate the extent to which previous studies address our questions, highlight remaining areas of conflict, and suggest future experiments to bridge these theories.
Collapse
Affiliation(s)
- Mia Borzello
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA
| | - Steve Ramirez
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | | | - Inah Lee
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, South Korea
| | - Helen Scharfman
- Departments of Child and Adolescent Psychiatry, Neuroscience and Physiology and Psychiatry and the Neuroscience Institute, New York University Langone Health, New York, NY, USA
- The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Craig Stark
- Department of Neurobiology and Behaviour, University of California, Irvine, Irvine, CA, USA
| | - James J Knierim
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Lara M Rangel
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
25
|
Biel-Nielsen TL, Hatton TA, Villadsen SNB, Jakobsen JS, Bonde JL, Spormann AM, Fosbøl PL. Electrochemistry-Based CO 2 Removal Technologies. CHEMSUSCHEM 2023; 16:e202202345. [PMID: 36861656 DOI: 10.1002/cssc.202202345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/16/2023] [Indexed: 06/10/2023]
Abstract
Unprecedented increase in atmospheric CO2 levels calls for efficient, sustainable, and cost-effective technologies for CO2 removal, including both capture and conversion approaches. Current CO2 abatement is largely based on energy-intensive thermal processes with a high degree of inflexibility. In this Perspective, it is argued that future CO2 technologies will follow the general societal trend towards electrified systems. This transition is largely promoted by decreasing electricity prices, continuous expansion of renewable energy infrastructure, and breakthroughs in carbon electrotechnologies, such as electrochemically modulated amine regeneration, redox-active quinones and other species, and microbial electrosynthesis. In addition, new initiatives make electrochemical carbon capture an integrated part of Power-to-X applications, for example, by linking it to H2 production. Selected electrochemical technologies crucial for a future sustainable society are reviewed. However, significant further development of these technologies within the next decade is needed, to meet the ambitious climate goals.
Collapse
Affiliation(s)
- Tessa Lund Biel-Nielsen
- Department of Chemical and Biochemical Engineering, Technical University of Denmark, Søltofts Plads, Building 229, DK-2800, Kgs. Lyngby, Denmark
| | - T Alan Hatton
- Department of Chemical Engineering, Massachusetts Institute of Technology, 02139, Cambridge, Massachusetts, USA
| | - Sebastian N B Villadsen
- Department of Chemical and Biochemical Engineering, Technical University of Denmark, Søltofts Plads, Building 229, DK-2800, Kgs. Lyngby, Denmark
| | | | - Jacob L Bonde
- ESTECH A/S, Sverigesvej 13, DK-5700, Svendborg, Denmark
| | - Alfred M Spormann
- Departments of Chemical Engineering and of Civil and Environmental Engineering, Stanford University, 94305, Stanford, California, USA
- Novo Nordisk Foundation CO2 Research Center, Aarhus University, Gustav Wieds Vej 10C, Building 3135, 214, DK-8000, Aarhus, Denmark
| | - Philip L Fosbøl
- Department of Chemical and Biochemical Engineering, Technical University of Denmark, Søltofts Plads, Building 229, DK-2800, Kgs. Lyngby, Denmark
| |
Collapse
|
26
|
Chen L, Xu Y, Cheng H, Li Z, Lai N, Li M, Ruan Y, Zheng Y, Fei F, Xu C, Ma J, Wang S, Gu Y, Han F, Chen Z, Wang Y. Adult-born neurons in critical period maintain hippocampal seizures via local aberrant excitatory circuits. Signal Transduct Target Ther 2023; 8:225. [PMID: 37280192 DOI: 10.1038/s41392-023-01433-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 03/15/2023] [Accepted: 04/06/2023] [Indexed: 06/08/2023] Open
Abstract
Temporal lobe epilepsy (TLE), one common type of medically refractory epilepsy, is accompanied with altered adult-born dentate granule cells (abDGCs). However, the causal role of abDGCs in recurrent seizures of TLE is not fully understood. Here, taking advantage of optogenetic and chemogenetic tools to selectively manipulate abDGCs in a reversible manner, combined with Ca2+ fiber photometry, trans-synaptic viral tracing, in vivo/vitro electrophysiology approaches, we aimed to test the role of abDGCs born at different period of epileptogenic insult in later recurrent seizures in mouse TLE models. We found that abDGCs were functionally inhibited during recurrent seizures. Optogenetic activation of abDGCs significantly extended, while inhibition curtailed, the seizure duration. This seizure-modulating effect was attributed to specific abDGCs born at a critical early phase after kindled status, which experienced specific type of circuit re-organization. Further, abDGCs extended seizure duration via local excitatory circuit with early-born granule cells (ebDGCs). Repeated modulation of "abDGC-ebDGC" circuit may easily induce a change of synaptic plasticity, and achieve long-term anti-seizure effects in both kindling and kainic acid-induced TLE models. Together, we demonstrate that abDGCs born at a critical period of epileptogenic insult maintain seizure duration via local aberrant excitatory circuits, and inactivation of these aberrant circuits can long-termly alleviate severity of seizures. This provides a deeper and more comprehensive understanding of the potential pathological changes of abDGCs circuit and may be helpful for the precise treatment in TLE.
Collapse
Affiliation(s)
- Liying Chen
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yingwei Xu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Heming Cheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhongxia Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Rehabilitation Medical Center Department, The Third Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Nanxi Lai
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Menghan Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yeping Ruan
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yang Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fan Fei
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiao Ma
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuang Wang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yan Gu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yi Wang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Zhejiang Rehabilitation Medical Center Department, The Third Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
27
|
Yun S, Soler I, Tran FH, Haas HA, Shi R, Bancroft GL, Suarez M, de Santis CR, Reynolds RP, Eisch AJ. Behavioral pattern separation and cognitive flexibility are enhanced in a mouse model of increased lateral entorhinal cortex-dentate gyrus circuit activity. Front Behav Neurosci 2023; 17:1151877. [PMID: 37324519 PMCID: PMC10267474 DOI: 10.3389/fnbeh.2023.1151877] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/26/2023] [Indexed: 06/17/2023] Open
Abstract
Behavioral pattern separation and cognitive flexibility are essential cognitive abilities that are disrupted in many brain disorders. A better understanding of the neural circuitry involved in these abilities will open paths to treatment. In humans and mice, discrimination and adaptation rely on the integrity of the hippocampal dentate gyrus (DG) which receives glutamatergic input from the entorhinal cortex (EC), including the lateral EC (LEC). An inducible increase of EC-DG circuit activity improves simple hippocampal-dependent associative learning and increases DG neurogenesis. Here, we asked if the activity of LEC fan cells that directly project to the DG (LEC → DG neurons) regulates the relatively more complex hippocampal-dependent abilities of behavioral pattern separation or cognitive flexibility. C57BL/6J male mice received bilateral LEC infusions of a virus expressing shRNA TRIP8b, an auxiliary protein of an HCN channel or a control virus (SCR shRNA). Prior work shows that 4 weeks post-surgery, TRIP8b mice have more DG neurogenesis and greater activity of LEC → DG neurons compared to SCR shRNA mice. Here, 4 weeks post-surgery, the mice underwent testing for behavioral pattern separation and reversal learning (touchscreen-based location discrimination reversal [LDR]) and innate fear of open spaces (elevated plus maze [EPM]) followed by quantification of new DG neurons (doublecortin-immunoreactive cells [DCX+] cells). There was no effect of treatment (SCR shRNA vs. TRIP8b) on performance during general touchscreen training, LDR training, or the 1st days of LDR testing. However, in the last days of LDR testing, the TRIP8b shRNA mice had improved pattern separation (reached the first reversal more quickly and had more accurate discrimination) compared to the SCR shRNA mice, specifically when the load on pattern separation was high (lit squares close together or "small separation"). The TRIP8b shRNA mice were also more cognitively flexible (achieved more reversals) compared to the SCR shRNA mice in the last days of LDR testing. Supporting a specific influence on cognitive behavior, the SCR shRNA and TRIP8b shRNA mice did not differ in total distance traveled or in time spent in the closed arms of the EPM. Supporting an inducible increase in LEC-DG activity, DG neurogenesis was increased. These data indicate that the TRIP8b shRNA mice had better pattern separation and reversal learning and more neurogenesis compared to the SCR shRNA mice. This study advances fundamental and translational neuroscience knowledge relevant to two cognitive functions critical for adaptation and survival-behavioral pattern separation and cognitive flexibility-and suggests that the activity of LEC → DG neurons merits exploration as a therapeutic target to normalize dysfunctional DG behavioral output.
Collapse
Affiliation(s)
- Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ivan Soler
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- University of Pennsylvania, Philadelphia, PA, United States
| | - Fionya H. Tran
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Harley A. Haas
- University of Pennsylvania, Philadelphia, PA, United States
| | - Raymon Shi
- University of Pennsylvania, Philadelphia, PA, United States
| | | | - Maiko Suarez
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Christopher R. de Santis
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ryan P. Reynolds
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Amelia J. Eisch
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
28
|
Rabiller G, Ip Z, Zarrabian S, Zhang H, Sato Y, Yazdan-Shahmorad A, Liu J. Type-2 diabetes alters hippocampal neural oscillations and disrupts synchrony between hippocampus and cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.25.542288. [PMID: 37292743 PMCID: PMC10245872 DOI: 10.1101/2023.05.25.542288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Type 2 diabetes mellitus (T2DM) increases the risk of neurological diseases, yet how brain oscillations change as age and T2DM interact is not well characterized. To delineate the age and diabetic effect on neurophysiology, we recorded local field potentials with multichannel electrodes spanning the somatosensory cortex and hippocampus (HPC) under urethane anesthesia in diabetic and normoglycemic control mice, at 200 and 400 days of age. We analyzed the signal power of brain oscillations, brain state, sharp wave associate ripples (SPW-Rs), and functional connectivity between the cortex and HPC. We found that while both age and T2DM were correlated with a breakdown in long-range functional connectivity and reduced neurogenesis in the dentate gyrus and subventricular zone, T2DM further slowed brain oscillations and reduced theta-gamma coupling. Age and T2DM also prolonged the duration of SPW-Rs and increased gamma power during SPW-R phase. Our results have identified potential electrophysiological substrates of hippocampal changes associated with T2DM and age. The perturbed brain oscillation features and diminished neurogenesis may underlie T2DM-accelerated cognitive impairment.
Collapse
|
29
|
Li YD, Luo YJ, Xie L, Tart DS, Sheehy RN, Zhang L, Coleman LG, Chen X, Song J. Activation of hypothalamic-enhanced adult-born neurons restores cognitive and affective function in Alzheimer's disease. Cell Stem Cell 2023; 30:415-432.e6. [PMID: 37028406 PMCID: PMC10150940 DOI: 10.1016/j.stem.2023.02.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/29/2022] [Accepted: 02/14/2023] [Indexed: 04/09/2023]
Abstract
Patients with Alzheimer's disease (AD) exhibit progressive memory loss, depression, and anxiety, accompanied by impaired adult hippocampal neurogenesis (AHN). Whether AHN can be enhanced in impaired AD brain to restore cognitive and affective function remains elusive. Here, we report that patterned optogenetic stimulation of the hypothalamic supramammillary nucleus (SuM) enhances AHN in two distinct AD mouse models, 5×FAD and 3×Tg-AD. Strikingly, the chemogenetic activation of SuM-enhanced adult-born neurons (ABNs) rescues memory and emotion deficits in these AD mice. By contrast, SuM stimulation alone or activation of ABNs without SuM modification fails to restore behavioral deficits. Furthermore, quantitative phosphoproteomics analyses reveal activation of the canonical pathways related to synaptic plasticity and microglia phagocytosis of plaques following acute chemogenetic activation of SuM-enhanced (vs. control) ABNs. Our study establishes the activity-dependent contribution of SuM-enhanced ABNs in modulating AD-related deficits and informs signaling mechanisms mediated by the activation of SuM-enhanced ABNs.
Collapse
Affiliation(s)
- Ya-Dong Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yan-Jia Luo
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dalton S Tart
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ryan N Sheehy
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Pharmacology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Libo Zhang
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leon G Coleman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
30
|
Li L, Miao J, Jiang Y, Dai CL, Iqbal K, Liu F, Chu D. Passive immunization inhibits tau phosphorylation and improves recognition learning and memory in 3xTg-AD mice. Exp Neurol 2023; 362:114337. [PMID: 36717015 DOI: 10.1016/j.expneurol.2023.114337] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/05/2023] [Accepted: 01/24/2023] [Indexed: 01/30/2023]
Abstract
Tau pathology is essential in the pathogenesis of Alzheimer's disease (AD) and related tauopathies. Tau immunotherapy aimed at reducing the progression of tau pathology provides a potential therapeutic strategy for treating these diseases. By screening monoclonal antibodies 43D, 63B, 39E10, and 77G7 that recognize epitopes ranging from tau's N-terminus to C-terminus, we found the 77G7, which targets the microtubule-binding domain promoted tau clearance in a dose-dependent manner by entering neuronal cells in vitro. Intra-cerebroventricular injection of 77G7 antibody reduced tau levels in the wild-type FVB mouse brain. Without influencing the levels of detergent-insoluble and aggregated tau, intravenous injection of 77G7 reduced tau hyperphosphorylation in the brain and improved novel object recognition but not spatial learning and memory in 15-18-month-old 3xTg-AD mice. These studies suggest that epitopes recognized by tau antibodies are crucial for the efficacy of immunotherapy. Immunization with antibody 77G7 provides a novel potential opportunity for tau-directed immunotherapy of AD and related tauopathies.
Collapse
Affiliation(s)
- Longfei Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Jin Miao
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA; Laboratory Animal Center, Nantong University, Nantong 226001, China
| | - Yanli Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Chun-Ling Dai
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA.
| | - Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA.
| |
Collapse
|
31
|
Li YD, Luo YJ, Song J. Optimizing memory performance and emotional states: multi-level enhancement of adult hippocampal neurogenesis. Curr Opin Neurobiol 2023; 79:102693. [PMID: 36822141 PMCID: PMC10023407 DOI: 10.1016/j.conb.2023.102693] [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: 11/03/2022] [Revised: 01/03/2023] [Accepted: 01/12/2023] [Indexed: 02/25/2023]
Abstract
Adult hippocampal neurogenesis (AHN) plays a key role in modulating memory and emotion processing. A fundamental question remains on how to effectively modulate AHN to improve hippocampal function. Here, we review recent work on how distinct aspects of hippocampal neurogenesis, including the number, maturation state, and activity of adult-born neurons (ABNs), contribute to overall hippocampal function. We propose multi-level enhancement of hippocampal neurogenesis with the combination of increased number, elevated activity, and enhanced maturation of ABNs as a potential strategy to optimize overall hippocampal performance. In addition, integration of ABNs induces significant remodeling of the local hippocampal circuits, which may in turn modulates brain-wide network dynamics. We discuss recent progress on how integration of ABNs contributes to local hippocampal circuit and brain-wide network dynamics during behavior.
Collapse
Affiliation(s)
- Ya-Dong Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA. https://twitter.com/yadlee2
| | - Yan-Jia Luo
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
32
|
Fuchigami T, Itokazu Y, Yu RK. Ganglioside GD3 regulates neural stem cell quiescence and controls postnatal neurogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532547. [PMID: 36993675 PMCID: PMC10055067 DOI: 10.1101/2023.03.14.532547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
The postnatal neural stem cell (NSC) pool hosts quiescent and activated radial glia-like NSCs contributing to neurogenesis throughout adulthood. However, the underlying regulatory mechanism during the transition from quiescent NSCs to activated NSCs in the postnatal NSC niche is not fully understood. Lipid metabolism and lipid composition play important roles in regulating NSC fate determination. Biological lipid membranes define the individual cellular shape and help maintain cellular organization and are highly heterogenous in structure and there exist diverse microdomains (also known as lipid rafts), which are enriched with sugar molecules, such as glycosphingolipids. An often overlooked but key aspect is that the functional activities of proteins and genes are highly dependent upon their molecular environments. We previously reported that ganglioside GD3 is the predominant species in NSCs and that the reduced postnatal NSC pools are observed in global GD3-synthase knockout (GD3S-KO) mouse brains. The specific roles of GD3 in determining the stage and cell-lineage determination of NSCs remain unclear, since global GD3S-KO mice cannot distinguish if GD3 regulates postnatal neurogenesis or developmental impacts. Here we show that inducible GD3 deletion in postnatal radial glia-like NSCs promotes the NSC activation, resulting in the loss of the long-term maintenance of the adult NSC pools. The reduced neurogenesis in the subventricular zone (SVZ) and the dentate gyrus (DG) of GD3S-conditional-knockout mice led to impaired olfactory and memory functions. Thus, our results provide convincing evidence that postnatal GD3 maintains the quiescent state of radial glia-like NSCs in the adult NSC niche.
Collapse
Affiliation(s)
- Takahiro Fuchigami
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yutaka Itokazu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Robert K. Yu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
33
|
Lafourcade CA, Sparks FT, Bordey A, Wyneken U, Mohammadi MH. Cannabinoid regulation of neurons in the dentate gyrus during epileptogenesis: Role of CB1R-associated proteins and downstream pathways. Epilepsia 2023. [PMID: 36869624 DOI: 10.1111/epi.17569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/05/2023]
Abstract
The hippocampal formation plays a central role in the development of temporal lobe epilepsy (TLE), a disease characterized by recurrent, unprovoked epileptic discharges. TLE is a neurologic disorder characterized by acute long-lasting seizures (i.e., abnormal electrical activity in the brain) or seizures that occur in close proximity without recovery, typically after a brain injury or status epilepticus. After status epilepticus, epileptogenic hyperexcitability develops gradually over the following months to years, resulting in the emergence of chronic, recurrent seizures. Acting as a filter or gate, the hippocampal dentate gyrus (DG) normally prevents excessive excitation from propagating through the hippocampus, and is considered a critical region in the progression of epileptogenesis in pathological conditions. Importantly, lipid-derived endogenous cannabinoids (endocannabinoids), which are produced on demand as retrograde messengers, are central regulators of neuronal activity in the DG circuit. In this review, we summarize recent findings concerning the role of the DG in controlling hyperexcitability and propose how DG regulation by cannabinoids (CBs) could provide avenues for therapeutic interventions. We also highlight possible pathways and manipulations that could be relevant for the control of hyperexcitation. The use of CB compounds to treat epilepsies is controversial, as anecdotal evidence is not always validated by clinical trials. Recent publications shed light on the importance of the DG as a region regulating incoming hippocampal excitability during epileptogenesis. We review recent findings concerning the modulation of the hippocampal DG circuitry by CBs and discuss putative underlying pathways. A better understanding of the mechanisms by which CBs exert their action during seizures may be useful to improve therapies.
Collapse
Affiliation(s)
- Carlos A Lafourcade
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Fraser T Sparks
- Department of Neuroscience, Columbia University, New York, New York, USA.,Current: Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | - Angelique Bordey
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ursula Wyneken
- Centro de Investigación e Innovación Biomédica, Laboratorio de Neurociencias, Universidad de Los Andes, Santiago, Chile.,Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | | |
Collapse
|
34
|
Wang J, Yang Y, Liu J, Qiu J, Zhang D, Ou M, Kang Y, Zhu T, Zhou C. Loss of sodium leak channel (NALCN) in the ventral dentate gyrus impairs neuronal activity of the glutamatergic neurons for inflammation-induced depression in male mice. Brain Behav Immun 2023; 110:13-29. [PMID: 36796706 DOI: 10.1016/j.bbi.2023.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND The dentate gyrus (DG) has been implicated in the pathophysiology of depression. Many studies have revealed the cellular types, neural circuits, and morphological changes of the DG involved in the development of depression. However, the molecular regulating its intrinsic activity in depression is unknown. METHODS Utilizing the mode of depression induced by lipopolysaccharide (LPS), we investigate the involvement of the sodium leak channel (NALCN) in inflammation-induced depressive-like behaviors of male mice. The expression of NALCN was detected by immunohistochemistry and real-time polymerase chain reaction. DG microinjection of the adeno-associated virus or lentivirus was carried out using a stereotaxic instrument and followed by behavioral tests. Neuronal excitability and NALCN conductance were recorded by whole-cell patch-clamp techniques. RESULTS The expression and function of NALCN were reduced in both the dorsal and ventral DG in LPS-treated mice; whereas, only knocking down NALCN in the ventral pole produced depressive-like behaviors and this effect of NALCN was specific to ventral glutamatergic neurons. The excitability of ventral glutamatergic neurons was impaired by both the knockdown of NALCN and/or the treatment of LPS. Then, the overexpression of NALCN in the ventral glutamatergic neurons decreased the susceptibility of mice to inflammation-induced depression, and the intracranial injection of substance P (non-selective NALCN activator) into the ventral DG rapidly ameliorated inflammation-induced depression-like behaviors in an NALCN-dependent manner. CONCLUSIONS NALCN, which drives the neuronal activity of the ventral DG glutamatergic neurons, uniquely regulates depressive-like behaviors and susceptibility to depression. Therefore, the NALCN of glutamatergic neurons in the ventral DG may present a molecular target for rapid antidepressant drugs.
Collapse
Affiliation(s)
- Jinping Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaoxin Yang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingxuan Qiu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Donghang Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mengchan Ou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Kang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
35
|
Salta E, Lazarov O, Fitzsimons CP, Tanzi R, Lucassen PJ, Choi SH. Adult hippocampal neurogenesis in Alzheimer's disease: A roadmap to clinical relevance. Cell Stem Cell 2023; 30:120-136. [PMID: 36736288 PMCID: PMC10082636 DOI: 10.1016/j.stem.2023.01.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 02/05/2023]
Abstract
Adult hippocampal neurogenesis (AHN) drops sharply during early stages of Alzheimer's disease (AD), via unknown mechanisms, and correlates with cognitive status in AD patients. Understanding AHN regulation in AD could provide a framework for innovative pharmacological interventions. We here combine molecular, behavioral, and clinical data and critically discuss the multicellular complexity of the AHN niche in relation to AD pathophysiology. We further present a roadmap toward a better understanding of the role of AHN in AD by probing the promises and caveats of the latest technological advancements in the field and addressing the conceptual and methodological challenges ahead.
Collapse
Affiliation(s)
- Evgenia Salta
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, 808 S Wood St., Chicago, IL 60612, USA
| | - Carlos P Fitzsimons
- Brain Plasticity group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Rudolph Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, McCance Center for Brain Health, 114 16th Street, Boston, MA 02129, USA.
| | - Paul J Lucassen
- Brain Plasticity group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands; Center for Urban Mental Health, University of Amsterdam, Kruislaan 404, 1098 SM, Amsterdam, The Netherlands.
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, McCance Center for Brain Health, 114 16th Street, Boston, MA 02129, USA.
| |
Collapse
|
36
|
Ash AM, Regele-Blasco E, Seib DR, Chahley E, Skelton PD, Luikart BW, Snyder JS. Adult-born neurons inhibit developmentally-born neurons during spatial learning. Neurobiol Learn Mem 2023; 198:107710. [PMID: 36572174 DOI: 10.1016/j.nlm.2022.107710] [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: 10/12/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Ongoing neurogenesis in the dentate gyrus (DG) subregion of the hippocampus results in a heterogenous population of neurons. Immature adult-born neurons (ABNs) have physiological and anatomical properties that may give them a unique role in learning. For example, compared to older granule neurons, they have greater somatic excitability, which could facilitate their recruitment into memory traces. However, recruitment is also likely to depend on interactions with other DG neurons through processes such as lateral inhibition. Immature ABNs target inhibitory interneurons and, compared to older neurons, they receive less GABAergic inhibition. Thus, they may induce lateral inhibition of mature DG neurons while being less susceptible to inhibition themselves. To test this we used a chemogenetic approach to silence immature ABNs as rats learned a spatial water maze task, and measured activity (Fos expression) in ABNs and developmentally-born neurons (DBNs). A retrovirus expressing the inhibitory DREADD receptor, hM4Di, was injected into the dorsal DG of male rats at 6w to infect neurons born in adulthood. Animals were also injected with BrdU to label DBNs or ABNs. DBNs were significantly more active than immature 4-week-old ABNs. Silencing 4-week-old ABNs did not alter learning but it increased activity in DBNs. However, silencing ABNs did not affect activation in other ABNs within the DG. Silencing ABNs also did not alter Fos expression in parvalbumin- and somatostatin-expressing interneurons. Collectively, these results suggest that ABNs may directly inhibit DBN activity during hippocampal-dependent learning, which may be relevant for maintaining sparse hippocampal representations of experienced events.
Collapse
Affiliation(s)
- Alyssa M Ash
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Elena Regele-Blasco
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Désirée R Seib
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Erin Chahley
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Patrick D Skelton
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Jason S Snyder
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
37
|
Wu T, Li S, Du D, Li R, Liu P, Yin Z, Zhang H, Qiao Y, Li A. Olfactory-auditory sensory integration in the lateral entorhinal cortex. Prog Neurobiol 2023; 221:102399. [PMID: 36581184 DOI: 10.1016/j.pneurobio.2022.102399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 12/27/2022]
Abstract
Multisensory integration plays an important role in animal cognition. Although many studies have focused on visual-auditory integration, studies on olfactory-auditory integration are rare. Here, we investigated neural activity patterns and odor decoding in the lateral entorhinal cortex (LEC) under uni-sensory and multisensory stimuli in awake, head-fixed mice. Using specific retrograde tracing, we verified that the LEC receives direct inputs from the primary auditory cortex (AC) and the medial geniculate body (MGB). Strikingly, we found that mitral/tufted cells (M/Ts) in the olfactory bulb (OB) and neurons in the LEC respond to both olfactory and auditory stimuli. Sound decreased the neural responses evoked by odors in both the OB and LEC, for both excitatory and inhibitory responses. Interestingly, significant changes in odor decoding performance and modulation of odor-evoked local field potentials (LFPs) were observed only in the LEC. These data indicate that the LEC is a critical center for olfactory-auditory multisensory integration, with direct projections from both olfactory and auditory centers.
Collapse
Affiliation(s)
- Tingting Wu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China; Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou 221004, China; Clinical Hearing Center, Department of Otorhinolaryngology - Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China; Department of Otolaryngology, Eye, Ear, Nose and Throat Hospital, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Fudan University, Shanghai 200031, China
| | - Shan Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Deliang Du
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China; Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou 221004, China; Clinical Hearing Center, Department of Otorhinolaryngology - Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China
| | - Ruochen Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Penglai Liu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Zhaoyang Yin
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Yuehua Qiao
- Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou 221004, China; Clinical Hearing Center, Department of Otorhinolaryngology - Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China.
| | - Anan Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
38
|
Yun S, Soler I, Tran F, Haas HA, Shi R, Bancroft GL, Suarez M, de Santis CR, Reynolds RP, Eisch AJ. Behavioral pattern separation and cognitive flexibility are enhanced in a mouse model of increased lateral entorhinal cortex-dentate gyrus circuit activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525756. [PMID: 36747871 PMCID: PMC9900985 DOI: 10.1101/2023.01.26.525756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Behavioral pattern separation and cognitive flexibility are essential cognitive abilities which are disrupted in many brain disorders. Better understanding of the neural circuitry involved in these abilities will open paths to treatment. In humans and mice, discrimination and adaptation rely on integrity of the hippocampal dentate gyrus (DG) which both receive glutamatergic input from the entorhinal cortex (EC), including the lateral EC (LEC). Inducible increase of EC-DG circuit activity improves simple hippocampal-dependent associative learning and increases DG neurogenesis. Here we asked if the activity of LEC fan cells that directly project to the DG (LEC➔DG neurons) regulates behavioral pattern separation or cognitive flexibility. C57BL6/J male mice received bilateral LEC infusions of a virus expressing shRNA TRIP8b, an auxiliary protein of an HCN channel or a control virus (SCR shRNA); this approach increases the activity of LEC➔DG neurons. Four weeks later, mice underwent testing for behavioral pattern separation and reversal learning (touchscreen-based Location Discrimination Reversal [LDR] task) and innate fear of open spaces (elevated plus maze [EPM]) followed by counting of new DG neurons (doublecortin-immunoreactive cells [DCX+] cells). TRIP8b and SCR shRNA mice performed similarly in general touchscreen training and LDR training. However, in late LDR testing, TRIP8b shRNA mice reached the first reversal more quickly and had more accurate discrimination vs. SCR shRNA mice, specifically when pattern separation was challenging (lit squares close together or "small separation"). Also, TRIP8b shRNA mice achieved more reversals in late LDR testing vs. SCR shRNA mice. Supporting a specific influence on cognitive behavior, SCR shRNA and TRIP8b shRNA mice did not differ in total distance traveled or in time spent in the closed arms of the EPM. Supporting an inducible increase in LEC-DG activity, DG neurogenesis was increased. These data indicate TRIP8b shRNA mice had better pattern separation and reversal learning and more neurogenesis vs. SCR shRNA mice. This work advances fundamental and translational neuroscience knowledge relevant to two cognitive functions critical for adaptation and survival - behavioral pattern separation and cognitive flexibility - and suggests the activity of LEC➔DG neurons merits exploration as a therapeutic target to normalize dysfunctional DG behavioral output.
Collapse
|
39
|
Fölsz O, Trouche S, Croset V. Adult-born neurons add flexibility to hippocampal memories. Front Neurosci 2023; 17:1128623. [PMID: 36875670 PMCID: PMC9975346 DOI: 10.3389/fnins.2023.1128623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Although most neurons are generated embryonically, neurogenesis is maintained at low rates in specific brain areas throughout adulthood, including the dentate gyrus of the mammalian hippocampus. Episodic-like memories encoded in the hippocampus require the dentate gyrus to decorrelate similar experiences by generating distinct neuronal representations from overlapping inputs (pattern separation). Adult-born neurons integrating into the dentate gyrus circuit compete with resident mature cells for neuronal inputs and outputs, and recruit inhibitory circuits to limit hippocampal activity. They display transient hyperexcitability and hyperplasticity during maturation, making them more likely to be recruited by any given experience. Behavioral evidence suggests that adult-born neurons support pattern separation in the rodent dentate gyrus during encoding, and they have been proposed to provide a temporal stamp to memories encoded in close succession. The constant addition of neurons gradually degrades old connections, promoting generalization and ultimately forgetting of remote memories in the hippocampus. This makes space for new memories, preventing saturation and interference. Overall, a small population of adult-born neurons appears to make a unique contribution to hippocampal information encoding and removal. Although several inconsistencies regarding the functional relevance of neurogenesis remain, in this review we argue that immature neurons confer a unique form of transience on the dentate gyrus that complements synaptic plasticity to help animals flexibly adapt to changing environments.
Collapse
Affiliation(s)
- Orsolya Fölsz
- Department of Biosciences, Durham University, Durham, United Kingdom.,MSc in Neuroscience Programme, University of Oxford, Oxford, United Kingdom
| | - Stéphanie Trouche
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Vincent Croset
- Department of Biosciences, Durham University, Durham, United Kingdom
| |
Collapse
|
40
|
Lei B, Kang B, Lin W, Chen H, Hao Y, Ma J, Shi S, Zhong Y. Adult newborn granule cells confer emotional state-dependent adaptability in memory retrieval. SCIENCE ADVANCES 2022; 8:eabn2136. [PMID: 36367932 PMCID: PMC9651853 DOI: 10.1126/sciadv.abn2136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
Achieving optimal behavior requires animals to flexibly retrieve prior knowledge. Here, we show that adult newborn granule cells (anbGCs) mediate emotional state-dependent adaptability of memory retrieval. We find that acute social reward (aSR) enhances memory retrieval by increasing the reactivation of engram cells, while acute social stress (aSS) weakens retrieval and reduces the reactivation. Such bidirectional regulation relies on the activation of distinct populations of anbGCs by aSR and aSS, triggering opposing modifications of dDG activity, which is sufficient to regulate and predict the performance of memory retrieval. Concordantly, in emotional disorder models, aSR-dependent memory adaptability is impaired, while the effect of aSS remains intact. Together, our data revealed that anbGCs mediate adaptability of memory retrieval, allowing animals to flexibly retrieve memory according to the current emotional state, and suggested the essential roles of anbGCs in translating emotional information to the regulation of memory expression.
Collapse
Affiliation(s)
- Bo Lei
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
| | - Bilin Kang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
| | - Wantong Lin
- Department of Computer Science, Brandeis University, Waltham, MA 02453, USA
| | - Haichao Chen
- School of Medicine, Tsinghua University, Beijing 100084, P.R. China
| | - Yuejun Hao
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
| | - Jian Ma
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Songhai Shi
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Yi Zhong
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
41
|
McHugh SB, Lopes-Dos-Santos V, Gava GP, Hartwich K, Tam SKE, Bannerman DM, Dupret D. Adult-born dentate granule cells promote hippocampal population sparsity. Nat Neurosci 2022; 25:1481-1491. [PMID: 36216999 PMCID: PMC9630129 DOI: 10.1038/s41593-022-01176-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 09/01/2022] [Indexed: 01/13/2023]
Abstract
The dentate gyrus (DG) gates neocortical information flow to the hippocampus. Intriguingly, the DG also produces adult-born dentate granule cells (abDGCs) throughout the lifespan, but their contribution to downstream firing dynamics remains unclear. Here, we show that abDGCs promote sparser hippocampal population spiking during mnemonic processing of novel stimuli. By combining triple-(DG-CA3-CA1) ensemble recordings and optogenetic interventions in behaving mice, we show that abDGCs constitute a subset of high-firing-rate neurons with enhanced activity responses to novelty and strong modulation by theta oscillations. Selectively activating abDGCs in their 4-7-week post-birth period increases sparsity of hippocampal population patterns, whereas suppressing abDGCs reduces this sparsity, increases principal cell firing rates and impairs novel object recognition with reduced dimensionality of the network firing structure, without affecting single-neuron spatial representations. We propose that adult-born granule cells transiently support sparser hippocampal population activity structure for higher-dimensional responses relevant to effective mnemonic information processing.
Collapse
Affiliation(s)
- Stephen B McHugh
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
- Department of Experimental Psychology, University of Oxford, Oxford, UK.
| | - Vítor Lopes-Dos-Santos
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Giuseppe P Gava
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Katja Hartwich
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Shu K E Tam
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - David Dupret
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| |
Collapse
|
42
|
Li L, Jiang Y, Wu G, Mahaman YAR, Ke D, Wang Q, Zhang B, Wang JZ, Li HL, Liu R, Wang X. Phosphorylation of Truncated Tau Promotes Abnormal Native Tau Pathology and Neurodegeneration. Mol Neurobiol 2022; 59:6183-6199. [PMID: 35896773 DOI: 10.1007/s12035-022-02972-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/20/2022] [Indexed: 11/29/2022]
Abstract
Abnormal posttranslational modifications of tau play important roles in mediating neurodegeneration in tauopathies including Alzheimer's disease. Both phosphorylation and truncation are implicated in the pathogenesis of tauopathies. However, whether phosphorylation aggravates truncated tau-induced pathology and neurodegeneration remains elusive. Here, we construct different tau fragments cleaved by delta secretase, with either phosphorylation or non-phosphorylation mimic mutations, and evaluate the contributions of phosphorylation to truncated tau-induced pathological and behavioral alterations in vitro and in vivo through biochemical methods including detergent insoluble tau extraction, western blot, immunofluorescence, flow cytometry, and behavior tests. Our results show that the self-aggregation of phospho-truncated tau is significantly influenced by the domain it contains. N-terminal inhibits, proline-rich domain promotes, and C-terminus have no impact on phospho-truncated tau aggregation. Phosphorylation of truncated tau1-368, which contains the microtubule-binding repeat domain and the proline-rich domain, induces endogenous tau phosphorylation and aggregation. In vivo, phospho-tau1-368 but not non-phospho-tau1-368 leads to a decrease in body weight of C57BL/6 J mice. Intriguingly, although tau1-368-induced anxiety behavior in C57BL/6 J mice is phosphorylation-independent, the recognition memory of mice is impaired by phospho-tau1-368, but not by non-phospho-tau1-368. Immunofluorescence staining shows that overexpressing phospho-tau1-368 results in neuronal loss and gliosis in the hippocampus, while the transmission of tau1-368 is phosphorylation-independent as revealed by the flow cytometry results in vitro and immunofluorescence staining in vivo. Our findings indicate that phosphorylation of truncated tau significantly fosters endogenous tau pathology and neurodegeneration.
Collapse
Affiliation(s)
- Longfei Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanli Jiang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gang Wu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yacoubou Abdoul Razak Mahaman
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bin Zhang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, JS, China
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Hong-Lian Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rong Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, JS, China.
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China.
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.
| |
Collapse
|
43
|
Kang YJ, Lee SH, Boychuk JA, Butler CR, Juras JA, Cloyd RA, Smith BN. Adult Born Dentate Granule Cell Mediated Upregulation of Feedback Inhibition in a Mouse Model of Traumatic Brain Injury. J Neurosci 2022; 42:7077-7093. [PMID: 36002261 PMCID: PMC9480876 DOI: 10.1523/jneurosci.2263-21.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/21/2022] Open
Abstract
Post-traumatic epilepsy (PTE) and behavioral comorbidities frequently develop after traumatic brain injury (TBI). Aberrant neurogenesis of dentate granule cells (DGCs) after TBI may contribute to the synaptic reorganization that occurs in PTE, but how neurogenesis at different times relative to the injury contributes to feedback inhibition and recurrent excitation in the dentate gyrus is unknown. Thus, we examined whether DGCs born at different postnatal ages differentially participate in feedback inhibition and recurrent excitation in the dentate gyrus using the controlled cortical impact (CCI) model of TBI. Both sexes of transgenic mice expressing channelrhodopsin2 (ChR2) in postnatally born DGCs were used for optogenetic activation of three DGC cohorts: postnatally early born DGCs, or those born just before or after CCI. We performed whole-cell patch-clamp recordings from ChR2-negative, mature DGCs and parvalbumin-expressing basket cells (PVBCs) in hippocampal slices to determine whether optogenetic activation of postnatally born DGCs increases feedback inhibition and/or recurrent excitation in mice 8-10 weeks after CCI and whether PVBCs are targets of ChR2-positive DGCs. In the dentate gyrus ipsilateral to CCI, activation of ChR2-expressing DGCs born before CCI produced increased feedback inhibition in ChR2-negative DGCs and increased excitation in PVBCs compared with those from sham controls. This upregulated feedback inhibition was less prominent in DGCs born early in life or after CCI. Surprisingly, ChR2-positive DGC activation rarely evoked recurrent excitation in mature DGCs from any cohort. These results support that DGC birth date-related increased feedback inhibition in of DGCs may contribute to altered excitability after TBI.SIGNIFICANCE STATEMENT Dentate granule cells (DGCs) control excitability of the dentate gyrus through synaptic interactions with inhibitory GABAergic interneurons. Persistent changes in DGC synaptic connectivity develop after traumatic brain injury, contributing to hyperexcitability in post-traumatic epilepsy (PTE). However, the impact of DGC neurogenesis on synaptic reorganization, especially on inhibitory circuits, after brain injury is not adequately described. Here, upregulation of feedback inhibition in mature DGCs from male and female mice was associated with increased excitation of parvalbumin-expressing basket cells by postnatally born DGCs, providing novel insights into underlying mechanisms of altered excitability after brain injury. A better understanding of these inhibitory circuit changes can help formulate hypotheses for development of novel, evidence-based treatments for post-traumatic epilepsy by targeting birth date-specific subsets of DGCs.
Collapse
Affiliation(s)
- Young-Jin Kang
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Sang-Hun Lee
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Epilepsy Research Center, University of Kentucky, Lexington, Kentucky 40536
| | - Jeffery A Boychuk
- Epilepsy Research Center, University of Kentucky, Lexington, Kentucky 40536
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Corwin R Butler
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - J Anna Juras
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Ryan A Cloyd
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Bret N Smith
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Epilepsy Research Center, University of Kentucky, Lexington, Kentucky 40536
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
| |
Collapse
|
44
|
Mishra R, Phan T, Kumar P, Morrissey Z, Gupta M, Hollands C, Shetti A, Lopez KL, Maienschein-Cline M, Suh H, Hen R, Lazarov O. Augmenting neurogenesis rescues memory impairments in Alzheimer's disease by restoring the memory-storing neurons. J Exp Med 2022; 219:e20220391. [PMID: 35984475 PMCID: PMC9399756 DOI: 10.1084/jem.20220391] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/16/2022] [Accepted: 06/28/2022] [Indexed: 12/04/2022] Open
Abstract
Hippocampal neurogenesis is impaired in Alzheimer's disease (AD) patients and familial Alzheimer's disease (FAD) mouse models. However, it is unknown whether new neurons play a causative role in memory deficits. Here, we show that immature neurons were actively recruited into the engram following a hippocampus-dependent task. However, their recruitment is severely deficient in FAD. Recruited immature neurons exhibited compromised spine density and altered transcript profile. Targeted augmentation of neurogenesis in FAD mice restored the number of new neurons in the engram, the dendritic spine density, and the transcription signature of both immature and mature neurons, ultimately leading to the rescue of memory. Chemogenetic inactivation of immature neurons following enhanced neurogenesis in AD, reversed mouse performance, and diminished memory. Notably, AD-linked App, ApoE, and Adam10 were of the top differentially expressed genes in the engram. Collectively, these observations suggest that defective neurogenesis contributes to memory failure in AD.
Collapse
Affiliation(s)
- Rachana Mishra
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Trongha Phan
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Pavan Kumar
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Zachery Morrissey
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
- Department of Psychiatry, College of Medicine, The University of Illinois at Chicago, Chicago, IL
- The Graduate Program in Neuroscience, The University of Illinois at Chicago, Chicago, IL
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Carolyn Hollands
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Aashutosh Shetti
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Kyra Lauren Lopez
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | | | - Hoonkyo Suh
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH
| | - Rene Hen
- Department of Psychiatry, Irving Medical Center, Columbia University, New York, NY
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
45
|
Segi-Nishida E, Suzuki K. Regulation of adult-born and mature neurons in stress response and antidepressant action in the dentate gyrus of the hippocampus. Neurosci Res 2022:S0168-0102(22)00233-4. [PMID: 36030966 DOI: 10.1016/j.neures.2022.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022]
Abstract
The dentate gyrus (DG) of the hippocampus has been implicated in the regulation of stress responses, and in the pathophysiology and treatment of depression. This review discusses the cellular changes caused by chronic stress and the cellular role of the DG in stress-induced behavioral changes and its antidepressant-like effects. Regarding adult-born neurogenic processes in the DG, chronic stress, such as repeated social defeat, suppresses cell proliferation during and immediately after stress; however, this effect is transient. The subsequent differentiation and survival processes are differentially regulated depending on the timing and sensitivity of stress. The activation of young adult-born neurons during stress contributes to stress resilience, while the transient increase in the survival of adult-born neurons after the cessation of stress seems to promote stress susceptibility. In mature granule neurons, the predominant cells in the DG, synaptic plasticity is suppressed by chronic stress. However, a group of mature granule neurons is activated by chronic stress. Chronic antidepressant treatment can transform mature granule neurons to a phenotype resembling that of immature neurons, characterized as "dematuration". Adult-born neurons suppress the activation of mature granule neurons during stress, indicating that local neural interactions within the DG are important for the stress response. Elucidating the stress-associated context- and timing-dependent cellular changes and functions in the DG will provide insights into stress-related psychiatric diseases.
Collapse
Affiliation(s)
- Eri Segi-Nishida
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, Japan.
| | - Kanzo Suzuki
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, Japan
| |
Collapse
|
46
|
Early life adversity shapes neural circuit function during sensitive postnatal developmental periods. Transl Psychiatry 2022; 12:306. [PMID: 35915071 PMCID: PMC9343623 DOI: 10.1038/s41398-022-02092-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
Early life adversity (ELA) is a major risk factor for mental illness, but the neurobiological mechanisms by which ELA increases the risk for future psychopathology are still poorly understood. Brain development is particularly malleable during prenatal and early postnatal life, when complex neural circuits are being formed and refined through an interplay of excitatory and inhibitory neural input, synaptogenesis, synaptic pruning, myelination, and neurogenesis. Adversity that influences these processes during sensitive periods of development can thus have long-lasting and pervasive effects on neural circuit maturation. In this review, we will discuss clinical and preclinical evidence for the impact of ELA on neural circuit formation with a focus on the early postnatal period, and how long-lasting impairments in these circuits can affect future behavior. We provide converging evidence from human and animal studies on how ELA alters the functional development of brain regions, neural circuits, and neurotransmitter systems that are crucial for cognition and affective behavior, including the hippocampus, the hypothalamus-pituitary-adrenal (HPA) axis, neural networks of fear responses and cognition, and the serotonin (5-HT) system. We also discuss how gene-by-environment (GxE) interactions can determine individual differences in susceptibility and resilience to ELA, as well as molecular pathways by which ELA regulates neural circuit development, for which we emphasize epigenetic mechanisms. Understanding the molecular and neurobiological mechanisms underlying ELA effects on brain function and psychopathology during early postnatal sensitive periods may have great potential to advance strategies to better treat or prevent psychiatric disorders that have their origin early in life.
Collapse
|
47
|
Luo H, Ren J, Sun Y, Liu Y, Zhou F, Shi G, Zhou J. Recent advances in chemical fixation of CO2 based on flow chemistry. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.107782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
48
|
Shridhar S, Mishra P, Narayanan R. Dominant role of adult neurogenesis-induced structural heterogeneities in driving plasticity heterogeneity in dentate gyrus granule cells. Hippocampus 2022; 32:488-516. [PMID: 35561083 PMCID: PMC9322436 DOI: 10.1002/hipo.23422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/21/2022] [Accepted: 04/28/2022] [Indexed: 02/02/2023]
Abstract
Neurons and synapses manifest pronounced variability in the amount of plasticity induced by identical activity patterns. The mechanisms underlying such plasticity heterogeneity, which have been implicated in context‐specific resource allocation during encoding, have remained unexplored. Here, we employed a systematic physiologically constrained parametric search to identify the cellular mechanisms behind plasticity heterogeneity in dentate gyrus granule cells. We used heterogeneous model populations to ensure that our conclusions were not biased by parametric choices in a single hand‐tuned model. We found that each of intrinsic, synaptic, and structural heterogeneities independently yielded heterogeneities in synaptic plasticity profiles obtained with two different induction protocols. However, among the disparate forms of neural‐circuit heterogeneities, our analyses demonstrated the dominance of neurogenesis‐induced structural heterogeneities in driving plasticity heterogeneity in granule cells. We found that strong relationships between neuronal intrinsic excitability and plasticity emerged only when adult neurogenesis‐induced heterogeneities in neural structure were accounted for. Importantly, our analyses showed that it was not imperative that the manifestation of neural‐circuit heterogeneities must translate to heterogeneities in plasticity profiles. Specifically, despite the expression of heterogeneities in structural, synaptic, and intrinsic neuronal properties, similar plasticity profiles were attainable across all models through synergistic interactions among these heterogeneities. We assessed the parametric combinations required for the manifestation of such degeneracy in the expression of plasticity profiles. We found that immature cells showed physiological plasticity profiles despite receiving afferent inputs with weak synaptic strengths. Thus, the high intrinsic excitability of immature granule cells was sufficient to counterbalance their low excitatory drive in the expression of plasticity profile degeneracy. Together, our analyses demonstrate that disparate forms of neural‐circuit heterogeneities could mechanistically drive plasticity heterogeneity, but also caution against treating neural‐circuit heterogeneities as proxies for plasticity heterogeneity. Our study emphasizes the need for quantitatively characterizing the relationship between neural‐circuit and plasticity heterogeneities across brain regions.
Collapse
Affiliation(s)
- Sameera Shridhar
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, India
| | - Poonam Mishra
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, India
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka, India
| |
Collapse
|
49
|
Rawat R, Tunc-Ozcan E, McGuire TL, Peng CY, Kessler JA. Ketamine activates adult-born immature granule neurons to rapidly alleviate depression-like behaviors in mice. Nat Commun 2022; 13:2650. [PMID: 35551462 PMCID: PMC9098911 DOI: 10.1038/s41467-022-30386-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 04/29/2022] [Indexed: 12/16/2022] Open
Abstract
Ketamine treatment decreases depressive symptoms within hours, but the mechanisms mediating these rapid antidepressant effects are unclear. Here, we demonstrate that activity of adult-born immature granule neurons (ABINs) in the mouse hippocampal dentate gyrus is both necessary and sufficient for the rapid antidepressant effects of ketamine. Ketamine treatment activates ABINs in parallel with its behavioral effects in both stressed and unstressed mice. Chemogenetic inhibition of ABIN activity blocks the antidepressant effects of ketamine, indicating that this activity is necessary for the behavioral effects. Conversely, chemogenetic activation of ABINs without any change in neuron numbers mimics both the cellular and the behavioral effects of ketamine, indicating that increased activity of ABINs is sufficient for rapid antidepressant effects. These findings thus identify a specific cell population that mediates the antidepressant actions of ketamine, indicating that ABINs can potentially be targeted to limit ketamine's side effects while preserving its therapeutic efficacy.
Collapse
Affiliation(s)
- Radhika Rawat
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| | - Elif Tunc-Ozcan
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Tammy L McGuire
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Chian-Yu Peng
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - John A Kessler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| |
Collapse
|
50
|
Ambient-pressure hydrogenation of CO 2 into long-chain olefins. Nat Commun 2022; 13:2396. [PMID: 35504867 PMCID: PMC9064975 DOI: 10.1038/s41467-022-29971-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/25/2022] [Indexed: 11/25/2022] Open
Abstract
The conversion of CO2 by renewable power-generated hydrogen is a promising approach to a sustainable production of long-chain olefins (C4+=) which are currently produced from petroleum resources. The decentralized small-scale electrolysis for hydrogen generation requires the operation of CO2 hydrogenation in ambient-pressure units to match the manufacturing scales and flexible on-demand production. Herein, we report a Cu-Fe catalyst which is operated under ambient pressure with comparable C4+= selectivity (66.9%) to that of the state-of-the-art catalysts (66.8%) optimized under high pressure (35 bar). The catalyst is composed of copper, iron oxides, and iron carbides. Iron oxides enable reverse-water-gas-shift to produce CO. The synergy of carbide path over iron carbides and CO insertion path over interfacial sites between copper and iron carbides leads to efficient C-C coupling into C4+=. This work contributes to the development of small-scale low-pressure devices for CO2 hydrogenation compatible with sustainable hydrogen production. The conversion of CO2 by renewable power-generated hydrogen is a promising approach to a sustainable production of long-chain olefins. Here the authors report a Cu-Fe catalyst which achieves the hydrogenation of CO2 into long-chain olefins under ambient pressure via the synergy of carbide mechanism and CO insertion mechanism.
Collapse
|