1
|
Liu Y, Chen S, Li J, Song Z, Wang J, Ren X, Qian Y, Ouyang W. Effects of high-intensity interval training and moderate-intensity continuous training on neural dynamics and firing in the CA1-MEC region of mice. J Appl Physiol (1985) 2025; 138:31-44. [PMID: 39589768 DOI: 10.1152/japplphysiol.00778.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
The aim of this study is to investigate the differential impacts of high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) on neural circuit dynamics and neuronal firing in the hippocampal CA1 subregion (CA1) region and medial entorhinal cortex (MEC) of mice. Forty-two male ICR mice were randomized into control, HIIT, and MICT groups. Electrophysiological recordings were performed pre- and postintervention to assess neural circuit dynamics and neuronal firing patterns in the CA1-MEC pathway. Both exercise protocols increased local field potential (LFP) coherence, with MICT showing a more pronounced effect on δ and γ coherences (P < 0.05). Both modalities reduced δ power spectral density (PSD) (HIIT, P < 0.05; MICT, P < 0.01) and elevated θ, β, and γ PSDs. Neuronal firing frequency improved in both CA1 and MEC following HIIT and MICT (P < 0.05). HIIT enhanced firing regularity in CA1 (P < 0.05), whereas MICT improved regularity in both regions (P < 0.05). Both protocols reduced firing latency (HIIT, P < 0.05; MICT, P < 0.01) and enhanced burst firing ratio, interburst interval (IBI), burst duration (BD), and LFP phase locking (P < 0.05 or P < 0.01). Notably, MICT significantly improved spatial working memory and novel recognition abilities, as evidenced by increased novel arm time, entries, and preference index (P < 0.01). This study reveals that both HIIT and MICT positively impact neural processing and information integration in the CA1-MEC network of mice. Notably, MICT exhibits a more pronounced impact on neural functional connectivity and cognitive function compared with HIIT. These findings, coupled with the similarities in hippocampal electrophysiological characteristics between rodents and humans, suggest potential exercise-mediated neural plasticity and cognitive benefits in humans.NEW & NOTEWORTHY This study is the first to investigate HIIT and MICT's effects on neural activity in the mouse CA1-MEC circuit, demonstrating that exercise modulates processing, enhances integration, and boosts cognitive performance. Due to similar hippocampal electrophysiology in rodents and humans during movement and navigation, our findings suggest implications for human brain neural changes, advancing the understanding of neurophysiological mechanisms underlying exercise-cognition interactions and informing exercise recommendations for cognitive health.
Collapse
Affiliation(s)
- Yuncheng Liu
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, People's Republic of China
| | - Shiqiang Chen
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, People's Republic of China
| | - Junliang Li
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, People's Republic of China
| | - Zengfei Song
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, People's Republic of China
| | - Jihui Wang
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, People's Republic of China
| | - Xiping Ren
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, People's Republic of China
| | - Yongdong Qian
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, People's Republic of China
| | - Wei Ouyang
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, People's Republic of China
| |
Collapse
|
2
|
Wu JL, Li ZM, Chen H, Chen WJ, Hu NY, Jin SY, Li XW, Chen YH, Yang JM, Gao TM. Distinct septo-hippocampal cholinergic projections separately mediate stress-induced emotional and cognitive deficits. SCIENCE ADVANCES 2024; 10:eado1508. [PMID: 39514666 PMCID: PMC11546849 DOI: 10.1126/sciadv.ado1508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
Patients suffering from chronic stress develop numerous symptoms, including emotional and cognitive deficits. The precise circuit mechanisms underlying different symptoms remain poorly understood. We identified two distinct basal forebrain cholinergic subpopulations in mice projecting to the dorsal hippocampus (dHPC) or ventral hippocampus (vHPC), which exhibited distinct input organizations, electrophysiological characteristics, transcriptomics, and responses to positive and negative valences of stimuli and were critical for cognitive and emotional modulation, respectively. Moreover, chronic stress induced elevated anxiety levels and cognitive deficits in mice, accompanied by enhanced vHPC but suppressed dHPC cholinergic projections. Chemogenetic activation of dHPC or inhibition of vHPC cholinergic projections alleviated stress-induced aberrant behaviors. Furthermore, we identified that the acetylcholinesterase inhibitor donepezil combined with blockade of muscarinic receptor 1-type muscarinic acetylcholine receptors in the vHPC rescued both stress-induced phenotypes. These data illuminated distinct septo-hippocampal cholinergic circuits mediated specific symptoms independently under stress, which may provide promising strategies for circuit-based treating of stress-related psychiatric disorders.
Collapse
Affiliation(s)
| | | | - Hao Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wen-Jun Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Neng-Yuan Hu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shi-Yang Jin
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao-Wen Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yi-Hua Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Lin X, Ghafuri A, Chen X, Kazmi M, Nitz DA, Xu X. Projection-specific circuits of retrosplenial cortex with differential contributions to spatial cognition. Mol Psychiatry 2024:10.1038/s41380-024-02819-8. [PMID: 39511453 DOI: 10.1038/s41380-024-02819-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/20/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024]
Abstract
Retrosplenial cortex (RSC) is a brain region involved in neuropsychiatric and neurodegenerative disorders. It has reciprocal connections with a diverse set of cortical and subcortical brain regions, but the afferent structure and behavioral function of circuits defined by its projection-specific sub-populations have yet to be determined. The corticocortical connections between RSC and secondary motor cortex (M2), as well as corticothalamic connections between RSC and anterodorsal thalamus (AD) have been hypothesized to function as semi-independent, but parallel pathways that impact spatial information processing in distinct ways. We used retrograde and anterograde viral tracers and monosynaptic retrograde rabies virus to quantitatively characterize and compare the afferent and efferent distributions of retrosplenial neuron sub-populations projecting to M2 and AD. AD-projecting and M2-projecting RSC neurons overlap in their collateral projections to other brain regions, but not in their projections to M2 and AD, respectively. Compared with AD-projecting RSC neurons, M2-projecting RSC neurons received much greater afferent input from the dorsal subiculum, AD, lateral dorsal and lateral posterior thalamus, and somatosensory cortex. AD-projecting RSC neurons received greater input from the anterior cingulate cortex and medial septum. We performed chemogenetic inhibition of M2- and AD-projecting RSC neurons and examined its impact on object-location memory, object-recognition, open-field exploration, and place-action association. Our findings indicate that inhibition of M2-projecting RSC neurons impairs object location memory as well as place-action association, while the RSC to AD pathway impacts only object-location memory. The findings indicate that RSC is composed of semi-independent circuits distinguishable by their afferent/efferent distributions and differing in the cognitive functions to which they contribute.
Collapse
Affiliation(s)
- Xiaoxiao Lin
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Ali Ghafuri
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Xiaojun Chen
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Musab Kazmi
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Douglas A Nitz
- Department of Cognitive Science, University of California, La Jolla, San Diego, CA, 92093, USA.
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, 92697, USA.
| | - Xiangmin Xu
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
4
|
Hui Y, Zhong Y, Kuang L, Xu J, Hao Y, Cao J, Zheng T. O-GlcNAcylation of circadian clock protein Bmal1 impairs cognitive function in diabetic mice. EMBO J 2024; 43:5667-5689. [PMID: 39375536 PMCID: PMC11574178 DOI: 10.1038/s44318-024-00263-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024] Open
Abstract
Neuronal damage in the hippocampus induced by high glucose has been shown to promote the onset and development of cognitive impairment in diabetes, but the underlying molecular mechanism remains unclear. Guided by single-cell RNA sequencing, we here report that high glucose increases O-GlcNAcylation of Bmal1 in hippocampal neurons. This glycosylation promotes the binding of Clock to Bmal1, resulting in the expression of transcription factor Bhlhe41 and its target Dnajb4. Upregulated Dnajb4 in turn leads to ubiquitination and degradation of the mitochondrial Na + /Ca2+ exchanger NCLX, thereby inducing mitochondrial calcium overload that causes neuronal damage and cognitive impairment in mice. Notably, Bhlhe41 downregulation or treatment with a short peptide that specifically blocks O-GlcNAcylation of Bmal1 on Ser424 mitigated these adverse effects in diabetic mouse models. These data highlight the crucial role of O-GlcNAcylation in circadian clock gene expression and may facilitate the design of targeted therapies for diabetes-associated cognitive impairment.
Collapse
Affiliation(s)
- Ya Hui
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Yuanmei Zhong
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Liuyu Kuang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Jingxi Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Yuqi Hao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Jingxue Cao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China
| | - Tianpeng Zheng
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, 541199, Guilin, Guangxi, P. R. China.
| |
Collapse
|
5
|
Hou R, Liu Z, Jin Z, Huang D, Hu Y, Du W, Zhu D, Yang L, Weng Y, Yuan T, Lu B, Wang Y, Ping Y, Xiao X. Coordinated Interactions between the Hippocampus and Retrosplenial Cortex in Spatial Memory. RESEARCH (WASHINGTON, D.C.) 2024; 7:0521. [PMID: 39483173 PMCID: PMC11525046 DOI: 10.34133/research.0521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/05/2024] [Accepted: 10/12/2024] [Indexed: 11/03/2024]
Abstract
While a hippocampal-cortical dialogue is generally thought to mediate memory consolidation, which is crucial for engram function, how it works remains largely unknown. Here, we examined the interplay of neural signals from the retrosplenial cortex (RSC), a neocortical region, and from the hippocampus in memory consolidation by simultaneously recording sharp-wave ripples (SWRs) of dorsal hippocampal CA1 and neural signals of RSC in free-moving mice during the delayed spatial alternation task (DSAT) and subsequent sleep. Hippocampal-RSC coordination during SWRs was identified in nonrapid eye movement (NREM) sleep, reflecting neural reactivation of decision-making in the task, as shown by a peak reactivation strength within SWRs. Using modified generalized linear models (GLMs), we traced information flow through the RSC-CA1-RSC circuit around SWRs during sleep following DSAT. Our findings show that after spatial training, RSC excitatory neurons typically increase CA1 activity prior to hippocampal SWRs, potentially initiating hippocampal memory replay, while inhibitory neurons are activated by hippocampal outputs in post-SWRs. We further identified certain excitatory neurons in the RSC that encoded spatial information related to the DSAT. These neurons, classified as splitters and location-related cells, showed varied responses to hippocampal SWRs. Overall, our study highlights the complex dynamics between the RSC and hippocampal CA1 region during SWRs in NREM sleep, underscoring their critical interplay in spatial memory consolidation.
Collapse
Affiliation(s)
- Ruiqing Hou
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Ziyue Liu
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Zichen Jin
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Dongxue Huang
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Yue Hu
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Wenjie Du
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Danyi Zhu
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Leiting Yang
- School of Life Science,
Fudan University, Shanghai 200032, China
| | - Yuanfeng Weng
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Tifei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Brain Health Institute, National Center for Mental Disorders, Shanghai Mental Health Center,
Shanghai Jiaotong University School of Medicine, Shanghai 200030, China
| | - Bin Lu
- Department of Endocrinology and Metabolism, Huadong Hospital,
Fudan University, Shanghai 200040, China
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education),
Shanghai JiaoTong University, Shanghai 200240, China
| | - Xiao Xiao
- Department of Anesthesiology, Huashan Hospital; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence, MOE Frontiers Center for Brain Science,
Fudan University, Shanghai 200433, China
| |
Collapse
|
6
|
Zhang XF, Li YD, Li Y, Li Y, Xu D, Bi LL, Xu HB. Ventral subiculum promotes wakefulness through several pathways in male mice. Neuropsychopharmacology 2024; 49:1468-1480. [PMID: 38734818 PMCID: PMC11251017 DOI: 10.1038/s41386-024-01875-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024]
Abstract
The ventral subiculum (vSUB), the major output structure of the hippocampal formation, regulates motivation, stress integration, and anxiety-like behaviors that rely on heightened arousal. However, the roles and underlying neural circuits of the vSUB in wakefulness are poorly known. Using in vivo fiber photometry and multichannel electrophysiological recordings in mice, we found that the vSUB glutamatergic neurons exhibited high activities during wakefulness. Moreover, activation of vSUB glutamatergic neurons caused an increase in wakefulness and anxiety-like behaviors and induced a rapid transition from sleep to wakefulness. In addition, optogenetic stimulation of vSUB glutamatergic terminals and retrograde-targeted chemogenetic activation of vSUB glutamatergic neurons revealed that vSUB promoted arousal by innervating the lateral hypothalamus (LH), nucleus accumbens (NAc) shell, and prefrontal cortex (PFC). Nevertheless, local microinjection of dopamine D1 or D2/D3 receptor antagonist blocked the wake-promoting effect induced by chemogenetic activation of vSUB pathways. Finally, chemogenetic inhibition of vSUB glutamatergic neurons decreased arousal. Altogether, our findings reveal a prominent contribution of vSUB glutamatergic neurons to the control of wakefulness through several pathways.
Collapse
Affiliation(s)
- Xue-Fen Zhang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yi-Dan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yue Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Ying Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Dan Xu
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Lin-Lin Bi
- Department of Pathology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China.
- Center for Pathology and Molecular Diagnostics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| | - Hai-Bo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
7
|
Demchuk AM, Esteves IM, Chang H, Sun J, McNaughton BL. Hierarchical Gradients of Encoded Spatial and Sensory Information in the Neocortex Are Attenuated by Dorsal Hippocampal Lesions. J Neurosci 2024; 44:e1619232024. [PMID: 38942472 PMCID: PMC11293447 DOI: 10.1523/jneurosci.1619-23.2024] [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: 08/21/2023] [Revised: 04/16/2024] [Accepted: 05/13/2024] [Indexed: 06/30/2024] Open
Abstract
During navigation, the neocortex actively integrates learned spatial context with current sensory experience to guide behaviors. However, the relative encoding of spatial and sensorimotor information among cortical cells, and whether hippocampal feedback continues to modify these properties after learning, remains poorly understood. Thus, two-photon microscopy of male and female Thy1-GCaMP6s mice was used to longitudinally image neurons spanning superficial retrosplenial cortex and layers II-Va of primary and secondary motor cortices before and after bilateral dorsal hippocampal lesions. During behavior on a familiar cued treadmill, the locations of two obstacles were interchanged to decouple place-tuning from cue-tuning among position-correlated cells with fields at those locations. Subpopulations of place and cue cells each formed interareal gradients such that higher-level cortical regions exhibited higher fractions of place cells, whereas lower-level regions exhibited higher fractions of cue cells. Position-correlated cells in the motor cortex also formed translaminar gradients; more superficial cells were more likely to exhibit fields and were more sparsely and precisely tuned than deeper cells. After dorsal hippocampal lesions, a neural representation of the learned environment persisted, but retrosplenial cortex exhibited significantly increased cue-tuning, and, in motor cortices, both position-correlated cell recruitment and population activity at the unstable obstacle locations became more homogeneously elevated across laminae. Altogether, these results support that the hippocampus continues to modulate cortical responses in familiar environments, and the relative impact of descending feedback obeys hierarchical interareal and interlaminar gradients opposite to the flow of ascending sensory inputs.
Collapse
Affiliation(s)
- Aubrey M Demchuk
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
| | - Ingrid M Esteves
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
| | - HaoRan Chang
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
| | - Jianjun Sun
- Hotchkiss Brain Institute, University of Calgary Foothills, Calgary, Alberta T2N 4N1, Canada
| | - Bruce L McNaughton
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
- Department of Neurobiology and Behaviour, University of California, Irvine, Irvine, California 92697
| |
Collapse
|
8
|
Bouin A, Wu G, Koyuncu OO, Ye Q, Kim KY, Wu MY, Tong L, Chen L, Phan S, Mackey MR, Ramachandra R, Ellisman MH, Holmes TC, Semler BL, Xu X. New rabies viral resources for multi-scale neural circuit mapping. Mol Psychiatry 2024; 29:1951-1967. [PMID: 38355784 PMCID: PMC11322437 DOI: 10.1038/s41380-024-02451-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/16/2024]
Abstract
Comparisons and linkage between multiple imaging scales are essential for neural circuit connectomics. Here, we report 20 new recombinant rabies virus (RV) vectors that we have developed for multi-scale and multi-modal neural circuit mapping tools. Our new RV tools for mesoscale imaging express a range of improved fluorescent proteins. Further refinements target specific neuronal subcellular locations of interest. We demonstrate the discovery power of these new tools including the detection of detailed microstructural changes of rabies-labeled neurons in aging and Alzheimer's disease mouse models, live imaging of neuronal activities using calcium indicators, and automated measurement of infected neurons. RVs that encode GFP and ferritin as electron microscopy (EM) and fluorescence microscopy reporters are used for dual EM and mesoscale imaging. These new viral variants significantly expand the scale and power of rabies virus-mediated neural labeling and circuit mapping across multiple imaging scales in health and disease.
Collapse
Affiliation(s)
- Alexis Bouin
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Ginny Wu
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Orkide O Koyuncu
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Qiao Ye
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Department Biomedical Engineering, University of California, Irvine, CA, 92697, USA
| | - Keun-Young Kim
- The National Center for Microscopy and Imaging Research (NCMIR) and the Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Michele Y Wu
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Liqi Tong
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Lujia Chen
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Department Biomedical Engineering, University of California, Irvine, CA, 92697, USA
| | - Sebastien Phan
- The National Center for Microscopy and Imaging Research (NCMIR) and the Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mason R Mackey
- The National Center for Microscopy and Imaging Research (NCMIR) and the Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ranjan Ramachandra
- The National Center for Microscopy and Imaging Research (NCMIR) and the Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mark H Ellisman
- The National Center for Microscopy and Imaging Research (NCMIR) and the Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Todd C Holmes
- Physiology & Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
- The Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
| | - Bert L Semler
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA, 92697, USA.
- The Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA.
| | - Xiangmin Xu
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Department Biomedical Engineering, University of California, Irvine, CA, 92697, USA.
- The Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
9
|
Yao R, Yamada K, Izawa S, Kito T, Sawada H, Chihara T, Aizu N, Iwata D, Nishii K. FNDC5/irisin mediates the protective effects of Innovative theta-shaking exercise on mouse memory. Heliyon 2024; 10:e29090. [PMID: 38638979 PMCID: PMC11024559 DOI: 10.1016/j.heliyon.2024.e29090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/23/2024] [Accepted: 03/29/2024] [Indexed: 04/20/2024] Open
Abstract
As a passive motion and non-invasive treatment, theta-shaking exercise is considered an alternative to traditional active exercise for slowing down brain ageing. Here, we studied the influence of theta-shaking exercise on fibronectin type III domain containing 5/irisin (FNDC5/irisin) in the anterior nucleus of the thalamus, hippocampus, and medial prefrontal cortex (ATN-HPC-MPFC). Further, we assessed memory in senescence-accelerated prone mice (SAMP-10 mice) using a behavioural test to confirm the protective effect of theta-shaking exercise against age-related memory decline. SAMP-10 mice were subjected to theta-shaking exercise for 9-30 weeks. Mice then performed the T-maze test and passive avoidance task. Immunohistochemical analysis and ELISA were used to assess FNDC5/irisin, nerve growth factor (NGF), and neurotrophin 4/5 (NT4/5) expression in the ATN-HPC-MPFC. In the shaking group, FNDC5 was locally upregulated within the hippocampus and MPFC area rather than exhibiting even distribution throughout brain tissue. Irisin levels were generally higher in the control group. Meanwhile, hippocampal NGF levels were significantly higher in the shaking group, with no differences noted in neurotrophin levels. Theta-shaking preserved normal neurons in certain sub-regions. However, no beneficial changes in neuronal density were noted in the ATN. Theta-shaking exercise positively affects memory function in SAMP-10 mice. FNDC5 upregulation and higher levels of NGF, along with the potential involvement of irisin, may have contributed to the preservation of normal neuronal density in the hippocampus and MPFC subregions.
Collapse
Affiliation(s)
- Runhong Yao
- Physical Therapy Course, Department of Rehabilitation, Faculty of Health Sciences, Nihon Institute of Medical Science, Irumagun, Saitama 350-0435, Japan
| | - Kouji Yamada
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Sho Izawa
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Takumi Kito
- Department of Physical Therapy, Faculty of Health Sciences, Kinjo University, Hakusan, Ishikawa 924-8511, Japan
| | - Hirohide Sawada
- Department of Medical Technology, School of Nursing and Medical Care, Yokkaichi Nursing and Medical Care University, Yokkaichi, Mie 512-8045, Japan
| | - Takeshi Chihara
- Department of Medical Technology, School of Nursing and Medical Care, Yokkaichi Nursing and Medical Care University, Yokkaichi, Mie 512-8045, Japan
| | - Naoki Aizu
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Daiki Iwata
- Department of Medical Technology, School of Nursing and Medical Care, Yokkaichi Nursing and Medical Care University, Yokkaichi, Mie 512-8045, Japan
| | - Kazuhiro Nishii
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
10
|
Ye Q, Gast G, Wilfley EG, Huynh H, Hays C, Holmes TC, Xu X. Monosynaptic Rabies Tracing Reveals Sex- and Age-Dependent Dorsal Subiculum Connectivity Alterations in an Alzheimer's Disease Mouse Model. J Neurosci 2024; 44:e1796232024. [PMID: 38503494 PMCID: PMC11026364 DOI: 10.1523/jneurosci.1796-23.2024] [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: 09/19/2023] [Revised: 02/20/2024] [Accepted: 02/28/2024] [Indexed: 03/21/2024] Open
Abstract
The subiculum (SUB), a hippocampal formation structure, is among the earliest brain regions impacted in Alzheimer's disease (AD). Toward a better understanding of AD circuit-based mechanisms, we mapped synaptic circuit inputs to dorsal SUB using monosynaptic rabies tracing in the 5xFAD mouse model by quantitatively comparing the circuit connectivity of SUB excitatory neurons in age-matched controls and 5xFAD mice at different ages for both sexes. Input-mapped brain regions include the hippocampal subregions (CA1, CA2, CA3), medial septum and diagonal band, retrosplenial cortex, SUB, postsubiculum (postSUB), visual cortex, auditory cortex, somatosensory cortex, entorhinal cortex, thalamus, perirhinal cortex (Prh), ectorhinal cortex, and temporal association cortex. We find sex- and age-dependent changes in connectivity strengths and patterns of SUB presynaptic inputs from hippocampal subregions and other brain regions in 5xFAD mice compared with control mice. Significant sex differences for SUB inputs are found in 5xFAD mice for CA1, CA2, CA3, postSUB, Prh, lateral entorhinal cortex, and medial entorhinal cortex: all of these areas are critical for learning and memory. Notably, we find significant changes at different ages for visual cortical inputs to SUB. While the visual function is not ordinarily considered defective in AD, these specific connectivity changes reflect that altered visual circuitry contributes to learning and memory deficits. Our work provides new insights into SUB-directed neural circuit mechanisms during AD progression and supports the idea that neural circuit disruptions are a prominent feature of AD.
Collapse
Affiliation(s)
- Qiao Ye
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California 92697
- Department of Biomedical Engineering, University of California, Irvine, California 92697
| | - Gocylen Gast
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California 92697
| | - Erik George Wilfley
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California 92697
| | - Hanh Huynh
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California 92697
| | - Chelsea Hays
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California 92697
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California 92697
- Center for Neural Circuit Mapping, University of California, Irvine, California 92697
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California 92697
- Department of Biomedical Engineering, University of California, Irvine, California 92697
- Center for Neural Circuit Mapping, University of California, Irvine, California 92697
| |
Collapse
|
11
|
Zeng YF, Yang KX, Cui Y, Zhu XN, Li R, Zhang H, Wu DC, Stevens RC, Hu J, Zhou N. Conjunctive encoding of exploratory intentions and spatial information in the hippocampus. Nat Commun 2024; 15:3221. [PMID: 38622129 PMCID: PMC11018604 DOI: 10.1038/s41467-024-47570-4] [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: 05/02/2023] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
The hippocampus creates a cognitive map of the external environment by encoding spatial and self-motion-related information. However, it is unclear whether hippocampal neurons could also incorporate internal cognitive states reflecting an animal's exploratory intention, which is not driven by rewards or unexpected sensory stimuli. In this study, a subgroup of CA1 neurons was found to encode both spatial information and animals' investigatory intentions in male mice. These neurons became active before the initiation of exploration behaviors at specific locations and were nearly silent when the same fields were traversed without exploration. Interestingly, this neuronal activity could not be explained by object features, rewards, or mismatches in environmental cues. Inhibition of the lateral entorhinal cortex decreased the activity of these cells during exploration. Our findings demonstrate that hippocampal neurons may bridge external and internal signals, indicating a potential connection between spatial representation and intentional states in the construction of internal navigation systems.
Collapse
Affiliation(s)
- Yi-Fan Zeng
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Ke-Xin Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yilong Cui
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xiao-Na Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Rui Li
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Hanqing Zhang
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
| | - Dong Chuan Wu
- Neuroscience and Brain Disease Center, Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, 404333, Taiwan
- Translational Medicine Research Center, China Medical University Hospital, Taichung City, 404333, Taiwan
| | - Raymond C Stevens
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Ji Hu
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Ning Zhou
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
12
|
Fragueiro A, Cury C, Santacroce F, Burles F, Iaria G, Committeri G. Medial positioning of the hippocampus and hippocampal fissure volume in developmental topographical disorientation. Hippocampus 2024; 34:204-216. [PMID: 38214182 DOI: 10.1002/hipo.23599] [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: 07/25/2023] [Revised: 11/08/2023] [Accepted: 12/18/2023] [Indexed: 01/13/2024]
Abstract
Developmental topographical disorientation (DTD) refers to the lifelong inability to orient by means of cognitive maps in familiar surroundings despite otherwise well-preserved general cognitive functions, and the absence of any acquired brain injury or neurological condition. While reduced functional connectivity between the hippocampus and other brain regions has been reported in DTD individuals, no structural differences in gray matter tissue for the whole brain neither for the hippocampus were detected. Considering that the human hippocampus is the main structure associated with cognitive map-based navigation, here, we investigated differences in morphological and morphometric hippocampal features between individuals affected by DTD (N = 20) and healthy controls (N = 238). Specifically, we focused on a developmental anomaly of the hippocampus that is characterized by the incomplete infolding of hippocampal subfields during fetal development, giving the hippocampus a more round or pyramidal shape, called incomplete hippocampal inversion (IHI). We rated IHI according to standard criteria and extracted hippocampal subfield volumes after FreeSurfer's automatic segmentation. We observed similar IHI prevalence in the group of individuals with DTD with respect to the control population. Neither differences in whole hippocampal nor major hippocampal subfield volumes have been observed between groups. However, when assessing the IHI independent criteria, we observed that the hippocampus in the DTD group is more medially positioned comparing to the control group. In addition, we observed bigger hippocampal fissure volume for the DTD comparing to the control group. Both of these findings were stronger for the right hippocampus comparing to the left. Our results provide new insights regarding the hippocampal morphology of individuals affected by DTD, highlighting the role of structural anomalies during early prenatal development in line with the developmental nature of the spatial disorientation deficit.
Collapse
Affiliation(s)
- Agustina Fragueiro
- Univ Rennes, CNRS, Inria, Inserm, IRISA UMR 6074, Empenn-ERL U 1228, Rennes, France
| | - Claire Cury
- Univ Rennes, CNRS, Inria, Inserm, IRISA UMR 6074, Empenn-ERL U 1228, Rennes, France
| | - Federica Santacroce
- Department of Neuroscience, Imaging and Clinical Sciences, and ITAB, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Ford Burles
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| | - Giuseppe Iaria
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| | - Giorgia Committeri
- Department of Neuroscience, Imaging and Clinical Sciences, and ITAB, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
13
|
Xiao W, Li P, Kong F, Kong J, Pan A, Long L, Yan X, Xiao B, Gong J, Wan L. Unraveling the Neural Circuits: Techniques, Opportunities and Challenges in Epilepsy Research. Cell Mol Neurobiol 2024; 44:27. [PMID: 38443733 PMCID: PMC10914928 DOI: 10.1007/s10571-024-01458-5] [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: 12/25/2023] [Accepted: 01/24/2024] [Indexed: 03/07/2024]
Abstract
Epilepsy, a prevalent neurological disorder characterized by high morbidity, frequent recurrence, and potential drug resistance, profoundly affects millions of people globally. Understanding the microscopic mechanisms underlying seizures is crucial for effective epilepsy treatment, and a thorough understanding of the intricate neural circuits underlying epilepsy is vital for the development of targeted therapies and the enhancement of clinical outcomes. This review begins with an exploration of the historical evolution of techniques used in studying neural circuits related to epilepsy. It then provides an extensive overview of diverse techniques employed in this domain, discussing their fundamental principles, strengths, limitations, as well as their application. Additionally, the synthesis of multiple techniques to unveil the complexity of neural circuits is summarized. Finally, this review also presents targeted drug therapies associated with epileptic neural circuits. By providing a critical assessment of methodologies used in the study of epileptic neural circuits, this review seeks to enhance the understanding of these techniques, stimulate innovative approaches for unraveling epilepsy's complexities, and ultimately facilitate improved treatment and clinical translation for epilepsy.
Collapse
Affiliation(s)
- Wenjie Xiao
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Peile Li
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Fujiao Kong
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jingyi Kong
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Lili Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiaoe Gong
- Department of Neurology, Hunan Children's Hospital, Changsha, Hunan Province, China.
| | - Lily Wan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China.
| |
Collapse
|
14
|
Sun Y, Nitz DA, Xu X, Giocomo LM. Subicular neurons encode concave and convex geometries. Nature 2024; 627:821-829. [PMID: 38448584 PMCID: PMC10972755 DOI: 10.1038/s41586-024-07139-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: 06/15/2023] [Accepted: 01/31/2024] [Indexed: 03/08/2024]
Abstract
Animals in the natural world constantly encounter geometrically complex landscapes. Successful navigation requires that they understand geometric features of these landscapes, including boundaries, landmarks, corners and curved areas, all of which collectively define the geometry of the environment1-12. Crucial to the reconstruction of the geometric layout of natural environments are concave and convex features, such as corners and protrusions. However, the neural substrates that could underlie the perception of concavity and convexity in the environment remain elusive. Here we show that the dorsal subiculum contains neurons that encode corners across environmental geometries in an allocentric reference frame. Using longitudinal calcium imaging in freely behaving mice, we find that corner cells tune their activity to reflect the geometric properties of corners, including corner angles, wall height and the degree of wall intersection. A separate population of subicular neurons encode convex corners of both larger environments and discrete objects. Both corner cells are non-overlapping with the population of subicular neurons that encode environmental boundaries. Furthermore, corner cells that encode concave or convex corners generalize their activity such that they respond, respectively, to concave or convex curvatures within an environment. Together, our findings suggest that the subiculum contains the geometric information needed to reconstruct the shape and layout of naturalistic spatial environments.
Collapse
Affiliation(s)
- Yanjun Sun
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA.
| | - Douglas A Nitz
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, Irvine, CA, USA
| | - Lisa M Giocomo
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
15
|
Han KA, Yoon TH, Kim J, Lee J, Lee JY, Jang G, Um JW, Kim JK, Ko J. Specification of neural circuit architecture shaped by context-dependent patterned LAR-RPTP microexons. Nat Commun 2024; 15:1624. [PMID: 38388459 PMCID: PMC10883964 DOI: 10.1038/s41467-024-45695-0] [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: 07/13/2023] [Accepted: 01/30/2024] [Indexed: 02/24/2024] Open
Abstract
LAR-RPTPs are evolutionarily conserved presynaptic cell-adhesion molecules that orchestrate multifarious synaptic adhesion pathways. Extensive alternative splicing of LAR-RPTP mRNAs may produce innumerable LAR-RPTP isoforms that act as regulatory "codes" for determining the identity and strength of specific synapse signaling. However, no direct evidence for this hypothesis exists. Here, using targeted RNA sequencing, we detected LAR-RPTP mRNAs in diverse cell types across adult male mouse brain areas. We found pronounced cell-type-specific patterns of two microexons, meA and meB, in Ptprd mRNAs. Moreover, diverse neural circuits targeting the same neuronal populations were dictated by the expression of different Ptprd variants with distinct inclusion patterns of microexons. Furthermore, conditional ablation of Ptprd meA+ variants at presynaptic loci of distinct hippocampal circuits impaired distinct modes of synaptic transmission and objection-location memory. Activity-triggered alterations of the presynaptic Ptprd meA code in subicular neurons mediates NMDA receptor-mediated postsynaptic responses in CA1 neurons and objection-location memory. Our data provide the evidence of cell-type- and/or circuit-specific expression patterns in vivo and physiological functions of LAR-RPTP microexons that are dynamically regulated.
Collapse
Affiliation(s)
- Kyung Ah Han
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea
| | - Taek-Han Yoon
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Jinhu Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Jusung Lee
- Department of New Biology, DGIST, Daegu, 42988, Korea
| | - Ju Yeon Lee
- Korea Basic Science Institute, Research Center for Bioconvergence Analysis, Cheongju, 28119, Korea
| | - Gyubin Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea
| | - Ji Won Um
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea
| | - Jong Kyoung Kim
- Department of New Biology, DGIST, Daegu, 42988, Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea.
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea.
| |
Collapse
|
16
|
Suganuma T, Hatori S, Chen CK, Hori S, Kanuka M, Liu CY, Tatsuzawa C, Yanagisawa M, Hayashi Y. Caffeoylquinic Acid Mitigates Neuronal Loss and Cognitive Decline in 5XFAD Mice Without Reducing the Amyloid-β Plaque Burden. J Alzheimers Dis 2024; 99:1285-1301. [PMID: 38788074 DOI: 10.3233/jad-240033] [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] [Indexed: 05/26/2024]
Abstract
Background Caffeoylquinic acid (CQA), which is abundant in coffee beans and Centella asiatica, reportedly improves cognitive function in Alzheimer's disease (AD) model mice, but its effects on neuroinflammation, neuronal loss, and the amyloid-β (Aβ) plaque burden have remained unclear. Objective To assess the effects of a 16-week treatment with CQA on recognition memory, working memory, Aβ levels, neuronal loss, neuroinflammation, and gene expression in the brains of 5XFAD mice, a commonly used mouse model of familial AD. Methods 5XFAD mice at 7 weeks of age were fed a 0.8% CQA-containing diet for 4 months and then underwent novel object recognition (NOR) and Y-maze tests. The Aβ levels and plaque burden were analyzed by enzyme-linked immunosorbent assay and immunofluorescent staining, respectively. Immunostaining of markers of mature neurons, synapses, and glial cells was analyzed. AmpliSeq transcriptome analysis and quantitative reverse-transcription-polymerase chain reaction were performed to assess the effect of CQA on gene expression levels in the cerebral cortex of the 5XFAD mice. Results CQA treatment for 4 months improved recognition memory and ameliorated the reduction of mature neurons and synaptic function-related gene mRNAs. The Aβ levels, plaque burden, and glial markers of neuroinflammation seemed unaffected. Conclusions These findings suggest that CQA treatment mitigates neuronal loss and improves cognitive function without reducing Aβ levels or neuroinflammation. Thus, CQA is a potential therapeutic compound for AD, improving cognitive function via as-yet unknown mechanisms independent of reductions in Aβ or neuroinflammation.
Collapse
Affiliation(s)
- Takaya Suganuma
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Biological Science Research Laboratories, Kao Corporation, Ichikai, Japan
| | - Sena Hatori
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Chung-Kuan Chen
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Satoshi Hori
- Biological Science Research Laboratories, Kao Corporation, Ichikai, Japan
| | - Mika Kanuka
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Chih-Yao Liu
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Chika Tatsuzawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Life Science Center for Survival Dynamics (TARA), University of Tsukuba, Tsukuba, Japan
- R&D Center for Frontiers of Mirai in Policy and Technology (F-MIRAI), University of Tsukuba, Tsukuba, Japan
| | - Yu Hayashi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
de Melo MB, Daldegan-Bueno D, Favaro VM, Oliveira MGM. The subiculum role on learning and memory tasks using rats and mice: A scoping review. Neurosci Biobehav Rev 2023; 155:105460. [PMID: 37939978 DOI: 10.1016/j.neubiorev.2023.105460] [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: 07/13/2023] [Revised: 10/24/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
This scoping review aimed to systematically identify and summarize data related to subiculum involvement in learning and memory behavioral tasks in rats and mice. Following a systematic strategy based on PICO and PRISMA guidelines, we searched five indexed databases (PubMed, Web of Science, EMBASE, Scopus, and PsycInfo) using a standardized search strategy to identify peer-reviewed articles published in English (pre-registration: osf.io/hm5ea). We identified 31 articles investigating the role of the subiculum in spatial, working, and recognition memories (n = 11), memories related to addiction models (n = 9), aversive memories (n = 7), and memories related to appetitive learning (n = 5). We highlight a dissociation in the dorsoventral axis of the subiculum with many studies exploring the ventral subiculum (n = 21) but only a few exploring the dorsal one (n = 10). We also observe the necessity of more data including mice, female animals, genetic tools, and better statistical approaches for replication purposes and research refinement. These findings provide a broad framework of the subiculum involvement in learning and memory, showing essential questions that can be explored by further studies.
Collapse
Affiliation(s)
- Márcio Braga de Melo
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Dimitri Daldegan-Bueno
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Vanessa Manchim Favaro
- Setor de Investigação de Doenças Neuromusculares, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
18
|
Yang L, Zhang Q, Wu XQ, Qiu XY, Fei F, Lai NX, Zheng YY, Zhang MD, Zhang QY, Wang Y, Wang F, Xu CL, Ruan YP, Wang Y, Chen Z. Chemogenetic inhibition of subicular seizure-activated neurons alleviates cognitive deficit in male mouse epilepsy model. Acta Pharmacol Sin 2023; 44:2376-2387. [PMID: 37488426 PMCID: PMC10692337 DOI: 10.1038/s41401-023-01129-z] [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: 04/07/2023] [Accepted: 06/28/2023] [Indexed: 07/26/2023] Open
Abstract
Cognitive deficit is a common comorbidity in temporal lobe epilepsy (TLE) and is not well controlled by current therapeutics. How epileptic seizure affects cognitive performance remains largely unclear. In this study we investigated the role of subicular seizure-activated neurons in cognitive impairment in TLE. A bipolar electrode was implanted into hippocampal CA3 in male mice for kindling stimulation and EEG recording; a special promoter with enhanced synaptic activity-responsive element (E-SARE) was used to label seizure-activated neurons in the subiculum; the activity of subicular seizure-activated neurons was manipulated using chemogenetic approach; cognitive function was assessed in object location memory (OLM) and novel object recognition (NOR) tasks. We showed that chemogenetic inhibition of subicular seizure-activated neurons (mainly CaMKIIα+ glutamatergic neurons) alleviated seizure generalization and improved cognitive performance, but inhibition of seizure-activated GABAergic interneurons had no effect on seizure and cognition. For comparison, inhibition of the whole subicular CaMKIIα+ neuron impaired cognitive function in naïve mice in basal condition. Notably, chemogenetic inhibition of subicular seizure-activated neurons enhanced the recruitment of cognition-responsive c-fos+ neurons via increasing neural excitability during cognition tasks. Our results demonstrate that subicular seizure-activated neurons contribute to cognitive impairment in TLE, suggesting seizure-activated neurons as the potential therapeutic target to alleviate cognitive impairment in TLE.
Collapse
Affiliation(s)
- Lin Yang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qi Zhang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xue-Qing Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiao-Yun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Fan Fei
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310013, China
| | - Nan-Xi Lai
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yu-Yi Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Meng-di Zhang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qing-Yang Zhang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yu Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Fei Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ceng-Lin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ye-Ping Ruan
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310013, China.
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
19
|
Chen YN, Kostka JK, Bitzenhofer SH, Hanganu-Opatz IL. Olfactory bulb activity shapes the development of entorhinal-hippocampal coupling and associated cognitive abilities. Curr Biol 2023; 33:4353-4366.e5. [PMID: 37729915 PMCID: PMC10617757 DOI: 10.1016/j.cub.2023.08.072] [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: 07/21/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/22/2023]
Abstract
The interplay between olfaction and higher cognitive processing has been documented in the adult brain; however, its development is poorly understood. In mice, shortly after birth, endogenous and stimulus-evoked activity in the olfactory bulb (OB) boosts the oscillatory entrainment of downstream lateral entorhinal cortex (LEC) and hippocampus (HP). However, it is unclear whether early OB activity has a long-lasting impact on entorhinal-hippocampal function and cognitive processing. Here, we chemogenetically silenced the synaptic outputs of mitral/tufted cells, the main projection neurons in the OB, during postnatal days 8-10. The transient manipulation leads to a long-lasting reduction of oscillatory coupling and weaker responsiveness to stimuli within developing entorhinal-hippocampal circuits accompanied by dendritic sparsification of LEC pyramidal neurons. Moreover, the transient silencing reduces the performance in behavioral tests involving entorhinal-hippocampal circuits later in life. Thus, neonatal OB activity is critical for the functional LEC-HP development and maturation of cognitive abilities.
Collapse
Affiliation(s)
- Yu-Nan Chen
- Institute of Developmental Neurophysiology, Center of Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Johanna K Kostka
- Institute of Developmental Neurophysiology, Center of Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sebastian H Bitzenhofer
- Institute of Developmental Neurophysiology, Center of Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center of Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| |
Collapse
|
20
|
Grieco SF, Holmes TC, Xu X. Probing neural circuit mechanisms in Alzheimer's disease using novel technologies. Mol Psychiatry 2023; 28:4407-4420. [PMID: 36959497 PMCID: PMC10827671 DOI: 10.1038/s41380-023-02018-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/25/2023]
Abstract
The study of Alzheimer's Disease (AD) has traditionally focused on neuropathological mechanisms that has guided therapies that attenuate neuropathological features. A new direction is emerging in AD research that focuses on the progressive loss of cognitive function due to disrupted neural circuit mechanisms. Evidence from humans and animal models of AD show that dysregulated circuits initiate a cascade of pathological events that culminate in functional loss of learning, memory, and other aspects of cognition. Recent progress in single-cell, spatial, and circuit omics informs this circuit-focused approach by determining the identities, locations, and circuitry of the specific cells affected by AD. Recently developed neuroscience tools allow for precise access to cell type-specific circuitry so that their functional roles in AD-related cognitive deficits and disease progression can be tested. An integrated systems-level understanding of AD-associated neural circuit mechanisms requires new multimodal and multi-scale interrogations that longitudinally measure and/or manipulate the ensemble properties of specific molecularly-defined neuron populations first susceptible to AD. These newly developed technological and conceptual advances present new opportunities for studying and treating circuits vulnerable in AD and represent the beginning of a new era for circuit-based AD research.
Collapse
Affiliation(s)
- Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA
| | - Todd C Holmes
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
21
|
Lin X, Cyrus N, Avila B, Holmes TC, Xu X. Hippocampal CA3 inhibitory neurons receive extensive noncanonical synaptic inputs from CA1 and subicular complex. J Comp Neurol 2023; 531:1333-1347. [PMID: 37312626 PMCID: PMC10525020 DOI: 10.1002/cne.25510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 04/26/2023] [Indexed: 06/15/2023]
Abstract
Hippocampal CA3 is traditionally conceptualized as a brain region within a unidirectional feedforward trisynaptic pathway that links major hippocampal subregions. Recent genomic and viral tracing studies indicate that the anatomical connectivity of CA3 and the trisynaptic pathway is more complex than initially expected and suggests that there may be cell type-specific input gradients throughout the three-dimensional hippocampal structure. In several recent studies using multiple viral tracing approaches, we describe subdivisions of the subiculum complex and ventral hippocampal CA1 that show significant back projections to CA1 and CA3 excitatory neurons. These novel connections form "noncanonical" circuits that run in the opposite direction relative to the well-characterized feedforward pathway. Diverse subtypes of GABAergic inhibitory neurons participate within the trisynaptic pathway. In the present study, we have applied monosynaptic retrograde viral tracing to examine noncanonical synaptic inputs from CA1 and subicular complex to the inhibitory neuron in hippocampal CA3. We quantitatively mapped synaptic inputs to CA3 inhibitory neurons to understand how they are connected within and beyond the hippocampus formation. Major brain regions that provide typical inputs to CA3 inhibitory neurons include the medial septum, the dentate gyrus, the entorhinal cortex, and CA3. Noncanonical inputs from ventral CA1 and subicular complex to CA3 inhibitory neurons follow a proximodistal topographic gradient with regard to CA3 subregions. We find novel noncanonical circuit connections between inhibitory CA3 neurons and ventral CA1, subiculum complex, and other brain regions. These results provide a new anatomical connectivity basis to further study the function of CA3 inhibitory neurons.
Collapse
Affiliation(s)
- Xiaoxiao Lin
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA 92697
| | - Neeyaz Cyrus
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA 92697
| | - Brenda Avila
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA 92697
| | - Todd C. Holmes
- Department Physiology & Biophysics, School of Medicine, University of California, Irvine, CA 92697
| | - Xiangmin Xu
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA 92697
| |
Collapse
|
22
|
Xie B, Zhen Z, Guo O, Li H, Guo M, Zhen J. Progress on the hippocampal circuits and functions based on sharp wave ripples. Brain Res Bull 2023:110695. [PMID: 37353037 DOI: 10.1016/j.brainresbull.2023.110695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Sharp wave ripples (SWRs) are high-frequency synchronization events generated by hippocampal neuronal circuits during various forms of learning and reactivated during memory consolidation and recall. There is mounting evidence that SWRs are essential for storing spatial and social memories in rodents and short-term episodic memories in humans. Sharp wave ripples originate mainly from the hippocampal CA3 and subiculum, and can be transmitted to modulate neuronal activity in cortical and subcortical regions for long-term memory consolidation and behavioral guidance. Different hippocampal subregions have distinct functions in learning and memory. For instance, the dorsal CA1 is critical for spatial navigation, episodic memory, and learning, while the ventral CA1 and dorsal CA2 may work cooperatively to store and consolidate social memories. Here, we summarize recent studies demonstrating that SWRs are essential for the consolidation of spatial, episodic, and social memories in various hippocampal-cortical pathways, and review evidence that SWR dysregulation contributes to cognitive impairments in neurodegenerative and neurodevelopmental diseases.
Collapse
Affiliation(s)
- Boxu Xie
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhihang Zhen
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ouyang Guo
- Department of Biology, Boston University, Boston, MA, United States
| | - Heming Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Moran Guo
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Junli Zhen
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China; Neurological Laboratory of Hebei Province, Shijiazhuang, China.
| |
Collapse
|
23
|
Chen L, Lin X, Ye Q, Nenadic Z, Holmes TC, Nitz DA, Xu X. Anatomical organization of temporally correlated neural calcium activity in the hippocampal CA1 region. iScience 2023; 26:106703. [PMID: 37250317 PMCID: PMC10214731 DOI: 10.1016/j.isci.2023.106703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 09/27/2022] [Accepted: 04/15/2023] [Indexed: 05/31/2023] Open
Abstract
Hippocampal CA1 neuronal ensembles generate sequential patterns of firing activity that contribute to episodic memory formation and spatial cognition. Here we used in vivo calcium imaging to record neural ensemble activities in mouse hippocampal CA1 and identified CA1 excitatory neuron sub-populations whose members are active across the same second-long period of time. We identified groups of hippocampal neurons sharing temporally correlated neural calcium activity during behavioral exploration and found that they also organized as clusters in anatomical space. Such clusters vary in membership and activity dynamics with respect to movement in different environments, but also appear during immobility in the dark suggesting an internal dynamic. The strong covariance between dynamics and anatomical location within the CA1 sub-region reveals a previously unrecognized form of topographic representation in hippocampus that may guide generation of hippocampal sequences across time and therefore organize the content of episodic memory.
Collapse
Affiliation(s)
- Lujia Chen
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697-1275, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697-2715, USA
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Xiaoxiao Lin
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697-1275, USA
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Qiao Ye
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697-1275, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697-2715, USA
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Zoran Nenadic
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697-2715, USA
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Todd C. Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA 92697- 4560, USA
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Douglas A. Nitz
- Department of Cognitive Science, University of California, La Jolla, La Jolla, CA 92093, San Diego
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697-1275, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697-2715, USA
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA 92697-4025, USA
- Department of Computer Science, University of California, Irvine, Irvine, CA 92697-3435, USA
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
24
|
Watkins de Jong L, Nejad MM, Yoon E, Cheng S, Diba K. Optogenetics reveals paradoxical network stabilizations in hippocampal CA1 and CA3. Curr Biol 2023; 33:1689-1703.e5. [PMID: 37023753 PMCID: PMC10175182 DOI: 10.1016/j.cub.2023.03.032] [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: 08/19/2022] [Revised: 02/22/2023] [Accepted: 03/10/2023] [Indexed: 04/08/2023]
Abstract
Recurrent connectivity between excitatory neurons and the strength of feedback from inhibitory neurons are critical determinants of the dynamics and computational properties of neuronal circuits. Toward a better understanding of these circuit properties in regions CA1 and CA3 of the hippocampus, we performed optogenetic manipulations combined with large-scale unit recordings in rats under anesthesia and in quiet waking, using photoinhibition and photoexcitation with different light-sensitive opsins. In both regions, we saw striking paradoxical responses: subsets of cells increased firing during photoinhibition, while other cells decreased firing during photoexcitation. These paradoxical responses were more prominent in CA3 than in CA1, but, notably, CA1 interneurons showed increased firing in response to photoinhibition of CA3. These observations were recapitulated in simulations where we modeled both CA1 and CA3 as inhibition-stabilized networks in which strong recurrent excitation is balanced by feedback inhibition. To directly test the inhibition-stabilized model, we performed large-scale photoinhibition directed at (GAD-Cre) inhibitory cells and found that interneurons in both regions increased firing when photoinhibited, as predicted. Our results highlight the often-paradoxical circuit dynamics that are evidenced during optogenetic manipulations and indicate that, contrary to long-standing dogma, both CA1 and CA3 hippocampal regions display strongly recurrent excitation, which is stabilized through inhibition.
Collapse
Affiliation(s)
- Laurel Watkins de Jong
- Department of Anesthesiology, Michigan Medicine, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA; Department of Psychology, University of Wisconsin-Milwaukee, 2441 E Hartford Ave, Milwaukee, WI 53211, USA
| | | | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, 1301 Beal Avenue, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sen Cheng
- Institute for Neural Computation, Ruhr University Bochum, Universitätsstr. 150, 44801 Bochum, Germany
| | - Kamran Diba
- Department of Anesthesiology, Michigan Medicine, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA; Department of Psychology, University of Wisconsin-Milwaukee, 2441 E Hartford Ave, Milwaukee, WI 53211, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
25
|
Sun Y, Zweifel LS, Holmes TC, Xu X. Whole-brain input mapping of the lateral versus medial anterodorsal bed nucleus of the stria terminalis in the mouse. Neurobiol Stress 2023; 23:100527. [PMID: 36861029 PMCID: PMC9969273 DOI: 10.1016/j.ynstr.2023.100527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The anterior portion of the bed nucleus of the stria terminalis (BNST) modulates fear and stress responses. The anterodorsal BNST (adBNST) can be anatomically subdivided further into the lateral and medial divisions. Although output projections of BNST subregions have been studied, the local and global input connections to these subregions remain poorly understood. To further understand BNST-centered circuit operations, we have applied new viral-genetic tracing and functional circuit mapping to determine detailed synaptic circuit inputs to lateral and medial subregions of adBNST in the mouse. Monosynaptic canine adenovirus type 2 (CAV2) and rabies virus-based retrograde tracers were injected in the adBNST subregions. The amygdalar complex, hypothalamus and hippocampal formation account for the majority of overall inputs to adBNST. However, lateral versus medial adBNST subregions have distinct patterns of long-range cortical and limbic brain inputs. The lateral adBNST has more input connections from prefrontal (prelimbic, infralimbic, cingulate) and insular cortices, anterior thalamus and ectorhinal/perirhinal cortices. In contrast, the medial adBNST received biased inputs from the medial amygdala, lateral septum, hypothalamus nuclei and ventral subiculum. We confirmed long-range functional inputs from the amydalohippocampal area and basolateral amygdala to the adBNST using ChR2-assisted circuit mapping. Selected novel BNST inputs are also validated with the AAV axonal tracing data from the Allen Institute Mouse Brain Connectivity Atlas. Together, these results provide a comprehensive map of the differential afferent inputs to lateral and medial adBNST subregions, and offer new insight into the functional operations of BNST circuitry for stress and anxiety-related behaviors.
Collapse
Affiliation(s)
- Yanjun Sun
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697-1275, USA
| | - Larry S. Zweifel
- Department of Psychiatry and Behavioral Sciences and Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA
| | - Todd C. Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697-4560, USA
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697-1275, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697-1275, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697-2715, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA, 92697-2715, USA
- Department of Computer Science, University of California, Irvine, CA, 92697-3435, USA
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697-1275, USA
| |
Collapse
|
26
|
Xiao Y, Deng P, Zhao Y, Yang S, Li B. Three-photon excited fluorescence imaging in neuroscience: From principles to applications. Front Neurosci 2023; 17:1085682. [PMID: 36891460 PMCID: PMC9986337 DOI: 10.3389/fnins.2023.1085682] [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: 10/31/2022] [Accepted: 02/02/2023] [Indexed: 02/22/2023] Open
Abstract
The development of three-photon microscopy (3PM) has greatly expanded the capability of imaging deep within biological tissues, enabling neuroscientists to visualize the structure and activity of neuronal populations with greater depth than two-photon imaging. In this review, we outline the history and physical principles of 3PM technology. We cover the current techniques for improving the performance of 3PM. Furthermore, we summarize the imaging applications of 3PM for various brain regions and species. Finally, we discuss the future of 3PM applications for neuroscience.
Collapse
Affiliation(s)
| | | | | | | | - Bo Li
- State Key Laboratory of Medical Neurobiology, Department of Neurology, Ministry of Education (MOE), Frontiers Center for Brain Science, Institute for Translational Brain Research, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Tactile cues are important to environmental novelty during repeated open field tests. Behav Processes 2023; 204:104796. [PMID: 36460136 DOI: 10.1016/j.beproc.2022.104796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 11/16/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022]
Abstract
The open field test (OFT) is a commonly used protocol to measure anxiety-like behaviors in rodents. Exploration in the central area of the open field and rearing frequency are often readouts of anxiety measurement. However, concerns about carry-over effects associated with repeated assessments limit its application, with the underlying mechanisms of this phenomenon still to be fully described. Here, we showed that repeated OFTs in the same mice led to reductions in the percentage of time spent in the central area and frequency of rearing. This effect reduced with an increase in the intervals between test. The decay caused by repeated OFTs was due to habituation, rather than frequent handling of the experimenter, since novel environments could prevent decay from repeated OFTs. Our results also indicated that tactile cues of the environment played important roles in the habituation of repeated OFTs. Furthermore, the decay of central area activity and rearing behavior during repeated OFTs would be blocked if the hippocampal CA1 was lesioned, suggesting that CA1 is a crucial region for habituation of the OFT in mice. Taken together, our study uncovers the important roles of tactile cues and hippocampal CA1 during repeated OFTs in mice.
Collapse
|
28
|
Zhang H, Chen L, Johnston KG, Crapser J, Green KN, Ha NML, Tenner AJ, Holmes TC, Nitz DA, Xu X. Degenerate mapping of environmental location presages deficits in object-location encoding and memory in the 5xFAD mouse model for Alzheimer's disease. Neurobiol Dis 2023; 176:105939. [PMID: 36462718 PMCID: PMC10187684 DOI: 10.1016/j.nbd.2022.105939] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/08/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022] Open
Abstract
A key challenge in developing diagnosis and treatments for Alzheimer's disease (AD) is to detect abnormal network activity at as early a stage as possible. To date, behavioral and neurophysiological investigations in AD model mice have yet to conduct a longitudinal assessment of cellular pathology, memory deficits, and neurophysiological correlates of neuronal activity. We therefore examined the temporal relationships between pathology, neuronal activities and spatial representation of environments, as well as object location memory deficits across multiple stages of development in the 5xFAD mice model and compared these results to those observed in wild-type mice. We performed longitudinal in vivo calcium imaging with miniscope on hippocampal CA1 neurons in behaving mice. We find that 5xFAD mice show amyloid plaque accumulation, depressed neuronal calcium activity during immobile states, and degenerate and unreliable hippocampal neuron spatial tuning to environmental location at early stages by 4 months of age while their object location memory (OLM) is comparable to WT mice. By 8 months of age, 5xFAD mice show deficits of OLM, which are accompanied by progressive degradation of spatial encoding and, eventually, impaired CA1 neural tuning to object-location pairings. Furthermore, depressed neuronal activity and unreliable spatial encoding at early stage are correlated with impaired performance in OLM at 8-month-old. Our results indicate the close connection between impaired hippocampal tuning to object-location and the presence of OLM deficits. The results also highlight that depressed baseline firing rates in hippocampal neurons during immobile states and unreliable spatial representation precede object memory deficits and predict memory deficits at older age, suggesting potential early opportunities for AD detecting.
Collapse
Affiliation(s)
- Hai Zhang
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, United States of America
| | - Lujia Chen
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, United States of America; Department of Biomedical Engineering, University of California, Irvine, CA 92697, United States of America
| | - Kevin G Johnston
- Department of Mathematics, University of California, Irvine, CA 92697, United States of America
| | - Joshua Crapser
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, CA 92697, United States of America
| | - Kim N Green
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, CA 92697, United States of America; Center for Neural Circuit Mapping, University of California, Irvine, CA 92697, United States of America
| | - Nicole My-Linh Ha
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, United States of America
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, CA 92697, United States of America
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA 92697, United States of America; Center for Neural Circuit Mapping, University of California, Irvine, CA 92697, United States of America
| | - Douglas A Nitz
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA 92093, United States of America; Center for Neural Circuit Mapping, University of California, Irvine, CA 92697, United States of America.
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, United States of America; Department of Biomedical Engineering, University of California, Irvine, CA 92697, United States of America; Center for Neural Circuit Mapping, University of California, Irvine, CA 92697, United States of America.
| |
Collapse
|
29
|
Honoré E, Lacaille JC. Object location learning in mice requires hippocampal somatostatin interneuron activity and is facilitated by mTORC1-mediated long-term potentiation of their excitatory synapses. Mol Brain 2022; 15:101. [PMID: 36544185 PMCID: PMC9769025 DOI: 10.1186/s13041-022-00988-7] [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: 10/18/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Hippocampus-dependent learning and memory originate from long-term synaptic changes in hippocampal networks. The activity of CA1 somatostatin interneurons (SOM-INs) during aversive stimulation is necessary for contextual fear memory formation. In addition, mTORC1-dependent long-term potentiation (LTP) of SOM-IN excitatory input synapses from local pyramidal cells (PC-SOM synapses) contributes to the consolidation of fear motivated spatial and contextual memories. Although, it remains unknown if SOM-IN activity and LTP are necessary and sufficient for novelty motivated spatial episodic memory such as the object location memory, and if so when it is required. Here we use optogenetics to examine whether dorsal CA1 SOM-IN activity and LTP are sufficient to regulate object location memory. First, we found that silencing SOM-INs during object location learning impaired memory. Second, optogenetic induction of PC-SOM synapse LTP (TBSopto) given 30 min before object location training, resulted in facilitation of memory. However, in mice with mTORC1 pathway genetically inactivated in SOM-INs, which blocks PC-SOM synapse LTP, TBSopto failed to facilitate object location memory. Our results indicate that SOM-IN activity is necessary during object location learning and that optogenetic induction of PC-SOM synapse LTP is sufficient to facilitate consolidation of object location memory. Thus, hippocampal somatostatin interneuron activity is required for object location learning, a hippocampus-dependent form of novelty motivated spatial learning that is facilitated by plasticity at PC-SOM synapses.
Collapse
Affiliation(s)
- Eve Honoré
- grid.14848.310000 0001 2292 3357Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, P.O. Box 6128, Station Downtown, QC H3C 3J7 Montreal, Canada
| | - Jean-Claude Lacaille
- grid.14848.310000 0001 2292 3357Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, P.O. Box 6128, Station Downtown, QC H3C 3J7 Montreal, Canada
| |
Collapse
|
30
|
Tan Z, Garduño BM, Aburto PF, Chen L, Ha N, Cogram P, Holmes TC, Xu X. Cognitively impaired aged Octodon degus recapitulate major neuropathological features of sporadic Alzheimer's disease. Acta Neuropathol Commun 2022; 10:182. [PMID: 36529803 PMCID: PMC9761982 DOI: 10.1186/s40478-022-01481-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/20/2022] [Indexed: 12/23/2022] Open
Abstract
The long-lived Chilean rodent (Octodon degus) has been reported to show spontaneous age-dependent neuropathology and cognitive impairments similar to those observed in human AD. However, the handful of published papers on degus of differing genetic backgrounds yield inconsistent findings about sporadic AD-like pathological features, with notably differing results between lab in-bred degus versus outbred degus. This motivates more extensive characterization of spontaneously occurring AD-like pathology and behavior in degus. In the present study, we show AD-like neuropathological markers in the form of amyloid deposits and tau abnormalities in a cognitively impaired subset of aged outbred degus. Compared to the aged degus that show normal burrowing behavior, the age-matched degus with burrowing behavior deficits correlatively exhibit detectable human AD-like Aβ deposits and tau neuropathology, along with neuroinflammatory markers that include enhanced microglial activation and higher numbers of reactive astrocytes in the brain. This subset of cognitively impaired aged degus also exhibits cerebral amyloid angiopathy and tauopathy. We find robust neurodegenerative features in behaviorally deficient aged degus, including hippocampal neuronal loss, altered parvalbumin and perineuronal net staining in the cortex, and increased c-Fos neuronal activation in the cortex that is consistent with the neural circuit hyperactivity reported in human AD patients. By focusing on the subset of aged degus that show AD-like behavioral deficits and correlative neuropathology, our findings establish outbred degus as a natural model of sporadic AD and demonstrate the potential importance of wild-type outbred genetic backgrounds for AD pathogenesis.
Collapse
Affiliation(s)
- Zhiqun Tan
- Department Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- The Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
| | - B Maximiliano Garduño
- Department Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Pedro Fernández Aburto
- Institute of Ecology and Biodiversity, Department of Ecological Sciences, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Lujia Chen
- Department Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Nicole Ha
- Department Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Patricia Cogram
- Institute of Ecology and Biodiversity, Department of Ecological Sciences, Faculty of Sciences, University of Chile, Santiago, Chile
- The Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
| | - Todd C Holmes
- Department Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
- The Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- The Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
31
|
Sun Y, Giocomo LM. Neural circuit dynamics of drug-context associative learning in the mouse hippocampus. Nat Commun 2022; 13:6721. [PMID: 36344498 PMCID: PMC9640587 DOI: 10.1038/s41467-022-34114-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022] Open
Abstract
The environmental context associated with previous drug consumption is a potent trigger for drug relapse. However, the mechanism by which neural representations of context are modified to incorporate information associated with drugs of abuse remains unknown. Using longitudinal calcium imaging in freely behaving mice, we find that unlike the associative learning of natural reward, drug-context associations for psychostimulants and opioids are encoded in a specific subset of hippocampal neurons. After drug conditioning, these neurons weakened their spatial coding for the non-drug paired context, resulting in an orthogonal representation for the drug versus non-drug context that was predictive of drug-seeking behavior. Furthermore, these neurons were selected based on drug-spatial experience and were exclusively tuned to animals' allocentric position. Together, this work reveals how drugs of abuse alter the hippocampal circuit to encode drug-context associations and points to the possibility of targeting drug-associated memory in the hippocampus.
Collapse
Affiliation(s)
- Yanjun Sun
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Lisa M Giocomo
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
32
|
Ye Q, Gast G, Su X, Saito T, Saido TC, Holmes TC, Xu X. Hippocampal neural circuit connectivity alterations in an Alzheimer's disease mouse model revealed by monosynaptic rabies virus tracing. Neurobiol Dis 2022; 172:105820. [PMID: 35843448 PMCID: PMC9482455 DOI: 10.1016/j.nbd.2022.105820] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/27/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with growing major health impacts, particularly in countries with aging populations. The examination of neural circuit mechanisms in AD mouse models is a recent focus for identifying new AD treatment strategies. We hypothesize that age-progressive changes of both long-range and local hippocampal neural circuit connectivity occur in AD. Recent advancements in viral-genetic technologies provide new opportunities for semi-quantitative mapping of cell-type-specific neural circuit connections in AD mouse models. We applied a recently developed monosynaptic rabies tracing method to hippocampal neural circuit mapping studies in AD model mice to determine how local and global circuit connectivity to hippocampal CA1 excitatory neurons may be altered in the single APP knock-in (APP-KI) AD mouse model. To determine age-related AD progression, we measured circuit connectivity in age-matched littermate control and AD model mice at two different ages (3-4 vs. 10-11 months old). We quantitatively mapped the connectivity strengths of neural circuit inputs to hippocampal CA1 excitatory neurons from brain regions including hippocampal subregions, medial septum, subiculum and entorhinal cortex, comparing different age groups and genotypes. We focused on hippocampal CA1 because of its clear relationship with learning and memory and that the hippocampal formation shows clear neuropathological changes in human AD. Our results reveal alterations in circuit connectivity of hippocampal CA1 in AD model mice. Overall, we find weaker extrinsic CA1 input connectivity strengths in AD model mice compared with control mice, including sex differences of reduced subiculum to CA1 inputs in aged female AD mice compared with aged male AD mice. Unexpectedly, we find a connectivity pattern shift with an increased proportion of inputs from the CA3 region to CA1 excitatory neurons when comparing young and old AD model mice, as well as old wild-type mice and old AD model mice. These unexpected shifts in CA3-CA1 input proportions in this AD mouse model suggest the possibility that compensatory circuit increases may occur in response to connectivity losses in other parts of the hippocampal circuits. We expect that this work provides new insights into the neural circuit mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Qiao Ye
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA.
| | - Gocylen Gast
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA.
| | - Xilin Su
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA.
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan; Lab for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0106, Japan.
| | - Takaomi C Saido
- Lab for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0106, Japan.
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA 92697, USA; Center for Neural Circuit Mapping, University of California, Irvine, CA 92697, USA.
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA; Center for Neural Circuit Mapping, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
33
|
Chao OY, Nikolaus S, Yang YM, Huston JP. Neuronal circuitry for recognition memory of object and place in rodent models. Neurosci Biobehav Rev 2022; 141:104855. [PMID: 36089106 PMCID: PMC10542956 DOI: 10.1016/j.neubiorev.2022.104855] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022]
Abstract
Rats and mice are used for studying neuronal circuits underlying recognition memory due to their ability to spontaneously remember the occurrence of an object, its place and an association of the object and place in a particular environment. A joint employment of lesions, pharmacological interventions, optogenetics and chemogenetics is constantly expanding our knowledge of the neural basis for recognition memory of object, place, and their association. In this review, we summarize current studies on recognition memory in rodents with a focus on the novel object preference, novel location preference and object-in-place paradigms. The evidence suggests that the medial prefrontal cortex- and hippocampus-connected circuits contribute to recognition memory for object and place. Under certain conditions, the striatum, medial septum, amygdala, locus coeruleus and cerebellum are also involved. We propose that the neuronal circuitry for recognition memory of object and place is hierarchically connected and constructed by different cortical (perirhinal, entorhinal and retrosplenial cortices), thalamic (nucleus reuniens, mediodorsal and anterior thalamic nuclei) and primeval (hypothalamus and interpeduncular nucleus) modules interacting with the medial prefrontal cortex and hippocampus.
Collapse
Affiliation(s)
- Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Susanne Nikolaus
- Department of Nuclear Medicine, University Hospital Düsseldorf, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Joseph P Huston
- Center for Behavioral Neuroscience, Institute of Experimental Psychology, Heinrich-Heine University, 40225 Düsseldorf, Germany.
| |
Collapse
|
34
|
Mizuseki K, Kitanishi T. Oscillation-coordinated, noise-resistant information distribution via the subiculum. Curr Opin Neurobiol 2022; 75:102556. [DOI: 10.1016/j.conb.2022.102556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 11/03/2022]
|
35
|
Lin X, Chen L, Jullienne A, Zhang H, Salehi A, Hamer M, C. Holmes T, Obenaus A, Xu X. Longitudinal dynamics of microvascular recovery after acquired cortical injury. Acta Neuropathol Commun 2022; 10:59. [PMID: 35468870 PMCID: PMC9036719 DOI: 10.1186/s40478-022-01361-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/05/2022] [Indexed: 01/04/2023] Open
Abstract
Acquired brain injuries due to trauma damage the cortical vasculature, which in turn impairs blood flow to injured tissues. There are reports of vascular morphological recovery following traumatic brain injury, but the remodeling process has not been examined longitudinally in detail after injury in vivo. Understanding the dynamic processes that influence recovery is thus critically important. We evaluated the longitudinal and dynamic microvascular recovery and remodeling up to 2 months post injury using live brain miniscope and 2-photon microscopic imaging. The new imaging approaches captured dynamic morphological and functional recovery processes at high spatial and temporal resolution in vivo. Vessel painting documented the initial loss and subsequent temporal morphological vascular recovery at the injury site. Miniscopes were used to longitudinally image the temporal dynamics of vascular repair in vivo after brain injury in individual mice across each cohort. We observe near-immediate nascent growth of new vessels in and adjacent to the injury site that peaks between 14 and 21 days post injury. 2-photon microscopy confirms new vascular growth and further demonstrates differences between cortical layers after cortical injury: large vessels persist in deeper cortical layers (> 200 μm), while superficial layers exhibit a dense plexus of fine (and often non-perfused) vessels displaying regrowth. Functionally, blood flow increases mirror increasing vascular density. Filopodia development and endothelial sprouting is measurable within 3 days post injury that rapidly transforms regions devoid of vessels to dense vascular plexus in which new vessels become increasingly perfused. Within 7 days post injury, blood flow is observed in these nascent vessels. Behavioral analysis reveals improved vascular modulation after 9 days post injury, consistent with vascular regrowth. We conclude that morphological recovery events are closely linked to functional recovery of blood flow to the compromised tissues, which subsequently leads to improved behavioral outcomes.
Collapse
|
36
|
López-Aranda MF, Boxx GM, Phan M, Bach K, Mandanas R, Herrera I, Taloma S, Thadani C, Lu O, Bui R, Liu S, Li N, Zhou Y, Cheng G, Silva AJ. Role of type I interferon signaling and microglia in the abnormal long term potentiation and object place recognition deficits of male mice with a mutation of the Tuberous Sclerosis 2 gene. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022. [PMID: 37519458 PMCID: PMC10382699 DOI: 10.1016/j.bpsgos.2022.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background Tuberous sclerosis complex is a genetic disorder associated with high rates of intellectual disability and autism. Mice with a heterozygous null mutation of the Tsc2 gene (Tsc2+/-) show deficits in hippocampal-dependent tasks and abnormal long-term potentiation (LTP) in the hippocampal CA1 region. Although previous studies focused on the role of neuronal deficits in the memory phenotypes of rodent models of tuberous sclerosis complex, the results presented here demonstrate a role for microglia in these deficits. Methods To test the possible role of microglia and type I interferon in abnormal hippocampal-dependent memory and LTP of Tsc2+/- mice, we used field recordings in CA1 and the object place recognition (OPR) task. We used the colony stimulating factor 1 receptor inhibitor PLX5622 to deplete microglia in Tsc2+/- mice and interferon alpha/beta receptor alpha chain null mutation (Ifnar1-/-) to manipulate a signaling pathway known to modulate microglia function. Results Unexpectedly, we demonstrate that male, but not female, Tsc2+/- mice show OPR deficits. These deficits can be rescued by depletion of microglia and by the Ifnar1-/- mutation. In addition to rescuing OPR deficits, depletion of microglia also reversed abnormal LTP of the Tsc2+/- mice. Altogether, our results suggest that altered IFNAR1 signaling in microglia causes the abnormal LTP and OPR deficits of male Tsc2+/- mice. Conclusions Microglia and IFNAR1 signaling have a key role in the hippocampal-dependent memory deficits and abnormal hippocampal LTP of Tsc2+/- male mice.
Collapse
|
37
|
Ouyang M, Zhang Q, Shu J, Wang Z, Fan J, Yu K, Lei L, Li Y, Wang Q. Capsaicin Ameliorates the Loosening of Mitochondria-Associated Endoplasmic Reticulum Membranes and Improves Cognitive Function in Rats With Chronic Cerebral Hypoperfusion. Front Cell Neurosci 2022; 16:822702. [PMID: 35370565 PMCID: PMC8968035 DOI: 10.3389/fncel.2022.822702] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/10/2022] [Indexed: 12/17/2022] Open
Abstract
Based on accumulating evidence, vascular factors contribute to cognitive decline and dementia. Mitochondrial dysfunction is the core pathophysiological mechanism. Mitochondria-associated endoplasmic reticulum membranes (MAMs) are subcellular structures that physically and biologically connect mitochondria with the endoplasmic reticulum (ER) and regulate multiple functions ranging from calcium transfer to mitochondrial dynamics and bioenergetics. MAMs dysfunction has been speculated to be a key factor contributing to the pathogenesis of cognitive disorders and a new therapeutic target. However, the alteration of MAMs in vascular cognitive impairment remains to be revealed. Capsaicin, a specific agonist known to activated the transient receptor potential vanilloid type 1 (TRPV1), is involved in hippocampal synaptic plasticity and memory, but the detailed mechanism is still unclear. In this study, chronic cerebral hypoperfusion (CCH) model rats were created by bilateral common carotid artery occlusion (BCCAO), which is a widely used model to study vascular dementia. We observed that CCH rats showed obvious cognitive deficits, and ER-mitochondria contacts were loosener with lower expression of mitofusin2 (MFN2), a key protein connecting MAMs, in the hippocampal CA1 region, compared to the sham group. After capsaicin treatment for 12 weeks, we found that cognitive deficits induced by CCH were significantly alleviated and loosened ER-mitochondrial interactions were obviously improved. In conclusion, the findings of this study highlight that MAMs may contribute to the pathogenesis of cognitive impairment induced by CCH, and our new evidence that capsaicin improves cognitive function highlights a novel opportunity for drug discovery.
Collapse
Affiliation(s)
- Mengqi Ouyang
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Qi Zhang
- Department of Pharmacology, Gaoping District People’s Hospital of Nanchong, Nanchong, China
| | - Jiahui Shu
- Department of Pharmacology, Yichang Yiling Hospital, Yichang, China
| | - Zhiqiang Wang
- Department of Neurology, Chengdu BOE Hospital, Chengdu, China
| | - Jin Fan
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Ke Yu
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Lei Lei
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Yuxia Li
- Department of Neurology, Chengdu BOE Hospital, Chengdu, China
| | - Qingsong Wang
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
- *Correspondence: Qingsong Wang,
| |
Collapse
|
38
|
Acetylcholine deficiency disrupts extratelencephalic projection neurons in the prefrontal cortex in a mouse model of Alzheimer's disease. Nat Commun 2022; 13:998. [PMID: 35194025 PMCID: PMC8863829 DOI: 10.1038/s41467-022-28493-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 01/25/2022] [Indexed: 11/21/2022] Open
Abstract
Short-term memory deficits have been associated with prefrontal cortex (PFC) dysfunction in Alzheimer’s disease (AD) and AD mouse models. Extratelencephalic projection (ET) neurons in the PFC play a key role in short-term working memory, but the mechanism between ET neuronal dysfunction in the PFC and short-term memory impairment in AD is not well understood. Here, using fiber photometry and optogenetics, we found reduced neural activity in the ET neurons in the medial prefrontal cortex (mPFC) of the 5×FAD mouse model led to object recognition memory (ORM) deficits. Activation of ET neurons in the mPFC of 5×FAD mice rescued ORM impairment, and inhibition of ET neurons in the mPFC of wild type mice impaired ORM expression. ET neurons in the mPFC that project to supramammillary nucleus were necessary for ORM expression. Viral tracing and in vivo recording revealed that mPFC ET neurons received fewer cholinergic inputs from the basal forebrain in 5×FAD mice. Furthermore, activation of cholinergic fibers in the mPFC rescued ORM deficits in 5×FAD mice, while acetylcholine deficiency reduced the response of ET neurons in the mPFC to familiar objects. Taken together, our results revealed a neural mechanism behind ORM impairment in 5×FAD mice. Short-term memory deficits are associated with prefrontal cortex dysfunction in Alzheimer’s disease. Here, the authors assessed extratelencephalic projection (ET) neurons and found reduced ET neural activity in the medial prefrontal cortex (mPFC) and showed ET neurons received fewer cholinergic inputs from the basal forebrain in 5×FAD mice which led to object recognition memory deficits.
Collapse
|
39
|
Sánchez-Bellot C, AlSubaie R, Mishchanchuk K, Wee RWS, MacAskill AF. Two opposing hippocampus to prefrontal cortex pathways for the control of approach and avoidance behaviour. Nat Commun 2022; 13:339. [PMID: 35039510 PMCID: PMC8763938 DOI: 10.1038/s41467-022-27977-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/13/2021] [Indexed: 11/09/2022] Open
Abstract
The decision to either approach or avoid a potentially threatening environment is thought to rely upon the coordinated activity of heterogeneous neural populations in the hippocampus and prefrontal cortex (PFC). However, how this circuitry is organized to flexibly promote both approach or avoidance at different times has remained elusive. Here, we show that the hippocampal projection to PFC is composed of two parallel circuits located in the superficial or deep pyramidal layers of the CA1/subiculum border. These circuits have unique upstream and downstream connectivity, and are differentially active during approach and avoidance behaviour. The superficial population is preferentially connected to widespread PFC inhibitory interneurons, and its activation promotes exploration; while the deep circuit is connected to PFC pyramidal neurons and fast spiking interneurons, and its activation promotes avoidance. Together this provides a mechanism for regulation of behaviour during approach avoidance conflict: through two specialized, parallel circuits that allow bidirectional hippocampal control of PFC.
Collapse
Affiliation(s)
- Candela Sánchez-Bellot
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St, London, WC1E 6BT, UK
| | - Rawan AlSubaie
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St, London, WC1E 6BT, UK
| | - Karyna Mishchanchuk
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St, London, WC1E 6BT, UK
| | - Ryan W S Wee
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St, London, WC1E 6BT, UK
| | - Andrew F MacAskill
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St, London, WC1E 6BT, UK.
| |
Collapse
|
40
|
Lin X, Chen L, Baglietto-Vargas D, Kamalipour P, Ye Q, LaFerla FM, Nitz DA, Holmes TC, Xu X. Spatial coding defects of hippocampal neural ensemble calcium activities in the triple-transgenic Alzheimer's disease mouse model. Neurobiol Dis 2022; 162:105562. [PMID: 34838667 PMCID: PMC9482454 DOI: 10.1016/j.nbd.2021.105562] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/24/2021] [Accepted: 11/22/2021] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) causes progressive age-related defects in memory and cognitive function and has emerged as a major health and socio-economic concern in the US and worldwide. To develop effective therapeutic treatments for AD, we need to better understand the neural mechanisms by which AD causes memory loss and cognitive deficits. Here we examine large-scale hippocampal neural population calcium activities imaged at single cell resolution in a triple-transgenic Alzheimer's disease mouse model (3xTg-AD) that presents both amyloid plaque and neurofibrillary pathological features along with age-related behavioral defects. To measure encoding of environmental location in hippocampal neural ensembles in the 3xTg-AD mice in vivo, we performed GCaMP6-based calcium imaging using head-mounted, miniature fluorescent microscopes ("miniscopes") on freely moving animals. We compared hippocampal CA1 excitatory neural ensemble activities during open-field exploration and track-based route-running behaviors in age-matched AD and control mice at young (3-6.5 months old) and old (18-21 months old) ages. During open-field exploration, 3xTg-AD CA1 excitatory cells display significantly higher calcium activity rates compared with Non-Tg controls for both the young and old age groups, suggesting that in vivo enhanced neuronal calcium ensemble activity is a disease feature. CA1 neuronal populations of 3xTg-AD mice show lower spatial information scores compared with control mice. The spatial firing of CA1 neurons of old 3xTg-AD mice also displays higher sparsity and spatial coherence, indicating less place specificity for spatial representation. We find locomotor speed significantly modulates the amplitude of hippocampal neural calcium ensemble activities to a greater extent in 3xTg-AD mice during open field exploration. Our data offer new and comprehensive information about age-dependent neural circuit activity changes in this important AD mouse model and provide strong evidence that spatial coding defects in the neuronal population activities are associated with AD pathology and AD-related memory behavioral deficits.
Collapse
Affiliation(s)
- Xiaoxiao Lin
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, United States of America
| | - Lujia Chen
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, United States of America; Department of Biomedical Engineering, University of California, Irvine, CA 92697, United States of America
| | - David Baglietto-Vargas
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Facultad de Ciencias, Universidad de Malaga, Malaga 29071, Spain; Institute for Memory Impairments and Neurological Disorder, University of California, Irvine, CA 92697, United States of America
| | - Parsa Kamalipour
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, United States of America
| | - Qiao Ye
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, United States of America; Department of Biomedical Engineering, University of California, Irvine, CA 92697, United States of America
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorder, University of California, Irvine, CA 92697, United States of America
| | - Douglas A Nitz
- Department of Cognitive Science, University of California, San Diego, CA 92093, United States of America; Center for Neural Circuit Mapping, University of California, Irvine, CA 92697, United States of America
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA 92697, United States of America; Center for Neural Circuit Mapping, University of California, Irvine, CA 92697, United States of America
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, United States of America; Institute for Memory Impairments and Neurological Disorder, University of California, Irvine, CA 92697, United States of America; Department of Biomedical Engineering, University of California, Irvine, CA 92697, United States of America; Center for Neural Circuit Mapping, University of California, Irvine, CA 92697, United States of America.
| |
Collapse
|
41
|
Lee SM, Seol JM, Lee I. Subicular neurons represent multiple variables of a hippocampal-dependent task by using theta rhythm. PLoS Biol 2022; 20:e3001546. [PMID: 35100261 PMCID: PMC8830791 DOI: 10.1371/journal.pbio.3001546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 02/10/2022] [Accepted: 01/18/2022] [Indexed: 01/31/2023] Open
Abstract
The subiculum is positioned at a critical juncture at the interface of the hippocampus with the rest of the brain. However, the exact roles of the subiculum in most hippocampal-dependent memory tasks remain largely unknown. One obstacle to make comparisons of neural firing patterns between the subiculum and hippocampus is the broad firing fields of the subicular cells. Here, we used spiking phases in relation to theta rhythm to parse the broad firing field of a subicular neuron into multiple subfields to find the unique functional contribution of the subiculum while male rats performed a hippocampal-dependent visual scene memory task. Some of the broad firing fields of the subicular neurons were successfully divided into multiple subfields similar to those in the CA1 by using the theta phase precession cycle. The new paradigm significantly improved the detection of task-relevant information in subicular cells without affecting the information content represented by CA1 cells. Notably, we found that multiple fields of a single subicular neuron, unlike those in the CA1, carried heterogeneous task-related information such as visual context and choice response. Our findings suggest that the subicular cells integrate multiple task-related factors by using theta rhythm to associate environmental context with action.
Collapse
Affiliation(s)
- Su-Min Lee
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Korea
| | - Jae-Min Seol
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Korea
| | - Inah Lee
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
42
|
Ahmed OJ. Mechanisms of Subiculum Hyperexcitability in Temporal Lobe Epilepsy. Epilepsy Curr 2021; 21:441-443. [PMID: 34924852 PMCID: PMC8652315 DOI: 10.1177/15357597211048601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
43
|
Guo L, Du QQ, Cheng PQ, Yang TT, Xing CQ, Luo XZ, Peng XC, Qian F, Huang JR, Tang FR. Neuroprotective Effects of Lycium barbarum Berry on Neurobehavioral Changes and Neuronal Loss in the Hippocampus of Mice Exposed to Acute Ionizing Radiation. Dose Response 2021; 19:15593258211057768. [PMID: 34887716 PMCID: PMC8649475 DOI: 10.1177/15593258211057768] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Brain exposure to ionizing radiation during the
radiotherapy of brain tumor or metastasis of peripheral cancer cells to the
brain has resulted in cognitive dysfunction by reducing neurogenesis in
hippocampus. The water extract of Lycium barbarum berry (Lyc),
containing water-soluble Lycium barbarum polysaccharides and
flavonoids, can protect the neuronal injury by reducing oxidative stress and
suppressing neuroinflammation. Reseach Design: To demonstrate the long-term radioprotective effect
of Lyc, we evaluated the neurobehavioral alterations and the numbers of NeuN,
calbindin (CB), and parvalbumin (PV) immunopositive hippocampal neurons in
BALB/c mice after acute 5.5 Gy radiation with/without oral administration of Lyc
at the dosage of 10 g/kg daily for 4 weeks. Results: The results showed that Lyc could improve
irradiation-induced animal weight loss, depressive behaviors, spatial memory
impairment, and hippocampal neuron loss. Immunohistochemistry study demonstrated
that the loss of NeuN-immunopositive neuron in the hilus of the dentate gyrus,
CB-immunopositive neuron in CA1 strata radiatum, lacunosum moleculare and
oriens, and PV-positive neuron in CA1 stratum pyramidum and stratum granulosum
of the dentate gyrus after irradiation were significantly improved by Lyc
treatment. Conclusion: The neuroprotective effect of Lyc on those hippocampal
neurons may benefit the configuration of learning related neuronal networks and
then improve radiation induced neurobehavioral changes such as cognitive
impairment and depression. It suggests that Lycium
barbarum berry may be an alternative food supplement to prevent
radiation-induced neuron loss and neuropsychological disorders.
Collapse
Affiliation(s)
- Lei Guo
- Department of Traditional Chinese Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Qian-Qian Du
- Department of Traditional Chinese Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Piao-Qin Cheng
- Department of Traditional Chinese Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Ting-Ting Yang
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Chao-Qun Xing
- Department of Traditional Chinese Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Xue-Zhi Luo
- Department of Traditional Chinese Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Xiao-Chun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Feng Qian
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Jiang-Rong Huang
- Department of Traditional Chinese Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Feng-Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| |
Collapse
|
44
|
Lin X, Amalraj M, Blanton C, Avila B, Holmes TC, Nitz DA, Xu X. Noncanonical projections to the hippocampal CA3 regulate spatial learning and memory by modulating the feedforward hippocampal trisynaptic pathway. PLoS Biol 2021; 19:e3001127. [PMID: 34928938 PMCID: PMC8741299 DOI: 10.1371/journal.pbio.3001127] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 01/07/2022] [Accepted: 11/24/2021] [Indexed: 12/27/2022] Open
Abstract
The hippocampal formation (HF) is well documented as having a feedforward, unidirectional circuit organization termed the trisynaptic pathway. This circuit organization exists along the septotemporal axis of the HF, but the circuit connectivity across septal to temporal regions is less well described. The emergence of viral genetic mapping techniques enhances our ability to determine the detailed complexity of HF circuitry. In earlier work, we mapped a subiculum (SUB) back projection to CA1 prompted by the discovery of theta wave back propagation from the SUB to CA1 and CA3. We reason that this circuitry may represent multiple extended noncanonical pathways involving the subicular complex and hippocampal subregions CA1 and CA3. In the present study, multiple retrograde viral tracing approaches produced robust mapping results, which supports this prediction. We find significant noncanonical synaptic inputs to dorsal hippocampal CA3 from ventral CA1 (vCA1), perirhinal cortex (Prh), and the subicular complex. Thus, CA1 inputs to CA3 run opposite the trisynaptic pathway and in a temporal to septal direction. Our retrograde viral tracing results are confirmed by anterograde-directed viral mapping of projections from input mapped regions to hippocampal dorsal CA3 (dCA3). We find that genetic inactivation of the projection of vCA1 to dCA3 impairs object-related spatial learning and memory but does not modulate anxiety-related behaviors. Our data provide a circuit foundation to explore novel functional roles contributed by these noncanonical hippocampal circuit connections to hippocampal circuit dynamics and learning and memory behaviors.
Collapse
Affiliation(s)
- Xiaoxiao Lin
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, California, United States of America
| | - Michelle Amalraj
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, California, United States of America
| | - Crisylle Blanton
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, California, United States of America
| | - Brenda Avila
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, California, United States of America
| | - Todd C. Holmes
- Department Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States of America
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, California, United States of America
| | - Douglas A. Nitz
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, California, United States of America
- Department of Cognitive Science, University of California San Diego, La Jolla, California, United States of America
| | - Xiangmin Xu
- Department Anatomy & Neurobiology, School of Medicine, University of California, Irvine, California, United States of America
- The Center for Neural Circuit Mapping, University of California, Irvine, Irvine, California, United States of America
| |
Collapse
|
45
|
Ross TW, Easton A. The Hippocampal Horizon: Constructing and Segmenting Experience for Episodic Memory. Neurosci Biobehav Rev 2021; 132:181-196. [PMID: 34826509 DOI: 10.1016/j.neubiorev.2021.11.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/29/2022]
Abstract
How do we recollect specific events that have occurred during continuous ongoing experience? There is converging evidence from non-human animals that spatially modulated cellular activity of the hippocampal formation supports the construction of ongoing events. On the other hand, recent human oriented event cognition models have outlined that our experience is segmented into discrete units, and that such segmentation can operate on shorter or longer timescales. Here, we describe a unification of how these dynamic physiological mechanisms of the hippocampus relate to ongoing externally and internally driven event segmentation, facilitating the demarcation of specific moments during experience. Our cross-species interdisciplinary approach offers a novel perspective in the way we construct and remember specific events, leading to the generation of many new hypotheses for future research.
Collapse
Affiliation(s)
- T W Ross
- Department of Psychology, Durham University, South Road, Durham, DH1 3LE, United Kingdom; Centre for Learning and Memory Processes, Durham University, United Kingdom.
| | - A Easton
- Department of Psychology, Durham University, South Road, Durham, DH1 3LE, United Kingdom; Centre for Learning and Memory Processes, Durham University, United Kingdom
| |
Collapse
|
46
|
Autistic-like behavior, spontaneous seizures, and increased neuronal excitability in a Scn8a mouse model. Neuropsychopharmacology 2021; 46:2011-2020. [PMID: 33658654 PMCID: PMC8429750 DOI: 10.1038/s41386-021-00985-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 02/05/2023]
Abstract
Patients with SCN8A epileptic encephalopathy exhibit a range of clinical features, including multiple seizure types, movement disorders, and behavioral abnormalities, such as developmental delay, mild-to-severe intellectual disability, and autism. Recently, the de novo heterozygous SCN8A R1620L mutation was identified in an individual with autism, intellectual disability, and behavioral seizures without accompanying electrographic seizure activity. To date, the effects of SCN8A mutations that are primarily associated with behavioral abnormalities have not been studied in a mouse model. To better understand the phenotypic and functional consequences of the R1620L mutation, we used CRISPR/Cas9 technology to generate mice expressing the corresponding SCN8A amino acid substitution. Homozygous mutants exhibit tremors and a maximum lifespan of 22 days, while heterozygous mutants (RL/+) exhibit autistic-like behaviors, such as hyperactivity and learning and social deficits, increased seizure susceptibility, and spontaneous seizures. Current clamp analyses revealed a reduced threshold for firing action potentials in heterozygous CA3 pyramidal neurons and reduced firing frequency, suggesting that the R1620L mutation has both gain- and loss-of-function effects. In vivo calcium imaging using miniscopes in freely moving RL/+ mutants showed hyperexcitability of cortical excitatory neurons that is likely to increase seizure susceptibility. Finally, we found that oxcarbazepine and Huperzine A, a sodium channel blocker and reversible acetylcholinesterase inhibitor, respectively, were capable of conferring robust protection against induced seizures in RL/+ mutants. This mouse line will provide the opportunity to better understand the range of clinical phenotypes associated with SCN8A mutations and to develop new therapeutic approaches.
Collapse
|
47
|
Maté de Gérando A, d'Orange M, Augustin E, Joséphine C, Aurégan G, Gaudin-Guérif M, Guillermier M, Hérard AS, Stimmer L, Petit F, Gipchtein P, Jan C, Escartin C, Selingue E, Carvalho K, Blum D, Brouillet E, Hantraye P, Gaillard MC, Bonvento G, Bemelmans AP, Cambon K. Neuronal tau species transfer to astrocytes and induce their loss according to tau aggregation state. Brain 2021; 144:1167-1182. [PMID: 33842937 DOI: 10.1093/brain/awab011] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/24/2020] [Accepted: 11/01/2020] [Indexed: 11/12/2022] Open
Abstract
Deposits of different abnormal forms of tau in neurons and astrocytes represent key anatomo-pathological features of tauopathies. Although tau protein is highly enriched in neurons and poorly expressed by astrocytes, the origin of astrocytic tau is still elusive. Here, we used innovative gene transfer tools to model tauopathies in adult mouse brains and to investigate the origin of astrocytic tau. We showed in our adeno-associated virus (AAV)-based models and in Thy-Tau22 transgenic mice that astrocytic tau pathology can emerge secondarily to neuronal pathology. By designing an in vivo reporter system, we further demonstrated bidirectional exchanges of tau species between neurons and astrocytes. We then determined the consequences of tau accumulation in astrocytes on their survival in models displaying various status of tau aggregation. Using stereological counting of astrocytes, we report that, as for neurons, soluble tau species are highly toxic to some subpopulations of astrocytes in the hippocampus, whereas the accumulation of tau aggregates does not affect their survival. Thus, astrocytes are not mere bystanders of neuronal pathology. Our results strongly suggest that tau pathology in astrocytes may significantly contribute to clinical symptoms.
Collapse
Affiliation(s)
- Anastasie Maté de Gérando
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Marie d'Orange
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Emma Augustin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Charlène Joséphine
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Gwénaelle Aurégan
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Mylène Gaudin-Guérif
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Martine Guillermier
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Anne-Sophie Hérard
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Lev Stimmer
- MIRCen, INSERM-CEA, Platform for experimental pathology, U1169 and US27, F-92265 Fontenay-aux-Roses, France
| | - Fanny Petit
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Pauline Gipchtein
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Caroline Jan
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Carole Escartin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Erwan Selingue
- Université Paris-Saclay, CEA, Neurospin, 91191, Gif-sur-Yvette, France
| | - Kévin Carvalho
- Université Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- Université Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, F-59000 Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Emmanuel Brouillet
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Philippe Hantraye
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Marie-Claude Gaillard
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Alexis-Pierre Bemelmans
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Karine Cambon
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| |
Collapse
|
48
|
Brymer KJ, Kulhaway EY, Howland JG, Caruncho HJ, Kalynchuk LE. Altered acoustic startle, prepulse facilitation, and object recognition memory produced by corticosterone withdrawal in male rats. Behav Brain Res 2021; 408:113291. [PMID: 33836169 DOI: 10.1016/j.bbr.2021.113291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 03/19/2021] [Accepted: 04/04/2021] [Indexed: 12/30/2022]
Abstract
The symptoms of human depression often include cognitive deficits. However, cognition is not frequently included in the behavioral assessments conducted in preclinical models of depression. For example, it is well known that repeated corticosterone (CORT) injections in rodents produce depression-like behavior as measured by the forced swim test, sucrose preference test, and tail suspension test, but the cognitive impairments produced by repeated CORT have not been thoroughly examined. The purpose of this experiment was to assess the effect of repeated CORT injections on several versions of object recognition memory and modulation of the acoustic startle response by relatively low intensity prepulses, along with the more traditional assessment of depression-like behavior using the forced swim test. Rats received 21 days of CORT (40 mg/kg) or vehicle injections followed by a battery of behavioral tests. Importantly, during behavioral testing CORT treatment did not occur (CORT withdrawal). Corticosterone decreased body weight, increased immobility in the forced swim test, lowered startle amplitudes, and facilitated responding to trials with a short interval (30 ms) between the prepulse and pulse. Corticosterone also impaired both object location and object-in-place recognition memory, while sparing performance on object recognition memory. Collectively, our data suggest that CORT produces selective disruptions in prepulse facilitation, object location, and object-in-place recognition memory, and that these impairments should be considered as part of the phenotype produced by repeated CORT, and perhaps chronic stress.
Collapse
Affiliation(s)
- Kyle J Brymer
- Faculty of Medicine, Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada.
| | - Erin Y Kulhaway
- Research Excellence and Innovation, University of Saskatchewan, Saskatoon, SK, S7N 5A5, Canada
| | - John G Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, S7N 5A5, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, V8W 2Y2, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, V8W 2Y2, Canada
| |
Collapse
|
49
|
Grieco SF, Qiao X, Johnston KG, Chen L, Nelson RR, Lai C, Holmes TC, Xu X. Neuregulin signaling mediates the acute and sustained antidepressant effects of subanesthetic ketamine. Transl Psychiatry 2021; 11:144. [PMID: 33627623 PMCID: PMC7904825 DOI: 10.1038/s41398-021-01255-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/09/2021] [Accepted: 02/01/2021] [Indexed: 01/03/2023] Open
Abstract
Subanesthetic ketamine evokes rapid antidepressant effects in human patients that persist long past ketamine's chemical half-life of ~2 h. Ketamine's sustained antidepressant action may be due to modulation of cortical plasticity. We find that ketamine ameliorates depression-like behavior in the forced swim test in adult mice, and this depends on parvalbumin-expressing (PV) neuron-directed neuregulin-1 (NRG1)/ErbB4 signaling. Ketamine rapidly downregulates NRG1 expression in PV inhibitory neurons in mouse medial prefrontal cortex (mPFC) following a single low-dose ketamine treatment. This NRG1 downregulation in PV neurons co-tracks with the decreases in synaptic inhibition to mPFC excitatory neurons for up to a week. This results from reduced synaptic excitation to PV neurons, and is blocked by exogenous NRG1 as well as by PV targeted ErbB4 receptor knockout. Thus, we conceptualize that ketamine's effects are mediated through rapid and sustained cortical disinhibition via PV-specific NRG1 signaling. Our findings reveal a novel neural plasticity-based mechanism for ketamine's acute and long-lasting antidepressant effects.
Collapse
Affiliation(s)
- Steven F. Grieco
- grid.266093.80000 0001 0668 7243Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697-1275 USA
| | - Xin Qiao
- grid.266093.80000 0001 0668 7243Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697-1275 USA
| | - Kevin G. Johnston
- grid.266093.80000 0001 0668 7243Department of Mathematics, University of California, Irvine, CA 92697-3875 USA
| | - Lujia Chen
- grid.266093.80000 0001 0668 7243Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697-1275 USA
| | - Renetta R. Nelson
- grid.266093.80000 0001 0668 7243Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697-1275 USA
| | - Cary Lai
- grid.411377.70000 0001 0790 959XDepartment of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405-7000 USA
| | - Todd C. Holmes
- grid.19006.3e0000 0000 9632 6718Department of Physiology and Biophysics, School of Medicine, Universityof California, Irvine, CA 92697- 4560 USA ,grid.266093.80000 0001 0668 7243The Center for Neural Circuit Mapping, University of California, Irvine, CA 92697 USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697-1275, USA. .,The Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA. .,Department of Biomedical Engineering, University of California, Irvine, CA, 92697-2715, USA. .,Department of Microbiology and Molecular Genetics, University of California, Irvine, CA, 92697-4025, USA. .,Department of Computer Science, University of California, Irvine, CA, 92697-3435, USA.
| |
Collapse
|
50
|
Opalka AN, Huang WQ, Liu J, Liang H, Wang DV. Hippocampal Ripple Coordinates Retrosplenial Inhibitory Neurons during Slow-Wave Sleep. Cell Rep 2021; 30:432-441.e3. [PMID: 31940487 PMCID: PMC7007963 DOI: 10.1016/j.celrep.2019.12.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/01/2019] [Accepted: 12/12/2019] [Indexed: 12/20/2022] Open
Abstract
The hippocampus and retrosplenial cortex (RSC) play indispensable roles in memory formation, and importantly, a hippocampal oscillation known as ripple is key to consolidation of new memories. However, it remains unclear how the hippocampus and RSC communicate and the role of ripple oscillation in coordinating the activity between these two brain regions. Here, we record from the dorsal hippocampus and RSC simultaneously in freely behaving mice during sleep and reveal that the RSC displays a pre-ripple activation associated with slow and fast oscillations. Immediately after ripples, a subpopulation of RSC putative inhibitory neurons increases firing activity, while most RSC putative excitatory neurons decrease activity. Consistently, optogenetic stimulation of this hippocampus-RSC pathway activates and suppresses RSC putative inhibitory and excitatory neurons, respectively. These results suggest that the dorsal hippocampus mainly inhibits RSC activity via its direct innervation of RSC inhibitory neurons, which overshadows the RSC in supporting learning and memory functions. Converging evidence suggests that hippocampal ripple oscillations and their interaction with the neocortex are critical for memory consolidation. By combining electrophysiology and optogenetic techniques in freely behaving mice, Opalka et al. provide direct evidence that hippocampal ripples communicate with retrosplenial cortex (RSC) interneurons and inhibit RSC population activity during sleep-associated memory consolidation.
Collapse
Affiliation(s)
- Ashley N Opalka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Wen-Qiang Huang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Jun Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Hualou Liang
- School of Biomedical Engineering, Drexel University, Philadelphia, PA 19104, USA
| | - Dong V Wang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| |
Collapse
|