1
|
Radahmadi M, Halabian A, Halabian A. An overview of extracellular field potentials: Different potentiation and measurable components, interpretations, and hippocampal synaptic activity models. Methods 2025; 239:50-63. [PMID: 40147603 DOI: 10.1016/j.ymeth.2025.03.015] [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: 12/29/2024] [Revised: 03/03/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
The hippocampus and some other brain regions are critically involved in synaptic plasticity. Electrophysiological recordings using extracellular field potentials (EFPs) reveal diverse synaptic activity within the hippocampus, including input/output functions (reflecting neural excitability), paired-pulse responses (reflecting short-term plasticity), and long-term potentiation (reflecting long-term plasticity). EFP techniques offer various measurable components for assessing multiple neural functions. These include fEPSP slope, amplitude, and area under curve (AUC), as well as latency (fEPSP onset or peak after stimulation), width at half amplitude, fiber volley, decay time, time-course (fEPSP rise and decay time constants; tau), initial slope/initial area and -/late area derived from a fEPSP waveform sample. Each of these parameters is separately evaluated and provides distinct electrophysiological interpretations. Despite the rich data offered by EFP techniques, many studies adopt a limited approach, focusing solely on fEPSP slope, amplitude, and occasionally AUC, thereby neglecting the potential insights provided by other parameters. Given the inherent variability of fEPSP components within a single recording and timeframe, a comprehensive analysis of synaptic activity within a specific hippocampal region is necessary for obtaining the full spectrum of fEPSP-related data. Researchers should consider the potential influence of additional factors contributing to the variability of synaptic activity magnitude. A detailed analysis considering different parts of extracellular fEPSP recordings and their properties is crucial for a deeper understanding of synaptic activity changes within the brain. Therefore, this review aims to provide a comprehensive overview of diverse forms of hippocampal synaptic activity, measurable components of EFP recordings, and their corresponding interpretations.
Collapse
Affiliation(s)
- Maryam Radahmadi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Alireza Halabian
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Arshia Halabian
- Department of Electrical Engineering, University of Isfahan, Isfahan, Iran
| |
Collapse
|
2
|
Lopes-Dos-Santos V, Brizee D, Dupret D. Spatio-temporal organization of network activity patterns in the hippocampus. Cell Rep 2025; 44:115808. [PMID: 40478735 PMCID: PMC7617751 DOI: 10.1016/j.celrep.2025.115808] [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: 02/10/2025] [Revised: 04/04/2025] [Accepted: 05/19/2025] [Indexed: 06/11/2025] Open
Abstract
Understanding how coordinated neural networks support brain functions remains a central goal in neuroscience. The hippocampus, with its layered architecture and structured inputs to diverse cell populations, is a tractable model for dissecting operating microcircuits through the analysis of electrophysiological signatures. We investigated hippocampal network patterns in behaving mice by developing a low-dimensional embedding of local field potentials recorded along the CA1-to-dentate gyrus axis. This embedding revealed layer-specific gamma profiles reflecting spatially organized rhythms and their associated principal cell-interneuron firing motifs. Moreover, firing behaviors along the CA1 radial axis distinguished between deep and superficial principal cells, as well as between interneurons from the pyramidal, radiatum, and lacunosum-moleculare layers. These findings provide a comprehensive map of spatiotemporal activity patterns underlying hippocampal network functions.
Collapse
Affiliation(s)
- Vítor Lopes-Dos-Santos
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK.
| | - Demi Brizee
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - David Dupret
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK.
| |
Collapse
|
3
|
Wei D, Tan S, Pang S, Liu B, Zhang Q, Zhu S, Fu G, Sun D, Wei W. Protective effects of anthocyanins on the nervous system injury caused by fluoride-induced endoplasmic reticulum stress in rats. Food Chem Toxicol 2025; 200:115386. [PMID: 40073964 DOI: 10.1016/j.fct.2025.115386] [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: 12/25/2024] [Revised: 02/14/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
Long-term fluoride exposure can produce neurotoxicity. Anthocyanins, as antioxidants, have a certain protective effect in nerve damage. This study aimed to investigate the protective role of anthocyanins in fluoride-induced neurological damage due to endoplasmic reticulum stress (ERS). Using a fluoride-exposed Wistar rat model, we assessed learning memory capacity and pathologic and ultrastructural injury. The level of oxidative stress (OS) in vivo was detected by colorimetric method, the level of ERS was analyzed by immunohistochemistry, and the apoptosis of neuronal cells was observed by TUNEL staining. The results showed that fluoride exposure could decrease the learning and memory ability in rats, and led to histopathological and ultrastructural damage in the hippocampal CA1, CA3 and cortical regions. Fluoride exposure-induced OS in vivo, which further activates ERS, which was manifested by increased levels of ERS-related proteins GRP78, Caspase 12, and Caspase 3 in hippocampal CA1, CA3, and cortical regions, and eventually led to a significant increase in neuronal apoptosis rate. Notably, after anthocyanins treatment, pathological and ultrastructural damage was restored, the level of OS and ERS were significantly restored, and the apoptosis rate of neuronal cells was significantly reduced. In summary, as nutritional interventions, anthocyanins exert a protective role in fluoride-induced neurological injury.
Collapse
Affiliation(s)
- Dan Wei
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Shiwen Tan
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo, 315010, China
| | - Shujuan Pang
- Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, Shandong Province, 266033, China
| | - Bingshu Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Qiong Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Siqi Zhu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Heilongjiang Provincial Key Lab of Trace Elements and Human Health Harbin Medical University, Harbin, 150081, China
| | - Guiyu Fu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Jining Center For Disease Control and Prevention, Jining, Shandong Province, 272000, China
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Heilongjiang Provincial Key Lab of Trace Elements and Human Health Harbin Medical University, Harbin, 150081, China.
| | - Wei Wei
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China; Heilongjiang Provincial Key Lab of Trace Elements and Human Health Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
4
|
Chen Z, Liu Y, Yang Y, Wang L, Qin M, Jiang Z, Xu M, Zhang S. Whole-brain mapping of basal forebrain cholinergic neurons reveals a long-range reciprocal input-output loop between distinct subtypes. SCIENCE ADVANCES 2025; 11:eadt1617. [PMID: 40446047 PMCID: PMC12124396 DOI: 10.1126/sciadv.adt1617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 04/24/2025] [Indexed: 06/02/2025]
Abstract
Basal forebrain cholinergic neurons (BFCNs) influence cognition and emotion through specific acetylcholine release in various brain regions, including the prefrontal cortices and basolateral amygdala (BLA). Acetylcholine release is controlled by distinct BFCN subtypes, modulated by excitatory and inhibitory inputs. However, the organization of the whole-brain input-output networks of these subtypes remains unclear. Here, we identified two distinct BFCN subtypes-BFCN→ACA and BFCN→BLA-innervating the anterior cingulate cortex (ACA) and BLA, each with unique distributions, electrophysiological properties, and projection patterns. Combining rabies-virus-assisted mapping and triple-plex RNAscope hybridization, we characterized their whole-brain input networks, identifying unique excitatory and shared inhibitory inputs for these subtypes. Moreover, our results reveal a long-range reciprocal input-output loop: BFCN→ACA neurons target the isocortex, their shared excitatory-input source, whereas BFCN→BLA neurons target shared inhibitory-input sources such as the striatum and pallidum, thus enabling dynamic interactions among these BFCN subtypes. Our study deepens understanding of cholinergic modulation in cognition and emotion and provides insights into their functional interactions.
Collapse
Affiliation(s)
- Zhaonan Chen
- Department of Ophthalmology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yanmei Liu
- Department of Ophthalmology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yunqi Yang
- Department of Ophthalmology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lizhao Wang
- Department of Ophthalmology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Meiling Qin
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhishan Jiang
- Department of Ophthalmology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Min Xu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Siyu Zhang
- Department of Ophthalmology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
5
|
Ikebara JM, Jorge RS, Marinho LSR, Higa GSV, Adhikari A, Reis FMCV, Borges FS, Ulrich H, Takada SH, De Pasquale R, Kihara AH. Hippocampal Interneurons Shape Spatial Coding Alterations in Neurological Disorders. Mol Neurobiol 2025:10.1007/s12035-025-05020-2. [PMID: 40392508 DOI: 10.1007/s12035-025-05020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Hippocampal interneurons (INs) play a fundamental role in regulating neural oscillations, modulating excitatory circuits, and shaping spatial representation. While historically overshadowed by excitatory pyramidal cells in spatial coding research, recent advances have demonstrated that inhibitory INs not only coordinate network dynamics but also contribute directly to spatial information processing. This review aims to provide a novel integrative perspective on how distinct IN subtypes participate in spatial coding and how their dysfunction contributes to cognitive deficits in neurological disorders such as epilepsy, Alzheimer's disease (AD), traumatic brain injury (TBI), and cerebral hypoxia-ischemia. We synthesize recent findings demonstrating that different IN classes-including parvalbumin (PV)-, somatostatin (SST)-, cholecystokinin (CCK)-, and calretinin (CR)-expressing neurons-exhibit spatially selective activity, challenging traditional views of spatial representation, and influence memory consolidation through network-level interactions. By leveraging cutting-edge techniques such as in vivo calcium imaging and optogenetics, new evidence suggests that INs encode spatial information with a level of specificity previously attributed only to pyramidal cells. Furthermore, we investigate the impact of inhibitory circuit dysfunction in neurological disorders, examining how disruptions in interneuronal activity lead to impaired theta-gamma coupling, altered sharp wave ripples, and destabilized place cell representations, ultimately resulting in spatial memory deficits. This review advances the field by shifting the focus from pyramidal-centered models to a more nuanced understanding of the hippocampal network, emphasizing the active role of INs in spatial coding. By highlighting the translational potential of targeting inhibitory circuits for therapeutic interventions, we propose novel strategies for restoring hippocampal network function in neurological conditions. Readers will gain a comprehensive understanding of the emerging role of INs in spatial representation and the critical implications of their dysfunction, paving the way for future research on interneuron-targeted treatments for cognitive disorders.
Collapse
Affiliation(s)
- Juliane Midori Ikebara
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Renata Silva Jorge
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Luciana Simões Rafagnin Marinho
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Guilherme Shigueto Vilar Higa
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Avishek Adhikari
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Fernando M C V Reis
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Fernando S Borges
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Roberto De Pasquale
- Neurophysiology Laboratory, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Alexandre Hiroaki Kihara
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil.
| |
Collapse
|
6
|
Xia T, Yan Z, Shen P, Chang M, Zhang N, Zhang Y, Chen Q, Wang R, Tong L, Zhou W, Ni Z, Gao Y. Neuroprotective Effects of Qi Jing Wan and Its Active Ingredient Diosgenin Against Cognitive Impairment in Plateau Hypoxia. Pharmaceuticals (Basel) 2025; 18:738. [PMID: 40430556 PMCID: PMC12114856 DOI: 10.3390/ph18050738] [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: 04/10/2025] [Revised: 05/03/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: High-altitude environments have a significant detrimental impact on the cognitive functions of the brain. Qi Jing Wan (QJW), a traditional herbal formula composed of Angelica sinensis, Astragalus membranaceus, and Rhizoma Polygonati Odorati, has demonstrated potential efficacy in treating cognitive disorders. However, its effects on cognitive dysfunction in plateau hypoxic environments remain unclear. Methods: In this study, acute and chronic plateau cognitive impairment mouse models were constructed to investigate the preventive and therapeutic effects of QJW and its significant active ingredient, diosgenin (Dio). Behavioral experiments were conducted to assess learning and memory in mice. Morphological changes in hippocampal neurons and synapses were assessed, and microglial activation and inflammatory factor levels were measured to evaluate brain damage. Potential active ingredients capable of crossing the blood-brain barrier were identified through chemical composition analysis and network database screening, followed by validation in animal and brain organoid experiments. Transcriptomics analysis, immunofluorescence staining, and molecular docking techniques were employed to explore the underlying mechanisms. Results: QJW significantly enhanced learning and memory abilities in plateau model mice, reduced structural damage to hippocampal neurons, restored NeuN expression, inhibited inflammatory factor levels and microglial activation, and improved hippocampal synaptic damage. Transcriptomics analysis revealed that Dio alleviated hypoxic brain damage and protected cognitive function by regulating the expression of PDE4C. Conclusions: These findings indicate that QJW and its significant active ingredient Dio effectively mitigate hypoxic brain injury and prevent cognitive impairment in high-altitude environments.
Collapse
Affiliation(s)
- Tiantian Xia
- Department of Tranditional Chinese Medicine, Qinghai Unversity Medical College, Xining 810016, China; (T.X.); (N.Z.); (L.T.)
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Y.); (P.S.); (M.C.); (Y.Z.); (Q.C.); (W.Z.)
| | - Ziqiao Yan
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Y.); (P.S.); (M.C.); (Y.Z.); (Q.C.); (W.Z.)
- Chinese PLA Medical School, Chinese People’s Liberation Army (PLA) General Hospital, Beijing 100036, China
| | - Pan Shen
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Y.); (P.S.); (M.C.); (Y.Z.); (Q.C.); (W.Z.)
| | - Mingyang Chang
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Y.); (P.S.); (M.C.); (Y.Z.); (Q.C.); (W.Z.)
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Nan Zhang
- Department of Tranditional Chinese Medicine, Qinghai Unversity Medical College, Xining 810016, China; (T.X.); (N.Z.); (L.T.)
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Y.); (P.S.); (M.C.); (Y.Z.); (Q.C.); (W.Z.)
| | - Yunan Zhang
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Y.); (P.S.); (M.C.); (Y.Z.); (Q.C.); (W.Z.)
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qi Chen
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Y.); (P.S.); (M.C.); (Y.Z.); (Q.C.); (W.Z.)
- Chinese PLA Medical School, Chinese People’s Liberation Army (PLA) General Hospital, Beijing 100036, China
| | - Rui Wang
- General Hospital of Xinjiang Military Command, PLA, Urumqi 830000, China;
| | - Li Tong
- Department of Tranditional Chinese Medicine, Qinghai Unversity Medical College, Xining 810016, China; (T.X.); (N.Z.); (L.T.)
| | - Wei Zhou
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Y.); (P.S.); (M.C.); (Y.Z.); (Q.C.); (W.Z.)
| | - Zhexin Ni
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Y.); (P.S.); (M.C.); (Y.Z.); (Q.C.); (W.Z.)
| | - Yue Gao
- Department of Tranditional Chinese Medicine, Qinghai Unversity Medical College, Xining 810016, China; (T.X.); (N.Z.); (L.T.)
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (Z.Y.); (P.S.); (M.C.); (Y.Z.); (Q.C.); (W.Z.)
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
7
|
Anderson MC, Crespo-Garcia M, Subbulakshmi S. Brain mechanisms underlying the inhibitory control of thought. Nat Rev Neurosci 2025:10.1038/s41583-025-00929-y. [PMID: 40379896 DOI: 10.1038/s41583-025-00929-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2025] [Indexed: 05/19/2025]
Abstract
Controlling action and thought requires the capacity to stop mental processes. Over the past two decades, evidence has grown that a domain-general inhibitory control mechanism supported by the right lateral prefrontal cortex achieves these functions. However, current views of the neural mechanisms of inhibitory control derive largely from research into the stopping of action. Whereas action stopping is a convenient empirical model, it does not invoke thought inhibition and cannot be used to identify the unique features of this process. Here, we review research that addresses how organisms stop a key process that drives thoughts: memory retrieval. This work has shown that retrieval stopping shares right dorsolateral and ventrolateral prefrontal mechanisms with action stopping, consistent with a domain-general inhibitory control mechanism, but also recruits a distinct fronto-temporal pathway that determines the success of mental control. As part of this pathway, GABAergic inhibition within the hippocampus influences the efficacy of prefrontal control over thought. These unique elements of mental control suggest that hippocampal disinhibition is a transdiagnostic factor underlying intrusive thinking, linking the fronto-temporal control pathway to preclinical models of psychiatric disorders and fear extinction. We suggest that retrieval-stopping deficits may underlie the intrusive thinking that is common across many psychiatric disorders.
Collapse
Affiliation(s)
- Michael C Anderson
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK.
- Behavioural and Clinical Neurosciences Unit, University of Cambridge, Cambridge, UK.
| | - Maite Crespo-Garcia
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - S Subbulakshmi
- Department of Psychology, Stanford University, Stanford, CA, USA
| |
Collapse
|
8
|
Chiang C, Chien M, Huang Y, Lin J, Liang S, Hsu K, Durand DM, Wu Y. Cathodal weak direct current decreases epileptic excitability with reduced neuronal activity and enhanced delta oscillations. J Physiol 2025; 603:2763-2782. [PMID: 40193544 PMCID: PMC12072238 DOI: 10.1113/jp287969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/12/2025] [Indexed: 04/09/2025] Open
Abstract
Seizures are manifestations of hyperexcitability in the brain. Non-invasive weak current stimulation, delivered through cathodal transcranial direct current stimulation (ctDCS), has emerged to treat refractory epilepsy and seizures, although the cellular-to-populational electrophysiological mechanisms remain unclear. Using the ctDCS in vivo model, we investigate how neural excitability is modulated through weak direct currents by analysing the local field potential (LFP) and extracellular unit spike recordings before, during and after ctDCS versus sham stimulation. In rats with kainic acid (KA)-induced acute hippocampal seizures, ctDCS reduced seizure excitability by decreasing the number and amplitude of epileptic spikes in LFP and enhancing delta (δ) power. We identified unit spikes of putative excitatory neurons in CA1 stratum pyramidale based on waveform sorting and validated via optogenetic inhibitions which increased aberrantly in seizure animals. Notably, cathodal stimulation significantly reduced these unit spikes, whereas anodal stimulation exhibited the opposite effect, showing polarity-specific and current strength-dependent responses. The reduced unit spikes after ctDCS coupled to δ oscillations with an increased coupling strength. These effects occurred during stimulation and lasted 90 min post-stimulation, accompanied by inhibitory short-term synaptic plasticity changes shown in paired-pulse stimulation after ctDCS. Consistently, neuronal activations measured by c-Fos significantly decreased after ctDCS, particularly in CaMKII+-excitatory neurons while increased in GAD+-inhibitory neurons. In conclusion, epileptic excitability was alleviated with cathodal weak direct current stimulation by diminishing excitatory neuronal activity and enhancing endogenous δ oscillations through strengthened coupling between unit spikes and δ waves, along with inhibitory plasticity changes, highlighting the potential implications to treat brain disorders characterized by hyperexcitability. KEY POINTS: Electric fields generated by transcranial weak electric current stimulation were measured at CA1, showing polarity-specific and current strength-dependent modulation of unit spike activity. Polyspike epileptiform discharges were observed in rats with kainic acid (KA)-induced hippocampal seizures. Cathodal transcranial direct current stimulation (ctDCS) reduced the number and amplitude of the epileptic spikes in local field potentials (LFPs) while increased δ oscillations. Neuronal unit spikes aberrantly increased in seizures and coupled with epileptiform discharges. ctDCS reduced excitatory neuronal firings at CA1 and strengthened the coupling between unit spikes and δ waves. Neuronal activations, measured by c-Fos, decreased in CaMKII+-excitatory neurons while increased in GAD+-inhibitory neurons after ctDCS. These effects on LFP and unit spikes lasted up to 90 min post-stimulation. Inhibitory short-term plasticity changes detected through paired-pulse stimulation underpin the enduring effects of ctDCS on seizures.
Collapse
Affiliation(s)
- Chia‐Chu Chiang
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
- Department of Biomedical Engineering, Neural Engineering CenterCase Western Reserve UniversityClevelandOhioUSA
| | - Miao‐Er Chien
- Institute of Clinical Medicine, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Yu‐Chieh Huang
- Institute of Clinical Medicine, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Jyun‐Ting Lin
- Department of Computer Science and Information EngineeringNational Cheng Kung UniversityTainanTaiwan
| | - Sheng‐Fu Liang
- Department of Computer Science and Information EngineeringNational Cheng Kung UniversityTainanTaiwan
| | - Kuei‐Sen Hsu
- Department of Pharmacology, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Institute of Basic Medical Sciences, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Dominique M. Durand
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
- Department of Biomedical Engineering, Neural Engineering CenterCase Western Reserve UniversityClevelandOhioUSA
| | - Yi‐Jen Wu
- Institute of Clinical Medicine, College of MedicineNational Cheng Kung UniversityTainanTaiwan
- Department of Neurology, National Cheng Kung University Hospital, College of MedicineNational Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
9
|
Speers LJ, Bilkey DK. Inflammation in Schizophrenia: The Role of Disordered Oscillatory Mechanisms. Cells 2025; 14:650. [PMID: 40358174 PMCID: PMC12071425 DOI: 10.3390/cells14090650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Schizophrenia is a chronic, debilitating disorder with diverse symptomatology, including disorganised cognition and behaviour. Despite considerable research effort, we have only a limited understanding of the underlying brain dysfunction. A significant proportion of individuals with schizophrenia exhibit high levels of inflammation, and inflammation associated with maternal immune system activation is a risk factor for the disorder. In this review, we outline the potential role of inflammation in the disorder, with a particular focus on how cytokine release might affect the development and function of GABAergic interneurons. One consequence of this change in inhibitory control is a disruption in oscillatory processes in the brain. These changes disrupt the spatial and temporal synchrony of neural activity in the brain, which, by disturbing representations of time and space, may underlie some of the disorganisation symptoms observed in the disorder.
Collapse
Affiliation(s)
| | - David K. Bilkey
- Psychology Department, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
10
|
Eyolfson E, Suesser KRB, Henry H, Bonilla-Del Río I, Grandes P, Mychasiuk R, Christie BR. The effect of traumatic brain injury on learning and memory: A synaptic focus. Neuroscientist 2025; 31:195-214. [PMID: 39316552 PMCID: PMC11909778 DOI: 10.1177/10738584241275583] [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] [Indexed: 09/26/2024]
Abstract
Deficits in learning and memory are some of the most commonly reported symptoms following a traumatic brain injury (TBI). We will examine whether the neural basis of these deficits stems from alterations to bidirectional synaptic plasticity within the hippocampus. Although the CA1 subregion of the hippocampus has been a focus of TBI research, the dentate gyrus should also be given attention as it exhibits a unique ability for adult neurogenesis, a process highly susceptible to TBI-induced damage. This review examines our current understanding of how TBI results in deficits in synaptic plasticity, as well as how TBI-induced changes in endocannabinoid (eCB) systems may drive these changes. Through the synthesis and amalgamation of existing data, we propose a possible mechanism for eCB-mediated recovery in synaptic plasticity deficits. This hypothesis is based on the plausible roles of CB1 receptors in regulating inhibitory tone, influencing astrocytes and microglia, and modulating glutamate release. Dysregulation of the eCBs may be responsible for deficits in synaptic plasticity and learning following TBI. Taken together, the existing evidence indicates eCBs may contribute to TBI manifestation, pathogenesis, and recovery, but it also suggests there may be a therapeutic role for the eCB system in TBI.
Collapse
Affiliation(s)
- Eric Eyolfson
- Division of Medical Sciences and Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Kirsten R. B. Suesser
- Division of Medical Sciences and Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Holly Henry
- Division of Medical Sciences and Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Itziar Bonilla-Del Río
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country, Leioa, Spain
| | - Pedro Grandes
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country, Leioa, Spain
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Brian R. Christie
- Division of Medical Sciences and Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, San Diego State University, San Diego, CA, USA
| |
Collapse
|
11
|
Hall AF, Wang DV. A cortical-hippocampal communication undergoes rebalancing after new learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645547. [PMID: 40196557 PMCID: PMC11974847 DOI: 10.1101/2025.03.26.645547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The brain's ability to consolidate a wide range of memories while maintaining their distinctiveness across experiences remains poorly understood. Sharp-wave ripples, neural oscillations that occur predominantly within CA1 of the hippocampus during immobility and sleep, have been shown to play a critical role in the consolidation process. More recently, evidence has uncovered functional heterogeneity of pyramidal neurons within distinct sublayers of CA1 that display unique properties during ripples, potentially contributing to memory specificity. Despite this, it remains unclear exactly how ripples shift the activity of CA1 neuronal populations to accommodate the consolidation of specific memories and how sublayer differences manifest. Here, we studied interactions between the anterior cingulate cortex (ACC) and CA1 neurons during ripples and discovered a reorganization of their communication following learning. Notably, this reorganization appeared specifically for CA1 superficial (CA1sup) sublayer neurons. Utilizing a generalized linear model decoder, we demonstrate the pre-existence of ACC-to-CA1sup communication, which is suppressed during new learning and subsequent sleep suggesting that ACC activity may reallocate the contribution of CA1sup neurons during memory acquisition and consolidation. Further supporting this notion, we found that optogenetic stimulations of the ACC preferentially suppressed CA1sup interneurons while activating a unique subset of CA1 interneurons. Overall, these findings highlight a possible role of the ACC in rebalancing CA1 neuronal populations' contribution to ripple contents surrounding learning.
Collapse
Affiliation(s)
- Arron F Hall
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Dong V Wang
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| |
Collapse
|
12
|
Li Y, Long S, Yu J, Feng J, Meng S, Li Y, Zhao L, Yu Y. Preoperative Sleep Deprivation Exacerbates Anesthesia/Surgery-induced Abnormal GABAergic Neurotransmission and Neuronal Damage in the Hippocampus in Aged Mice. Mol Neurobiol 2025:10.1007/s12035-025-04851-3. [PMID: 40106167 DOI: 10.1007/s12035-025-04851-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Older adults with anesthesia and surgery often suffer from postoperative cognitive dysfunction (POCD), which puts a heavy burden on rehabilitation. Preoperative sleep disorder, a common phenomenon in elderly anesthesia patients, is closely associated with POCD, but the underlying mechanism is still not fully understood. Hippocampal gamma-aminobutyric acid (GABA)ergic neurotransmission has been reported to play an important role in sleep disorder and cognitive impairment. The aim of this study was to elucidate the effect of preoperative acute sleep deprivation (SD) on anesthesia/surgery-induced POCD and the potential mechanism of hippocampal GABAergic neurotransmission. In the aged (18-20-month-old) male mice, we used a rotating rod to deprive sleep for 24 h and induced a POCD model using sevoflurane exposure combined with laparotomy exploration. A sequential set of behavioral tests, including open field test (OFT), Y-maze, and novel object recognition (NOR), was conducted to assess cognitive performances. In vivo magnetic resonance imaging (MRI) technique was used to observe hippocampal axonal microstructural changes. The levels of GABAergic neurotransmitter markers glutamic acid decarboxylase (GAD) 67, vesicular GABA transporter (VGAT), GABA transporter (GAT)-1, and GABA in the hippocampus were detected with enzyme-linked immunosorbent assay (ELISA). The reactivity of GABAergic neurons and neuronal damage in different subregions of the hippocampus were observed by immunofluorescence and Nissl staining, respectively. Compared the anesthesia/surgery (A/S) mice, 24-h SD combined with A/S induced shorter stay time in the central area of the open field, less the percent of novel arm preference in the Y maze, and lower recognition index in the NOR, as well as significantly enhanced hippocampal GABAergic neurotransmission, decreased hippocampal axonal integrity and density, and increased GAD67 reactivity and reduced the number of neurons in hippocampal CA1. Preoperative 24-h SD exacerbated anesthesia/surgery-induced POCD in aged mice, with the cumulative effect of abnormal GABAergic neurotransmission and neuronal damage in the hippocampus.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Siwen Long
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Jiafeng Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Jingyu Feng
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Shuqi Meng
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Lina Zhao
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China.
| |
Collapse
|
13
|
Berden L, Rajan N, Mbouombouo Mfossa AC, De Bie I, Etlioglu E, Benotmane MA, Verslegers M, Aourz N, Smolders I, Rigo JM, Brône B, Quintens R. Interneuron migration impairment and brain region-specific DNA damage response following irradiation during early neurogenesis in mice. Cell Mol Life Sci 2025; 82:118. [PMID: 40095026 PMCID: PMC11914712 DOI: 10.1007/s00018-025-05643-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025]
Abstract
Embryonic DNA damage resulting from DNA repair deficiencies or exposure to ionizing radiation during early neurogenesis can lead to neurodevelopmental disorders, including microcephaly. This has been linked to an excessive DNA damage response in dorsal neural progenitor cells (NPCs), resulting in p53-dependent apoptosis and premature neuronal differentiation which culminates in depletion of the NPC pool. However, the effect of DNA damage on ventral forebrain NPCs, the origin of interneurons, remains unclear. In this study, we investigated the sequelae of irradiation of mouse fetuses at an early timepoint of forebrain neurogenesis. We focused on the neocortex (NCX) and medial ganglionic eminence (MGE), key regions for developing dorsal and ventral NPCs, respectively. Although both regions showed a typical p53-mediated DNA damage response consisting of cell cycle arrest, DNA repair and apoptosis, NCX cells displayed prolonged cell cycle arrest, while MGE cells exhibited more sustained apoptosis. Moreover, irradiation reduced the migration speed of interneurons in acute living brain slices and MGE explants, the latter indicating a cell-intrinsic component in the defect. RNA sequencing and protein analyses revealed disruptions in actin and microtubule cytoskeletal-related cellular machinery, particularly in MGE cells. Despite massive acute apoptosis and an obvious interneuron migration defect, prenatally irradiated animals did not show increased sensitivity to pentylenetetrazole-induced seizures, nor was there a reduction in cortical interneurons in young adult mice. This suggests a high plasticity of the developing brain to acute insults during early neurogenesis. Overall, our findings indicate that embryonic DNA damage induces region-specific responses, potentially linked to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lisa Berden
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Laboratory for Neurophysiology, BIOMED Research Institute, UHasselt, Hasselt, Belgium
| | - Nicholas Rajan
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | | | - Isabeau De Bie
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- 4BRAIN, Department of Head and Skin, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Emre Etlioglu
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Mohammed Abderrafi Benotmane
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Mieke Verslegers
- Preclinical Sciences and Translational Safety, Johnson & Johnson IM, Beerse, Belgium
| | - Najat Aourz
- Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Faculteit Geneeskunde en Farmacie, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Ilse Smolders
- Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Faculteit Geneeskunde en Farmacie, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Jean-Michel Rigo
- Laboratory for Neurophysiology, BIOMED Research Institute, UHasselt, Hasselt, Belgium
| | - Bert Brône
- Laboratory for Neurophysiology, BIOMED Research Institute, UHasselt, Hasselt, Belgium
| | - Roel Quintens
- Radiobiology Unit, Nuclear Medical Applications Institute, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium.
| |
Collapse
|
14
|
Wang C, Liu J, Su L, Wang X, Bian Y, Wang Z, Ye L, Lu X, Zhou L, Chen W, Yang W, Liu J, Wang L, Shen Y. GABAergic Progenitor Cell Graft Rescues Cognitive Deficits in Fragile X Syndrome Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411972. [PMID: 39823534 PMCID: PMC11904963 DOI: 10.1002/advs.202411972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/17/2024] [Indexed: 01/19/2025]
Abstract
Fragile X syndrome (FXS) is an inherited neurodevelopmental disorder characterized by a range of clinical manifestations with no effective treatment strategy to date. Here, transplantation of GABAergic precursor cells from the medial ganglionic eminence (MGE) is demonstrated to significantly improve cognitive performance in Fmr1 knockout (KO) mice. Within the hippocampus of Fmr1-KO mice, MGE-derived cells from wild-type donor mice survive, migrate, differentiate into functionally mature interneurons, and form inhibitory synaptic connections with host pyramidal neurons. MGE cell transplantation restores Ras-PKB signaling in pyramidal neurons, enhances AMPA receptor trafficking, rescues synaptic plasticity, and corrects abnormal hippocampal neural oscillations. These findings highlight the potential of GABAergic precursor cell transplantation as a promising therapeutic strategy for FXS.
Collapse
Affiliation(s)
- Chen Wang
- Department of NeurologyInstitute of NeuroscienceKey Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaThe Second Affiliated HospitalGuangzhou Medical UniversityGuangzhou510260China
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
| | - Jia‐Yu Liu
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Zhejiang Development & Planning InstituteHangzhou310030China
| | - Li‐Da Su
- Neuroscience Care UnitKey Laboratory of Multiple Organ Failure of Ministry of Educationthe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhou310009China
- Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang ProvinceHangzhou310009China
| | - Xin‐Tai Wang
- Institute of Life SciencesCollege of Life and Environmental SciencesHangzhou Normal UniversityHangzhou311121China
| | - Yu‐Peng Bian
- Center for Brain Healththe Fourth Affiliated Hospital of School of Medicineand International School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
| | | | - Lu‐Yu Ye
- Department of BiophysicsZhejiang University School of MedicineHangzhou310058China
| | - Xin‐Jiang Lu
- Department of PhysiologyZhejiang University School of MedicineHangzhou310058China
| | - Lin Zhou
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
| | - Wei Chen
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
| | - Wei Yang
- Department of BiophysicsZhejiang University School of MedicineHangzhou310058China
| | - Jun Liu
- Department of NeurologyInstitute of NeuroscienceKey Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaThe Second Affiliated HospitalGuangzhou Medical UniversityGuangzhou510260China
| | - Luxi Wang
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Center for Brain Healththe Fourth Affiliated Hospital of School of Medicineand International School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
| | - Ying Shen
- Department of NeurologyInstitute of NeuroscienceKey Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of ChinaThe Second Affiliated HospitalGuangzhou Medical UniversityGuangzhou510260China
- Department of Physiology and Department of PsychiatrySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Center for Brain Healththe Fourth Affiliated Hospital of School of Medicineand International School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Key Laboratory for Precision DiagnosisTreatmentand Clinical Translation of Rare Diseases of Zhejiang ProvinceZhejiang University School of MedicineHangzhou310058China
| |
Collapse
|
15
|
Strauch C, Böge J, Shchyglo O, Dubovyk V, Manahan‐Vaughan D. The Suprapyramidal and Infrapyramidal Blades of the Dentate Gyrus Exhibit Different GluN Subunit Content and Dissimilar Frequency-Dependent Synaptic Plasticity In Vivo. Hippocampus 2025; 35:e70002. [PMID: 39994965 PMCID: PMC11850964 DOI: 10.1002/hipo.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025]
Abstract
The entorhinal cortex sends afferent information to the hippocampus by means of the perforant path (PP). The PP input to the dentate gyrus (DG) terminates in the suprapyramidal (sDG) and infrapyramidal (iDG) blades. Different electrophysiological stimulation patterns of the PP can generate hippocampal synaptic plasticity. Whether frequency-dependent synaptic plasticity differs in the sDG and iDG is unclear. Here, we compared medial PP-DG responses in freely behaving adult rats and found that synaptic plasticity in the sDG is broadly frequency dependent, whereby long-term depression (LTD, > 24 h) is induced with stimulation at 1 Hz, short-term depression (< 2 h) is triggered by 5 or 10 Hz, and long-term potentiation (LTP) of increasing magnitudes is induced by 200 and 400 Hz stimulation, respectively. By contrast, although the iDG expresses STD following 5 or 10 Hz stimulation, LTD induced by 1 Hz is weaker, LTP is not induced by 200 Hz and LTP induced by 400 Hz stimulation is significantly smaller in magnitude than LTP induced in sDG. Furthermore, the stimulus-response relationship of iDG is suppressed compared to sDG. These differences may arise from differences in granule cell properties, or the complement of NMDA receptors. Patch clamp recordings, in vitro, revealed reduced firing frequencies in response to high currents, and different action potential thresholds in iDG compared to sDG. Assessment of the expression of GluN subunits revealed significantly lower expression levels of GluN1, GluN2A, and GluN2B in the middle molecular layer of iDG compared to sDG. Taken together, these data indicate that synaptic plasticity in the iDG is weaker, less persistent and less responsive to afferent frequencies than synaptic plasticity in sDG. Effects may be mediated by weaker NMDA receptor expression and differences in neuronal responses in iDG versus sDG. These characteristics may explain reported differences in experience-dependent information processing in the suprapyramidal and infrapyramidal blades of the DG.
Collapse
Affiliation(s)
- Christina Strauch
- Department of Neurophysiology, Medical FacultyRuhr University BochumBochumGermany
| | - Juliane Böge
- Department of Neurophysiology, Medical FacultyRuhr University BochumBochumGermany
| | - Olena Shchyglo
- Department of Neurophysiology, Medical FacultyRuhr University BochumBochumGermany
| | - Valentyna Dubovyk
- Department of Neurophysiology, Medical FacultyRuhr University BochumBochumGermany
| | | |
Collapse
|
16
|
Yang S, Xue J, Li Z, Zhang S, Zhang Z, Huang Z, Yung KKL, Lai KWC. Deep Learning-Based Ion Channel Kinetics Analysis for Automated Patch Clamp Recording. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404166. [PMID: 39737527 PMCID: PMC12083860 DOI: 10.1002/advs.202404166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/07/2024] [Indexed: 01/01/2025]
Abstract
The patch clamp technique is a fundamental tool for investigating ion channel dynamics and electrophysiological properties. This study proposes the first artificial intelligence framework for characterizing multiple ion channel kinetics of whole-cell recordings. The framework integrates machine learning for anomaly detection and deep learning for multi-class classification. The anomaly detection excludes recordings that are incompatible with ion channel behavior. The multi-class classification combined a 1D convolutional neural network, bidirectional long short-term memory, and an attention mechanism to capture the spatiotemporal patterns of the recordings. The framework achieves an accuracy of 97.58% in classifying 124 test datasets into six categories based on ion channel kinetics. The utility of the novel framework is demonstrated in two applications: Alzheimer's disease drug screening and nanomatrix-induced neuronal differentiation. In drug screening, the framework illustrates the inhibitory effects of memantine on endogenous channels, and antagonistic interactions among potassium, magnesium, and calcium ion channels. For nanomatrix-induced differentiation, the classifier indicates the effects of differentiation conditions on sodium and potassium channels associated with action potentials, validating the functional properties of differentiated neurons for Parkinson's disease treatment. The proposed framework is promising for enhancing the efficiency and accuracy of ion channel kinetics analysis in electrophysiological research.
Collapse
Affiliation(s)
- Shengjie Yang
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| | - Jiaqi Xue
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| | - Ziqi Li
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| | - Shiqing Zhang
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug ResearchCollege of PharmacyJinan University601 West Huangpu Road, TianheGuangzhou510632China
| | - Zhang Zhang
- School of Public HealthGuangzhou Medical UniversityXinzao, PanyuGuangzhou511436China
| | - Zhifeng Huang
- Department of ChemistryChinese University of Hong KongShatinNew TerritoriesHong Kong SARChina
| | - Ken Kin Lam Yung
- Department of Science and Environmental StudiesEducation University of Hong Kong10 Lo Ping RoadTai PoNew TerritoriesHong Kong SARChina
| | - King Wai Chiu Lai
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| |
Collapse
|
17
|
Garofalo S, Mormino A, Mazzarella L, Cocozza G, Rinaldi A, Di Pietro E, Di Castro MA, De Felice E, Maggi L, Chece G, Andolina D, Ventura R, Ielpo D, Piacentini R, Catalano M, Stefanini L, Limatola C. Platelets tune fear memory in mice. Cell Rep 2025; 44:115261. [PMID: 39903668 DOI: 10.1016/j.celrep.2025.115261] [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: 07/03/2024] [Revised: 11/13/2024] [Accepted: 01/13/2025] [Indexed: 02/06/2025] Open
Abstract
Several lines of evidence have shown that platelet-derived factors are key molecules in brain-body communication in pathological conditions. Here, we identify platelets as key actors in the modulation of fear behaviors in mice through the control of inhibitory neurotransmission and plasticity in the hippocampus. Interfering with platelet number or activation reduces hippocampal serotonin (5-HT) and modulates fear learning and memory in mice, and this effect is reversed by serotonin replacement by serotonin precursor (5-HTP)/benserazide. In addition, we unravel that natural killer (NK) cells participate in this mechanism, regulating interleukin-13 (IL-13) levels in the gut, with effects on serotonin production by enterochromaffin cells and uptake by platelets. Both NK cells and platelet depletion reduce the activation of hippocampal inhibitory neurons and increase the long-term potentiation of synaptic transmission. Understanding the role of platelets in the modulation of neuro-immune interactions offers additional tools for the definition of the molecular and cellular elements involved in the growing field of brain-body communication.
Collapse
Affiliation(s)
- Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.
| | - Alessandro Mormino
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Letizia Mazzarella
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Germana Cocozza
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Arianna Rinaldi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Erika Di Pietro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | | | - Eleonora De Felice
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Giuseppina Chece
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Diego Andolina
- Department of Psychology and Center for Research in Neurobiology 'D. Bovet', Sapienza University of Rome, Rome, Italy
| | - Rossella Ventura
- Department of Psychology and Center for Research in Neurobiology 'D. Bovet', Sapienza University of Rome, Rome, Italy
| | - Donald Ielpo
- Department of Psychology and Center for Research in Neurobiology 'D. Bovet', Sapienza University of Rome, Rome, Italy
| | - Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; IRCCS Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli 1, Roma, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Lucia Stefanini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Cristina Limatola
- Laboratory affiliated with Istituto Pasteur, Department of Physiology and Pharmacology, Sapienza University, Rome, Italy; IRCCS Neuromed, Pozzilli, Italy.
| |
Collapse
|
18
|
Ramnauth AD, Tippani M, Divecha HR, Papariello AR, Miller RA, Nelson ED, Thompson JR, Pattie EA, Kleinman JE, Maynard KR, Collado-Torres L, Hyde TM, Martinowich K, Hicks SC, Page SC. Spatiotemporal analysis of gene expression in the human dentate gyrus reveals age-associated changes in cellular maturation and neuroinflammation. Cell Rep 2025; 44:115300. [PMID: 40009515 DOI: 10.1016/j.celrep.2025.115300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/19/2024] [Accepted: 01/21/2025] [Indexed: 02/28/2025] Open
Abstract
The dentate gyrus of the hippocampus is important for many cognitive functions, including learning, memory, and mood. Here, we present transcriptome-wide spatial gene expression maps of the human dentate gyrus and investigate age-associated changes across the lifespan. Genes associated with neurogenesis and the extracellular matrix are enriched in infants and decline throughout development and maturation. Following infancy, inhibitory neuron markers increase, and cellular proliferation markers decrease. We also identify spatio-molecular signatures that support existing evidence for protracted maturation of granule cells during adulthood and age-associated increases in neuroinflammation-related gene expression. Our findings support the notion that the hippocampal neurogenic niche undergoes major changes following infancy and identify molecular regulators of brain aging in glial- and neuropil-enriched tissue.
Collapse
Affiliation(s)
- Anthony D Ramnauth
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Heena R Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Alexis R Papariello
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Ryan A Miller
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Erik D Nelson
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Jacqueline R Thompson
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Elizabeth A Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD 21205, USA
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Center for Computational Biology, Johns Hopkins University, Baltimore, MD 21205, USA; Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
19
|
Griesius S, Richardson A, Kullmann DM. Supralinear dendritic integration in murine dendrite-targeting interneurons. eLife 2025; 13:RP100268. [PMID: 39887034 PMCID: PMC11785373 DOI: 10.7554/elife.100268] [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] [Indexed: 02/01/2025] Open
Abstract
Non-linear summation of synaptic inputs to the dendrites of pyramidal neurons has been proposed to increase the computation capacity of neurons through coincidence detection, signal amplification, and additional logic operations such as XOR. Supralinear dendritic integration has been documented extensively in principal neurons, mediated by several voltage-dependent conductances. It has also been reported in parvalbumin-positive hippocampal basket cells, in dendrites innervated by feedback excitatory synapses. Whether other interneurons, which support feed-forward or feedback inhibition of principal neuron dendrites, also exhibit local non-linear integration of synaptic excitation is not known. Here, we use patch-clamp electrophysiology, and two-photon calcium imaging and glutamate uncaging, to show that supralinear dendritic integration of near-synchronous spatially clustered glutamate-receptor mediated depolarization occurs in NDNF-positive neurogliaform cells and oriens-lacunosum moleculare interneurons in the mouse hippocampus. Supralinear summation was detected via recordings of somatic depolarizations elicited by uncaging of glutamate on dendritic fragments, and, in neurogliaform cells, by concurrent imaging of dendritic calcium transients. Supralinearity was abolished by blocking NMDA receptors (NMDARs) but resisted blockade of voltage-gated sodium channels. Blocking L-type calcium channels abolished supralinear calcium signalling but only had a minor effect on voltage supralinearity. Dendritic boosting of spatially clustered synaptic signals argues for previously unappreciated computational complexity in dendrite-projecting inhibitory cells of the hippocampus.
Collapse
Affiliation(s)
- Simonas Griesius
- Department of Clinical Experimental and Epilepsy, UCL Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Amy Richardson
- Department of Clinical Experimental and Epilepsy, UCL Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Dimitri Michael Kullmann
- Department of Clinical Experimental and Epilepsy, UCL Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
| |
Collapse
|
20
|
Zerlaut Y, Tzilivaki A. Interneuronal modulations as a functional switch for cortical computations: mechanisms and implication for disease. Front Cell Neurosci 2025; 18:1479579. [PMID: 39916937 PMCID: PMC11799556 DOI: 10.3389/fncel.2024.1479579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/27/2024] [Indexed: 02/09/2025] Open
Abstract
Understanding cortical inhibition and its diverse roles remains a key challenge in neurophysiological research. Traditionally, inhibition has been recognized for controlling the stability and rhythmicity of network dynamics, or refining the spatiotemporal properties of cortical representations. In this perspective, we propose that specific types of interneurons may play a complementary role, by modulating the computational properties of neural networks. We review experimental and theoretical evidence, mainly from rodent sensory cortices, that supports this view. Additionally, we explore how dysfunctions in these interneurons may disrupt the network's ability to switch between computational modes, impacting the flexibility of cortical processing and potentially contributing to various neurodevelopmental and psychiatric disorders.
Collapse
Affiliation(s)
- Yann Zerlaut
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Alexandra Tzilivaki
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, Berlin, Germany
- Einstein Center for Neurosciences, Chariteplatz, Berlin, Germany
- NeuroCure Cluster of Excellence, Chariteplatz, Berlin, Germany
| |
Collapse
|
21
|
Kuang X, Chen S, Ye Q. The lactate metabolism and protein lactylation in epilepsy. Front Cell Neurosci 2025; 18:1464169. [PMID: 39876842 PMCID: PMC11772370 DOI: 10.3389/fncel.2024.1464169] [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: 07/13/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025] Open
Abstract
Protein lactylation is a new form of post-translational modification that has recently been proposed. Lactoyl groups, derived mainly from the glycolytic product lactate, have been linked to protein lactylation in brain tissue, which has been shown to correlate with increased neuronal excitability. Ischemic stroke may promote neuronal glycolysis, leading to lactate accumulation in brain tissue. This accumulation of lactate accumulation may heighten neuronal excitability by upregulating protein lactylation levels, potentially triggering post-stroke epilepsy. Although current clinical treatments for seizures have advanced significantly, approximately 30% of patients with epilepsy remain unresponsive to medication, and the prevalence of epilepsy continues to rise. This study explores the mechanisms of epilepsy-associated neuronal death mediated by lactate metabolism and protein lactylation. This study also examines the potential for histone deacetylase inhibitors to alleviate seizures by modifying lactylation levels, thereby offering fresh perspectives for future research into the pathogenesis and clinical treatment of epilepsy.
Collapse
Affiliation(s)
- Xi Kuang
- Hainan Health Vocational College, Haikou, China
| | - Shuang Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Qingmei Ye
- Hainan General Hospital and Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
22
|
E Said S, Miyamoto D. Multi-region processing during sleep for memory and cognition. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2025; 101:107-128. [PMID: 40074337 DOI: 10.2183/pjab.101.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Over the past decades, the understanding of sleep has evolved to be a fundamental physiological mechanism integral to the processing of different types of memory rather than just being a passive brain state. The cyclic sleep substates, namely, rapid eye movement (REM) sleep and non-REM (NREM) sleep, exhibit distinct yet complementary oscillatory patterns that form inter-regional networks between different brain regions crucial to learning, memory consolidation, and memory retrieval. Technical advancements in imaging and manipulation approaches have provided deeper understanding of memory formation processes on multi-scales including brain-wide, synaptic, and molecular levels. The present review provides a short background and outlines the current state of research and future perspectives in understanding the role of sleep and its substates in memory processing from both humans and rodents, with a focus on cross-regional brain communication, oscillation coupling, offline reactivations, and engram studies. Moreover, we briefly discuss how sleep contributes to other higher-order cognitive functions.
Collapse
Affiliation(s)
- Salma E Said
- Laboratory for Sleeping-Brain Dynamics, Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Daisuke Miyamoto
- Laboratory for Sleeping-Brain Dynamics, Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
23
|
Cupolillo D, Regio V, Barberis A. Synaptic microarchitecture: the role of spatial interplay between excitatory and inhibitory inputs in shaping dendritic plasticity and neuronal output. Front Cell Neurosci 2024; 18:1513602. [PMID: 39758273 PMCID: PMC11695373 DOI: 10.3389/fncel.2024.1513602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Affiliation(s)
| | | | - Andrea Barberis
- Istituto Italiano di Tecnologia, Synaptic Plasticity of Inhibitory Networks, Genova, Italy
| |
Collapse
|
24
|
Wang C, Wu B, Lin R, Cheng Y, Huang J, Chen Y, Bai J. Vagus nerve stimulation: a physical therapy with promising potential for central nervous system disorders. Front Neurol 2024; 15:1516242. [PMID: 39734634 PMCID: PMC11671402 DOI: 10.3389/fneur.2024.1516242] [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: 10/24/2024] [Accepted: 11/29/2024] [Indexed: 12/31/2024] Open
Abstract
The diseases of the central nervous system (CNS) often cause irreversible damage to the human body and have a poor prognosis, posing a significant threat to human health. They have brought enormous burdens to society and healthcare systems. However, due to the complexity of their causes and mechanisms, effective treatment methods are still lacking. Vagus nerve stimulation (VNS), as a physical therapy, has been utilized in the treatment of various diseases. VNS has shown promising outcomes in some CNS diseases and has been approved by the Food and Drug Administration (FDA) in the United States for epilepsy and depression. Moreover, it has demonstrated significant potential in the treatment of stroke, consciousness disorders, and Alzheimer's disease. Nevertheless, the exact efficacy of VNS, its beneficiaries, and its mechanisms of action remain unclear. This article discusses the current clinical evidence supporting the efficacy of VNS in CNS diseases, providing updates on the progress, potential, and potential mechanisms of action of VNS in producing effects on CNS diseases.
Collapse
Affiliation(s)
- Chaoran Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Postgraduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bangqi Wu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Ruolan Lin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Postgraduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yupei Cheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Postgraduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingjie Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Postgraduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuyan Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Postgraduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Bai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Postgraduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
25
|
Nakagawa N. The neuronal Golgi in neural circuit formation and reorganization. Front Neural Circuits 2024; 18:1504422. [PMID: 39703196 PMCID: PMC11655203 DOI: 10.3389/fncir.2024.1504422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
The Golgi apparatus is a central hub in the intracellular secretory pathway. By positioning in the specific intracellular region and transporting materials to spatially restricted compartments, the Golgi apparatus contributes to the cell polarity establishment and morphological specification in diverse cell types. In neurons, the Golgi apparatus mediates several essential steps of initial neural circuit formation during early brain development, such as axon-dendrite polarization, neuronal migration, primary dendrite specification, and dendritic arbor elaboration. Moreover, neuronal activity-dependent remodeling of the Golgi structure enables morphological changes in neurons, which provides the cellular basis of circuit reorganization during postnatal critical period. In this review, I summarize recent findings illustrating the unique Golgi positioning and its developmental dynamics in various types of neurons. I also discuss the upstream regulators for the Golgi positioning in neurons, and functional roles of the Golgi in neural circuit formation and reorganization. Elucidating how Golgi apparatus sculpts neuronal connectivity would deepen our understanding of the cellular/molecular basis of neural circuit development and plasticity.
Collapse
Affiliation(s)
- Naoki Nakagawa
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Japan
| |
Collapse
|
26
|
Rappe A, Vihinen HA, Suomi F, Hassinen AJ, Ehsan H, Jokitalo ES, McWilliams TG. Longitudinal autophagy profiling of the mammalian brain reveals sustained mitophagy throughout healthy aging. EMBO J 2024; 43:6199-6231. [PMID: 39367235 PMCID: PMC11612485 DOI: 10.1038/s44318-024-00241-y] [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: 11/17/2023] [Revised: 07/24/2024] [Accepted: 08/16/2024] [Indexed: 10/06/2024] Open
Abstract
Mitophagy neutralizes mitochondrial damage, thereby preventing cellular dysfunction and apoptosis. Defects in mitophagy have been strongly implicated in age-related neurodegenerative disorders such as Parkinson's and Alzheimer's disease. While mitophagy decreases throughout the lifespan of short-lived model organisms, it remains unknown whether such a decline occurs in the aging mammalian brain-a question of fundamental importance for understanding cell type- and region-specific susceptibility to neurodegeneration. Here, we define the longitudinal dynamics of basal mitophagy and macroautophagy across neuronal and non-neuronal cell types within the intact aging mouse brain in vivo. Quantitative profiling of reporter mouse cohorts from young to geriatric ages reveals cell- and tissue-specific alterations in mitophagy and macroautophagy between distinct subregions and cell populations, including dopaminergic neurons, cerebellar Purkinje cells, astrocytes, microglia and interneurons. We also find that healthy aging is hallmarked by the dynamic accumulation of differentially acidified lysosomes in several neural cell subsets. Our findings argue against any widespread age-related decline in mitophagic activity, instead demonstrating dynamic fluctuations in mitophagy across the aging trajectory, with strong implications for ongoing theragnostic development.
Collapse
Affiliation(s)
- Anna Rappe
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Helena A Vihinen
- Electron Microscopy Unit (EMBI), Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland
| | - Fumi Suomi
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Antti J Hassinen
- High Content Imaging and Analysis Unit (FIMM-HCA), Institute for Molecular Medicine, Helsinki Institute of Life Science, University of Helsinki, Tukholmankatu 8, Helsinki, 00290, Finland
| | - Homa Ehsan
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Eija S Jokitalo
- Electron Microscopy Unit (EMBI), Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland
| | - Thomas G McWilliams
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
| |
Collapse
|
27
|
Lacagnina AF, Dong TN, Iyer RR, Boesch LF, Khan S, Mohamed MK, Clem RL. Ventral hippocampal interneurons govern extinction and relapse of contextual associations. Cell Rep 2024; 43:114880. [PMID: 39425930 PMCID: PMC11665204 DOI: 10.1016/j.celrep.2024.114880] [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: 12/06/2023] [Revised: 08/02/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
Contextual memories are critical for survival but must be extinguished when new conditions render them nonproductive. By most accounts, extinction forms a new memory that competes with the original association for control over behavior, but the underlying circuit mechanisms remain largely enigmatic. Here, we demonstrate that extinction of contextual fear conditioning recruits somatostatin interneurons (SST-INs) in the ventral hippocampus. Correspondingly, real-time activity of SST-INs correlates with transitions between immobility and movement, signaling exit from defensive freezing bouts. Optogenetic manipulation of SST-INs but not parvalbumin interneurons (PV-INs) elicits bidirectional changes in freezing that are specific to the context in which extinction was acquired. Finally, similar effects were obtained following extinction of sucrose-based appetitive conditioning, in which SST-IN inhibition triggers relapse to reward seeking. These data suggest that ventral hippocampal SST-INs play a fundamental role in extinction that is independent of affective valence and may be related to their disruption of spontaneous emotional responses.
Collapse
Affiliation(s)
- Anthony F Lacagnina
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tri N Dong
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rasika R Iyer
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leonie F Boesch
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saqib Khan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mazen K Mohamed
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roger L Clem
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
28
|
Asim M, Qianqian G, Waris A, Wang H, Lai Y, Chen X. Unraveling the role of cholecystokinin in epilepsy: Mechanistic insight into neuroplasticity. Neurochem Int 2024; 180:105870. [PMID: 39343303 DOI: 10.1016/j.neuint.2024.105870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Epilepsy is a disorder characterized by an imbalance between excitability and inhibition, leading to uncontrolled hyperexcitability of neurons in the central nervous system. Despite the prevalence of epileptic seizures, the underlying mechanisms driving this hyperexcitability remain poorly understood. This review article aims to enhance our understanding of the mechanisms of epilepsy, with a specific focus on the role of cholecystokinin (CCK) in this debilitating disease. We will begin with an introduction to the topic, followed by an examination of the role of GABAergic neurons and the synaptic plasticity mechanisms associated with seizures. As we delve deeper, we will elucidate how CCK and its receptors contribute to seizure behavior. Finally, we will discuss the CCK-dependent synaptic plasticity mechanisms and highlight their potential implications in seizure activity. Through a comprehensive examination of these aspects, this review provides valuable insights into the involvement of CCK and its receptors in epilepsy. By improving our understanding of the mechanisms underlying this condition, particularly the role of CCK, we aim to contribute to the development of more effective treatment strategies.
Collapse
Affiliation(s)
- Muhammad Asim
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong; Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong.
| | - Gao Qianqian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Abdul Waris
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Huajie Wang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Yuanying Lai
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Xi Chen
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong; Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong
| |
Collapse
|
29
|
Sonsungsan P, Aimauthon S, Sriwichai N, Namchaiw P. Unveiling mitochondria as central components driving cognitive decline in alzheimer's disease through cross-transcriptomic analysis of hippocampus and entorhinal cortex microarray datasets. Heliyon 2024; 10:e39378. [PMID: 39498000 PMCID: PMC11534180 DOI: 10.1016/j.heliyon.2024.e39378] [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: 05/21/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 11/07/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by symptoms such as memory loss and impaired learning. This study conducted a cross-transcriptomic analysis of AD using existing microarray datasets from the hippocampus (HC) and entorhinal cortex (EC), comparing them with age-matched non-AD controls. Both of these brain regions are critical for learning and memory processing and are vulnerable areas that exhibit abnormalities in early AD. The cross-transcriptomic analysis identified 564 significantly differentially expressed genes in HC and 479 in EC. Among these, 151 genes were significantly differentially expressed in both tissues, with functions related to synaptic vesicle clustering, synaptic vesicle exocytosis/endocytosis, mitochondrial ATP synthesis, hydrogen ion transmembrane transport, and structural constituent of cytoskeleton, suggesting a potential association between cognitive decline in AD, synaptic vesicle dynamics, dysregulation of cytoskeleton organization, and mitochondrial dysfunction. Further gene ontology analysis specific to the HC revealed the gene ontology enrichment in aerobic respiration, synaptic vesicle cycle, and oxidative phosphorylation. The enrichment analysis in CA1 of HC revealed differentiation in gene expression related to mitochondrial membrane functions involved in bioenergetics, mitochondrial electron transport, and biological processes associated with microtubule-based process, while analysis in the EC region showed enrichment in synaptic vesicle dynamics which is associated with neurotransmitter release and the regulation of postsynaptic membrane potential and synaptic transmission of GABAergic and glutamatergic synapse. Protein-protein interaction analysis highlighted central hub proteins predominantly expressed in mitochondria, involved in regulation of oxidative stress and ATP synthesis. These hub proteins interact not only within the mitochondria but also with proteins in the vesicular membrane and neuronal cytoskeleton, indicating a central role of mitochondria. This finding underscores the association between clinical symptoms and mitochondrial dysregulation of synaptic vesicle dynamics, cytoskeleton organization, and mitochondrial processes in both the HC and EC of AD. Therefore, targeting these dysregulated pathways could provide promising therapeutic targets aimed at cognitive decline and memory impairment in early AD stages.
Collapse
Affiliation(s)
- Pajaree Sonsungsan
- Mathematics and Statistics, School of Science, Walailak University, Nakhon Si Thammarat, Thailand
| | - Supatha Aimauthon
- Chemical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
- Center for Biologics Research and Development, Chulabhorn Research Institute, Bangkok, Thailand
| | - Nattawet Sriwichai
- Center for Agricultural Systems Biology, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi (Bang Khun Thian), Bangkok, Thailand
| | - Poommaree Namchaiw
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
- Neuroscience Center for Research and Innovation, Learning Institute, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| |
Collapse
|
30
|
Xu T, Tan D, Wang Y, Gong C, Yuan J, Yang X, Wen Y, Ban Y, Liang M, Hu Y, Cao Y, Chen Y, Ran H. Targeted sonogenetic modulation of GABAergic interneurons in the hippocampal CA1 region in status epilepticus. Theranostics 2024; 14:6373-6391. [PMID: 39431014 PMCID: PMC11488105 DOI: 10.7150/thno.96598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 09/06/2024] [Indexed: 10/22/2024] Open
Abstract
Rationale: Sonogenetics is an advanced ultrasound-based neurostimulation approach for targeting neurons in specific brain regions. However, the role of sonogenetics in treating status epilepticus (SE) remains unclear. Here, we aimed to investigate the effects of ultrasound neurostimulation and MscL-G22S (a mechanosensitive ion channel that mediates Ca2+ influx)-mediated sonogenetics (MG-SOG) in a mouse model of kainic acid (KA)-induced SE. Methods: For MG-SOG, a Cre-dependent AAV expressing MscL-G22S was injected into parvalbumin (PV)-cre and somatostatin (SST)-cre mice to induce the expression of MscL-G22S-EGFP in PV interneurons (PV-INs) and SST interneurons (SST-INs), respectively; mice were stimulated with continuous pulses of ultrasound stimulation during the latency of generalized seizures (GSs), the latency to SE, in SE model mice. We performed calcium fiber photometry, patch-clamp recording, local field potential recording, and SE monitoring to investigate the role of MG-SOG in treating SE. Results: First, we observed obvious neuronal activation in the hippocampal CA1 region in SE model mice. Both excitatory neurons (ENs) and GABAergic interneurons (GABA-INs) in the CA1 region were activated in SE model mice; however, the inhibitory effect of GABA-INs on ENs seemed to be insufficient to reduce EN excitability despite the increased activation of GABA-INs in SE model mice. Thus, we speculated that MG-SOG-induced activation of GABA-INs, mainly SST-INs and PV-INs, in the CA1 region may protect against SE. We found that MG-SOG-mediated PV-IN activation in the CA1 region ameliorated SE and changed SE-related electrophysiological abnormalities in the CA1 region; however, MG-SOG-induced SST-IN activation in the CA1 region did not ameliorate SE. Conclusions: MG-SOG-mediated activation of PV-INs had a positive effect on relieving SE. Our work may promote the development of sonogenetic neurostimulation techniques for treating SE.
Collapse
Affiliation(s)
- Tao Xu
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
- Department of Ultrasound, Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Dandan Tan
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - You Wang
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Chen Gong
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jinxian Yuan
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiaolan Yang
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yuetao Wen
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yuenan Ban
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Minxue Liang
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yaqin Hu
- Department of Ultrasound, Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yang Cao
- Department of Ultrasound, Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yangmei Chen
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Haitao Ran
- Department of Ultrasound, Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
31
|
Lin ZJ, Gu X, Gong WK, Wang M, Wu YJ, Wang Q, Wu XR, Zhao XY, Zhu MX, Wang LY, Liu Q, Yuan TF, Li WG, Xu TL. Stimulation of an entorhinal-hippocampal extinction circuit facilitates fear extinction in a post-traumatic stress disorder model. J Clin Invest 2024; 134:e181095. [PMID: 39316444 PMCID: PMC11563685 DOI: 10.1172/jci181095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024] Open
Abstract
Effective psychotherapy of post-traumatic stress disorder (PTSD) remains challenging owing to the fragile nature of fear extinction, for which the ventral hippocampal CA1 (vCA1) region is considered as a central hub. However, neither the core pathway nor the cellular mechanisms involved in implementing extinction are known. Here, we unveil a direct pathway, where layer 2a fan cells in the lateral entorhinal cortex (LEC) target parvalbumin-expressing interneurons (PV-INs) in the vCA1 region to propel low-gamma-band synchronization of the LEC-vCA1 activity during extinction learning. Bidirectional manipulations of either hippocampal PV-INs or LEC fan cells sufficed for fear extinction. Gamma entrainment of vCA1 by deep brain stimulation (DBS) or noninvasive transcranial alternating current stimulation (tACS) of LEC persistently enhanced the PV-IN activity in vCA1, thereby promoting fear extinction. These results demonstrate that the LEC-vCA1 pathway forms a top-down motif to empower low-gamma-band oscillations that facilitate fear extinction. Finally, application of low-gamma DBS and tACS to a mouse model with persistent PTSD showed potent efficacy, suggesting that the dedicated LEC-vCA1 pathway can be stimulated for therapy to remove traumatic memory trace.
Collapse
Affiliation(s)
- Ze-Jie Lin
- Department of Anesthesiology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders (LEAD)
- Department of Anatomy and Physiology
| | - Xue Gu
- Department of Anatomy and Physiology
- Department of Anesthesiology, Shanghai General Hospital, and
| | - Wan-Kun Gong
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mo Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Yan-Jiao Wu
- Department of Anesthesiology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders (LEAD)
- Department of Anatomy and Physiology
| | - Qi Wang
- Department of Anesthesiology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders (LEAD)
- Department of Anatomy and Physiology
| | - Xin-Rong Wu
- Department of Anesthesiology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders (LEAD)
- Department of Anatomy and Physiology
| | - Xin-Yu Zhao
- Department of Anesthesiology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders (LEAD)
- Department of Anatomy and Physiology
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Lu-Yang Wang
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Quanying Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Guang Li
- Department of Anatomy and Physiology
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Ministry of Education–Shanghai Key Laboratory for Children’s Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian-Le Xu
- Department of Anesthesiology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders (LEAD)
- Department of Anatomy and Physiology
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
| |
Collapse
|
32
|
Ojima D, Tominaga Y, Kubota T, Tada A, Takahashi H, Kishimoto Y, Tominaga T, Yamamoto T. Impaired Hippocampal Long-Term Potentiation and Memory Deficits upon Haploinsufficiency of MDGA1 Can Be Rescued by Acute Administration of D-Cycloserine. Int J Mol Sci 2024; 25:9674. [PMID: 39273620 PMCID: PMC11394992 DOI: 10.3390/ijms25179674] [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: 08/08/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
The maintenance of proper brain function relies heavily on the balance of excitatory and inhibitory neural circuits, governed in part by synaptic adhesion molecules. Among these, MDGA1 (MAM domain-containing glycosylphosphatidylinositol anchor 1) acts as a suppressor of synapse formation by interfering with Neuroligin-mediated interactions, crucial for maintaining the excitatory-inhibitory (E/I) balance. Mdga1-/- mice exhibit selectively enhanced inhibitory synapse formation in their hippocampal pyramidal neurons, leading to impaired hippocampal long-term potentiation (LTP) and hippocampus-dependent learning and memory function; however, it has not been fully investigated yet if the reduction in MDGA1 protein levels would alter brain function. Here, we examined the behavioral and synaptic consequences of reduced MDGA1 protein levels in Mdga1+/- mice. As observed in Mdga1-/- mice, Mdga1+/- mice exhibited significant deficits in hippocampus-dependent learning and memory tasks, such as the Morris water maze and contextual fear-conditioning tests, along with a significant deficit in the long-term potentiation (LTP) in hippocampal Schaffer collateral CA1 synapses. The acute administration of D-cycloserine, a co-agonist of NMDAR (N-methyl-d-aspartate receptor), significantly ameliorated memory impairments and restored LTP deficits specifically in Mdga1+/- mice, while having no such effect on Mdga1-/- mice. These results highlight the critical role of MDGA1 in regulating inhibitory synapse formation and maintaining the E/I balance for proper cognitive function. These findings may also suggest potential therapeutic strategies targeting the E/I imbalance to alleviate cognitive deficits associated with neuropsychiatric disorders.
Collapse
Grants
- 16K08237, 19K07065, 19K07337, 16H06532, 24H01497, 23K18485, 23K21755, 21H03606, 23H03488, 23K28178, 23K21713, 22H05698, 24K18267, 21K15247, 19K07337, 22K06618 the Ministry of Education, Culture, Sports, Science and Technology, Japan
Collapse
Affiliation(s)
- Daiki Ojima
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Kagawa, Japan (A.T.); (H.T.)
| | - Yoko Tominaga
- Institute of Neuroscience, Tokushima Bunri University, Sanuki 769-2193, Kagawa, Japan
| | - Takashi Kubota
- Department of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki 769-2193, Kagawa, Japan; (T.K.); (Y.K.)
| | - Atsushi Tada
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Kagawa, Japan (A.T.); (H.T.)
| | - Hiroo Takahashi
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Kagawa, Japan (A.T.); (H.T.)
| | - Yasushi Kishimoto
- Department of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki 769-2193, Kagawa, Japan; (T.K.); (Y.K.)
- Laboratory of Physical Chemistry, Faculty of Pharmaceutical Sciences, Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan
| | - Takashi Tominaga
- Institute of Neuroscience, Tokushima Bunri University, Sanuki 769-2193, Kagawa, Japan
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki 769-2193, Kagawa, Japan
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho 761-0793, Kagawa, Japan (A.T.); (H.T.)
| |
Collapse
|
33
|
Maliković J, Amrein I, Vinciguerra L, Wolfer DP, Slomianka L. NECAB1-3, parvalbumin, calbindin, and calretinin in the hippocampus of the European mole. Front Neuroanat 2024; 18:1452722. [PMID: 39296922 PMCID: PMC11408328 DOI: 10.3389/fnana.2024.1452722] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024] Open
Abstract
Many calcium-binding proteins are expressed in a region-and cell-type specific manner in the mammalian hippocampus. Neuronal calcium-binding proteins (NECABs) are also expressed in hippocampal neurons, but few species have been investigated, with partly controversial findings. We here describe NECAB1, NECAB2 and NECAB3 as well as parvalbumin, calbindin, and calretinin in the European mole, and compare staining patterns of these proteins with those in mouse and other species. While subtle differences are present, NECAB staining in the European mole was generally similar to those in mouse. Common to European moles, mice, and other species we investigated, large hilar polymorphic cells, likely to represent mossy cells, were positive for all three NECABs. NECAB1 and 2 are suitable as markers for these cells along the entire septotemporal axis of the hippocampus. In the European mole, parvalbumin, calbindin and calretinin showed traits that have been described in other species before, albeit in a unique combination. In summary, we provide the first description of distribution of these proteins in the hippocampus of the European mole. This subterranean, insectivorous, and solitary living species belongs to the Order of Eulipotyphla. Despite many similarities with other subterranean species from the rodent order in terms of lifestyle, its hippocampus is cytoarchitecturally much more elaborated than in, e.g., mole-rats. It remains an open question if the hippocampal structure of the European mole reflects evolutionary constraints or ecology. Our descriptive study highlights the diversity in hippocampal cytoarchitecture even in small mammalian species.
Collapse
Affiliation(s)
- Jovana Maliković
- Division of Functional Neuroanatomy, Institute of Anatomy, University of Zürich, Zürich, Switzerland
| | - Irmgard Amrein
- Division of Functional Neuroanatomy, Institute of Anatomy, University of Zürich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | | | - David P Wolfer
- Division of Functional Neuroanatomy, Institute of Anatomy, University of Zürich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Lutz Slomianka
- Division of Functional Neuroanatomy, Institute of Anatomy, University of Zürich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
34
|
Moreira JVME, Bernardi LP, Teixeira FC, Paniago J, Teixeira LV, Bifi F, Souza DO, Rohden F. Spontaneously Hypertensive Rats Present Exacerbated Focal Stroke Behavioral Outcomes. Brain Sci 2024; 14:838. [PMID: 39199529 PMCID: PMC11352869 DOI: 10.3390/brainsci14080838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/13/2024] [Accepted: 08/17/2024] [Indexed: 09/01/2024] Open
Abstract
This study aimed to analyze the effects of systemic arterial hypertension (SAH) in a model of permanent ischemic stroke (focal ischemia due to thermocoagulation of pial vessels) on sensorimotor function (cylinder test and patch removal test), behavioral tasks (novelty habituation memory open field task) and cerebral infarct size in adult male spontaneously hypertensive rats (SHR) and normotensive Wistar Kyoto rats (WKY) for 42 days after the occurrence of a stroke. We observed that the stroke caused asymmetry in the front paws and delayed adhesive removal. These effects were spontaneously reduced in WKY rats, but not in SHR. Short- and long-term novelty habituation memories were abolished by stroke in WYK and SHR. On the 3rd day after stroke, the size of the focal cerebral infarct was the same in WKY and SHR. However, on the 7th day, the infarct size decreased in WKY rats, but not SHR. These results suggested that SAH impairment of sensorimotor recovery in rats subjected to cerebral ischemia could be related to augmented focal cerebral infarct size. Moreover, the behavioral tasks used in this study were unaffected by Systemic Arterial Hypertension. Our results highlight the need for animal models of comorbidities in stroke research.
Collapse
Affiliation(s)
- João Victor Matos e Moreira
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Annex Building, Ramiro Barcelos Street 2600, Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (J.V.M.e.M.); (L.P.B.); (J.P.); (L.V.T.); (F.B.); (D.O.S.)
| | - Luis Pedro Bernardi
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Annex Building, Ramiro Barcelos Street 2600, Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (J.V.M.e.M.); (L.P.B.); (J.P.); (L.V.T.); (F.B.); (D.O.S.)
| | - Fernanda Cardoso Teixeira
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre—UFCSPA, Porto Alegre 90050-170, Rio Grande do Sul, Brazil;
| | - Jerônimo Paniago
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Annex Building, Ramiro Barcelos Street 2600, Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (J.V.M.e.M.); (L.P.B.); (J.P.); (L.V.T.); (F.B.); (D.O.S.)
| | - Luciele Varaschini Teixeira
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Annex Building, Ramiro Barcelos Street 2600, Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (J.V.M.e.M.); (L.P.B.); (J.P.); (L.V.T.); (F.B.); (D.O.S.)
| | - Felippo Bifi
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Annex Building, Ramiro Barcelos Street 2600, Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (J.V.M.e.M.); (L.P.B.); (J.P.); (L.V.T.); (F.B.); (D.O.S.)
| | - Diogo Onofre Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Annex Building, Ramiro Barcelos Street 2600, Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (J.V.M.e.M.); (L.P.B.); (J.P.); (L.V.T.); (F.B.); (D.O.S.)
| | - Francieli Rohden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Annex Building, Ramiro Barcelos Street 2600, Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (J.V.M.e.M.); (L.P.B.); (J.P.); (L.V.T.); (F.B.); (D.O.S.)
| |
Collapse
|
35
|
Méndez P, de la Vega-Ruiz R, Montes-Mellado A. Estrogenic regulation of hippocampal inhibitory system across lifespan. J Neuroendocrinol 2024:e13441. [PMID: 39143852 DOI: 10.1111/jne.13441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
Estrogens produced in peripheral tissues and locally in the brain are potent neuromodulators. The function of the hippocampus, a brain region essential for episodic memory and spatial navigation, relies on the activity of ensembles of excitatory neurons whose activity is temporally and spatially coordinated by a wide diversity of inhibitory neurons (INs) types. Over the last years, we have accumulated evidence that indicates that estrogens regulate the function of hippocampal INs through different mechanisms, including transcriptional regulation and rapid nongenomic signaling. Here, we argue that the well-documented influence of estrogens on episodic memory may be related to the actions of local and peripheral estrogens on the heterogenous populations of hippocampal INs. We discuss how physiological changes in peripheral sex hormone levels throughout lifespan may interact with local brain sources to regulate IN function at different stages of life, from early hippocampal development to the aging brain. We conclude that considering INs as mediators of sex hormone actions in the hippocampus across the healthy life span will benefit our understanding of sex-biased neurodevelopmental disorders and physiological aging.
Collapse
|
36
|
Liao Z, Losonczy A. Learning, Fast and Slow: Single- and Many-Shot Learning in the Hippocampus. Annu Rev Neurosci 2024; 47:187-209. [PMID: 38663090 PMCID: PMC11519319 DOI: 10.1146/annurev-neuro-102423-100258] [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] [Indexed: 08/09/2024]
Abstract
The hippocampus is critical for memory and spatial navigation. The ability to map novel environments, as well as more abstract conceptual relationships, is fundamental to the cognitive flexibility that humans and other animals require to survive in a dynamic world. In this review, we survey recent advances in our understanding of how this flexibility is implemented anatomically and functionally by hippocampal circuitry, during both active exploration (online) and rest (offline). We discuss the advantages and limitations of spike timing-dependent plasticity and the more recently discovered behavioral timescale synaptic plasticity in supporting distinct learning modes in the hippocampus. Finally, we suggest complementary roles for these plasticity types in explaining many-shot and single-shot learning in the hippocampus and discuss how these rules could work together to support the learning of cognitive maps.
Collapse
Affiliation(s)
- Zhenrui Liao
- Department of Neuroscience and Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA;
| | - Attila Losonczy
- Department of Neuroscience and Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA;
| |
Collapse
|
37
|
Aksic M, Jakovcevski I, Hamad MIK, Jakovljevic V, Stankovic S, Vulovic M. The Neuroprotective Effect of Neural Cell Adhesion Molecule L1 in the Hippocampus of Aged Alzheimer's Disease Model Mice. Biomedicines 2024; 12:1726. [PMID: 39200191 PMCID: PMC11351965 DOI: 10.3390/biomedicines12081726] [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: 06/11/2024] [Revised: 07/22/2024] [Accepted: 07/27/2024] [Indexed: 09/02/2024] Open
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder and the most common form of dementia, causing the loss of cognitive function. Our previous study has shown, using a doubly mutated mouse model of AD (APP/PS1), that the neural adhesion molecule L1 directly binds amyloid peptides and decreases plaque load and gliosis when injected as an adeno-associated virus construct (AAV-L1) into APP/PS1 mice. In this study, we microinjected AAV-L1, using a Hamilton syringe, directly into the 3-month-old APP/PS1 mouse hippocampus and waited for a year until significant neurodegeneration developed. We stereologically counted the principal neurons and parvalbumin-positive interneurons in the hippocampus, estimated the density of inhibitory synapses around principal cells, and compared the AAV-L1 injection models with control injections of green fluorescent protein (AAV-GFP) and the wild-type hippocampus. Our results show that there is a significant loss of granule cells in the dentate gyrus of the APP/PS1 mice, which was improved by AAV-L1 injection, compared with the AAV-GFP controls (p < 0.05). There is also a generalized loss of parvalbumin-positive interneurons in the hippocampus of APP/PS1 mice, which is ameliorated by AAV-L1 injection, compared with the AAV-GFP controls (p < 0.05). Additionally, AAV-L1 injection promotes the survival of inhibitory synapses around the principal cells compared with AAV-GFP controls in all three hippocampal subfields (p < 0.01). Our results indicate that L1 promotes neuronal survival and protects the synapses in an AD mouse model, which could have therapeutic implications.
Collapse
Affiliation(s)
- Miljana Aksic
- Center for Medical Biochemistry, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (M.A.); (S.S.)
| | - Igor Jakovcevski
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany
- Department of Neuroanatomy and Molecular Brain Research, Institute of Anatomy, Ruhr Universität Bochum, 44801 Bochum, Germany
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 64141, United Arab Emirates
| | - Vladimir Jakovljevic
- Center of Excellence for Redox Balance Research, Cardiovascular and Metabolic Disorders, Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Sanja Stankovic
- Center for Medical Biochemistry, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (M.A.); (S.S.)
- Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Maja Vulovic
- Department of Anatomy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| |
Collapse
|
38
|
Sun H, Shen Y, Ni P, Liu X, Li Y, Qiu Z, Su J, Wang Y, Wu M, Kong X, Cao JL, Xie W, An S. Autism-associated neuroligin 3 deficiency in medial septum causes social deficits and sleep loss in mice. J Clin Invest 2024; 134:e176770. [PMID: 39058792 PMCID: PMC11444198 DOI: 10.1172/jci176770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Patients with autism spectrum disorder (ASD) frequently experience sleep disturbance. Genetic mutations in the neuroligin 3 (NLG3) gene are highly correlative with ASD and sleep disturbance. However, the cellular and neural circuit bases of this correlation remain elusive. Here, we found that the conditional knockout of Nlg3 (Nlg3-CKO) in the medial septum (MS) impairs social memory and reduces sleep. Nlg3 CKO in the MS caused hyperactivity of MSGABA neurons during social avoidance and wakefulness. Activation of MSGABA neurons induced social memory deficits and sleep loss in C57BL/6J mice. In contrast, inactivation of these neurons ameliorated social memory deficits and sleep loss in Nlg3-CKO mice. Sleep deprivation led to social memory deficits, while social isolation caused sleep loss, both resulting in a reduction in NLG3 expression and an increase in activity of GABAergic neurons in the MS from C57BL/6J mice. Furthermore, MSGABA-innervated CA2 neurons specifically regulated social memory without impacting sleep, whereas MSGABA-innervating neurons in the preoptic area selectively controlled sleep without affecting social behavior. Together, these findings demonstrate that the hyperactive MSGABA neurons impair social memory and disrupt sleep resulting from Nlg3 CKO in the MS, and achieve the modality specificity through their divergent downstream targets.
Collapse
Affiliation(s)
- Haiyan Sun
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Yu Shen
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Pengtao Ni
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Xin Liu
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Yan Li
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Zhentong Qiu
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Jiawen Su
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Yihan Wang
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Miao Wu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Xiangxi Kong
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Shuming An
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
39
|
Asmerian H, Alberts J, Sanetra AM, Diaz AJ, Silm K. Role of adaptor protein complexes in generating functionally distinct synaptic vesicle pools. J Physiol 2024. [PMID: 39034608 DOI: 10.1113/jp286179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024] Open
Abstract
The synaptic vesicle (SV) cycle ensures the release of neurotransmitters and the replenishment of SVs to sustain neuronal activity. Multiple endocytosis and sorting pathways contribute to the recapture of the SV membrane and proteins after fusion. Adaptor protein (AP) complexes are among the critical components of the SV retrieval machinery. The canonical clathrin adaptor AP2 ensures the replenishment of most SVs across many neuronal populations. An alternative AP1/AP3-dependent process mediates the formation of a subset of SVs that differ from AP2 vesicles in molecular composition and respond preferentially during higher frequency firing. Furthermore, recent studies show that vesicular transporters for different neurotransmitters depend to a different extent on the AP3 pathway and this affects the release properties of the respective neurotransmitters. This review focuses on the current understanding of the AP-dependent molecular and functional diversity among SVs. We also discuss the contribution of these pathways to the regulation of neurotransmitter release across neuronal populations.
Collapse
Affiliation(s)
- Hrach Asmerian
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jacob Alberts
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anna M Sanetra
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexia J Diaz
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Katlin Silm
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
40
|
Hadler MD, Alle H, Geiger JRP. Parvalbumin interneuron cell-to-network plasticity: mechanisms and therapeutic avenues. Trends Pharmacol Sci 2024; 45:586-601. [PMID: 38763836 DOI: 10.1016/j.tips.2024.04.003] [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: 03/31/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/21/2024]
Abstract
Alzheimer's disease (AD) and schizophrenia (SCZ) represent two major neuropathological conditions with a high disease burden. Despite their distinct etiologies, patients suffering from AD or SCZ share a common burden of disrupted memory functions unattended by current therapies. Recent preclinical analyses highlight cell-type-specific contributions of parvalbumin interneurons (PVIs), particularly the plasticity of their cellular excitability, towards intact neuronal network function (cell-to-network plasticity) and memory performance. Here we argue that deficits of PVI cell-to-network plasticity may underlie memory deficits in AD and SCZ, and we explore two therapeutic avenues: the targeting of PVI-specific neuromodulation, including by neuropeptides, and the recruitment of network synchrony in the gamma frequency range (40 Hz) by external stimulation. We finally propose that these approaches be merged under consideration of recent insights into human brain physiology.
Collapse
Affiliation(s)
- Michael D Hadler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Henrik Alle
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg R P Geiger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
41
|
Lai W, Luo R, Tang Y, Yu Z, Zhou B, Yang Z, Brown J, Hong G. Salidroside directly activates HSC70, revealing a new role for HSC70 in BDNF signalling and neurogenesis after cerebral ischemia. Phytother Res 2024; 38:2619-2640. [PMID: 38488455 DOI: 10.1002/ptr.8178] [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: 09/26/2023] [Revised: 01/17/2024] [Accepted: 02/11/2024] [Indexed: 06/13/2024]
Abstract
Salidroside, a principal bioactive component of Rhodiola crenulata, is neuroprotective across a wide time window in stroke models. We investigated whether salidroside induced neurogenesis after cerebral ischemia and aimed to identify its primary molecular targets. Rats, subjected to transient 2 h of middle cerebral artery occlusion (MCAO), received intraperitoneal vehicle or salidroside ± intracerebroventricular HSC70 inhibitor VER155008 or TrkB inhibitor ANA-12 for up to 7 days. MRI, behavioural tests, immunofluorescent staining and western blotting measured effects of salidroside. Reverse virtual docking and enzymatic assays assessed interaction of salidroside with purified recombinant HSC70. Salidroside dose-dependently decreased cerebral infarct volumes and neurological deficits, with maximal effects by 50 mg/kg/day. This dose also improved performance in beam balance and Morris water maze tests. Salidroside significantly increased BrdU+/nestin+, BrdU+/DCX+, BrdU+/NeuN+, BrdU-/NeuN+ and BDNF+ cells in the peri-infarct cortex, with less effect in striatum and no significant effect in the subventricular zone. Salidroside was predicted to bind with HSC70. Salidroside dose-dependently increased HSC70 ATPase and HSC70-dependent luciferase activities, but it did not activate HSP70. HSC70 immunoreactivity concentrated in the peri-infarct cortex and was unchanged by salidroside. However, VER155008 prevented salidroside-dependent increases of neurogenesis, BrdU-/NeuN+ cells and BDNF+ cells in peri-infarct cortex. Salidroside also increased BDNF protein and p-TrkB/TrkB ratio in ischemic brain, changes prevented by VER155008 and ANA-12, respectively. Additionally, ANA-12 blocked salidroside-dependent neurogenesis and increased BrdU-/NeuN+ cells in the peri-infarct cortex. Salidroside directly activates HSC70, thereby stimulating neurogenesis and neuroprotection via BDNF/TrkB signalling after MCAO. Salidroside and similar activators of HSC70 might provide clinical therapies for ischemic stroke.
Collapse
Affiliation(s)
- Wenfang Lai
- College of Pharmacology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Rui Luo
- College of Pharmacology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yuheng Tang
- College of Pharmacology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhengshuang Yu
- College of Pharmacology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Binbin Zhou
- College of Pharmacology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zelin Yang
- College of Pharmacology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - John Brown
- College of Pharmacology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Guizhu Hong
- College of Pharmacology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
42
|
Jakovljević A, Stamenković V, Poleksić J, Hamad MIK, Reiss G, Jakovcevski I, Andjus PR. The Role of Tenascin-C on the Structural Plasticity of Perineuronal Nets and Synaptic Expression in the Hippocampus of Male Mice. Biomolecules 2024; 14:508. [PMID: 38672524 PMCID: PMC11047978 DOI: 10.3390/biom14040508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Neuronal plasticity is a crucial mechanism for an adapting nervous system to change. It is shown to be regulated by perineuronal nets (PNNs), the condensed forms of the extracellular matrix (ECM) around neuronal bodies. By assessing the changes in the number, intensity, and structure of PNNs, the ultrastructure of the PNN mesh, and the expression of inhibitory and excitatory synaptic inputs on these neurons, we aimed to clarify the role of an ECM glycoprotein, tenascin-C (TnC), in the dorsal hippocampus. To enhance neuronal plasticity, TnC-deficient (TnC-/-) and wild-type (TnC+/+) young adult male mice were reared in an enriched environment (EE) for 8 weeks. Deletion of TnC in TnC-/- mice showed an ultrastructural reduction of the PNN mesh and an increased inhibitory input in the dentate gyrus (DG), and an increase in the number of PNNs with a rise in the inhibitory input in the CA2 region. EE induced an increased inhibitory input in the CA2, CA3, and DG regions; in DG, the change was also followed by an increased intensity of PNNs. No changes in PNNs or synaptic expression were found in the CA1 region. We conclude that the DG and CA2 regions emerged as focal points of alterations in PNNs and synaptogenesis with EE as mediated by TnC.
Collapse
Affiliation(s)
- Ana Jakovljević
- Center for Laser Microscopy, Institute for Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| | - Vera Stamenković
- Center for Integrative Brain Research, Seattle Children’s Research Institute, 1900 9th Ave, Seattle, WA 98125, USA;
| | - Joko Poleksić
- Institute of Anatomy “Niko Miljanic”, School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Gebhard Reiss
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany;
| | - Igor Jakovcevski
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany;
| | - Pavle R. Andjus
- Center for Laser Microscopy, Institute for Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
43
|
Heesen SH, Köhr G. GABAergic interneuron diversity and organization are crucial for the generation of human-specific functional neural networks in cerebral organoids. Front Cell Neurosci 2024; 18:1389335. [PMID: 38665372 PMCID: PMC11044699 DOI: 10.3389/fncel.2024.1389335] [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: 02/21/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
This mini review investigates the importance of GABAergic interneurons for the network function of human-induced pluripotent stem cells (hiPSC)-derived brain organoids. The presented evidence suggests that the abundance, diversity and three-dimensional cortical organization of GABAergic interneurons are the primary elements responsible for the creation of synchronous neuronal firing patterns. Without intricate inhibition, coupled oscillatory patterns cannot reach a sufficient complexity to transfer spatiotemporal information constituting physiological network function. Furthermore, human-specific brain network function seems to be mediated by a more complex and interconnected inhibitory structure that remains developmentally flexible for a longer period when compared to rodents. This suggests that several characteristics of human brain networks cannot be captured by rodent models, emphasizing the need for model systems like organoids that adequately mimic physiological human brain function in vitro.
Collapse
Affiliation(s)
- Sebastian H. Heesen
- Molecular and Behavioural Neurobiology, Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Georg Köhr
- Department of Neurophysiology, Mannheim Center for Translational Neurosciences, Heidelberg University, Mannheim, Germany
- Physiology of Neural Networks, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| |
Collapse
|
44
|
Wu Y, Hu H, Liu W, Zhao Y, Xie F, Sun Z, Zhang L, Dong H, Wang X, Qian L. Hippocampal Lactate-Infusion Enhances Spatial Memory Correlated with Monocarboxylate Transporter 2 and Lactylation. Brain Sci 2024; 14:327. [PMID: 38671979 PMCID: PMC11048250 DOI: 10.3390/brainsci14040327] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Lactate has emerged as a key player in regulating neural functions and cognitive processes. Beyond its function as an energy substrate and signal molecule, recent research has revealed lactate to serve as an epigenetic regulator in the brain. However, the molecular mechanisms by which lactate regulates spatial memory and its role in the prevention of cognitive disorders remain unclear. Herein, we injected L-lactate (10 μmol/kg/d for 6 d) into the mouse's hippocampus, followed by the Morris water maze (MWM) test and molecular analyses. Improved spatial memory performances were observed in mice injected with lactate. Besides, lactate upregulated the expression of synaptic proteins post-synaptic density 95 (PSD95), synaptophysin (SYP), and growth associated protein 43 (GAP43) in hippocampal tissues and HT22 cells, suggesting a potential role in synaptic transmission and memory formation. The facilitative role of monocarboxylate transporter 2 (MCT2), a neuron-specific lactate transporter, in this process was confirmed, as MCT2 antagonists attenuated the lactate-induced upregulation of synaptic proteins. Moreover, lactate induced protein lactylation, a post-translational modification, which could be suppressed by MCT2 inhibition. RNA sequencing of lactated-injected hippocampal tissues revealed a comprehensive gene expression profile influenced by lactate, with significant changes in genes associated with transcriptional progress. These data demonstrate that hippocampal lactate injection enhances spatial memory in mice, potentially through the upregulation of synaptic proteins and induction of protein lactylation, with MCT2 playing a crucial role in these processes. Our findings shed light on the multi-faceted role of lactate in neural function and memory regulation, opening new avenues for therapeutic interventions targeting cognitive disorders.
Collapse
Affiliation(s)
- Yuhan Wu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Hui Hu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Weiwei Liu
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
- College of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Yun Zhao
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Fang Xie
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Zhaowei Sun
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Ling Zhang
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Huafeng Dong
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| | - Xue Wang
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China
| | - Lingjia Qian
- Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, China; (Y.W.); (H.H.); (W.L.); (Y.Z.); (F.X.); (Z.S.); (L.Z.); (H.D.)
| |
Collapse
|
45
|
Nguyen R, Sivakumaran S, Lambe EK, Kim JC. Ventral hippocampal cholecystokinin interneurons gate contextual reward memory. iScience 2024; 27:108824. [PMID: 38303709 PMCID: PMC10831933 DOI: 10.1016/j.isci.2024.108824] [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: 04/21/2023] [Revised: 11/06/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Associating contexts with rewards depends on hippocampal circuits, with local inhibitory interneurons positioned to play an important role in shaping activity. Here, we demonstrate that the encoding of context-reward memory requires a ventral hippocampus (vHPC) to nucleus accumbens (NAc) circuit that is gated by cholecystokinin (CCK) interneurons. In a sucrose conditioned place preference (CPP) task, optogenetically inhibiting vHPC-NAc terminals impaired the acquisition of place preference. Transsynaptic rabies tracing revealed vHPC-NAc neurons were monosynaptically innervated by CCK interneurons. Using intersectional genetic targeting of CCK interneurons, ex vivo optogenetic activation of CCK interneurons increased GABAergic transmission onto vHPC-NAc neurons, while in vivo optogenetic inhibition of CCK interneurons increased cFos in these projection neurons. Notably, CCK interneuron inhibition during sucrose CPP learning increased time spent in the sucrose-associated location, suggesting enhanced place-reward memory. Our findings reveal a previously unknown hippocampal microcircuit crucial for modulating the strength of contextual reward learning.
Collapse
Affiliation(s)
- Robin Nguyen
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | | | - Evelyn K. Lambe
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of OBGYN, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Jun Chul Kim
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
46
|
García-Carlos CA, Basurto-Islas G, Perry G, Mondragón-Rodríguez S. Meta-Analysis in Transgenic Alzheimer's Disease Mouse Models Reveals Opposite Brain Network Effects of Amyloid-β and Phosphorylated Tau Proteins. J Alzheimers Dis 2024; 99:595-607. [PMID: 38669540 DOI: 10.3233/jad-231365] [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: 04/28/2024]
Abstract
Background Cognitive deficits observed in Alzheimer's disease (AD) patients have been correlated with altered hippocampal activity. Although the mechanism remains under extensive study, neurofibrillary tangles and amyloid plaques have been proposed as responsible for brain activity alterations. Aiming to unveil the mechanism, researchers have developed several transgenic models of AD. Nevertheless, the variability in hippocampal oscillatory alterations found in different genetic backgrounds and ages remains unclear. Objective To assess the oscillatory alterations in relation to animal developmental age and protein inclusion, amyloid-β (Aβ) load, and abnormally phosphorylated tau (pTau), we reviewed and analyzed the published data on peak power, frequency, and quantification of theta-gamma cross-frequency coupling (modulation index values). Methods To ensure that the search was as current as possible, a systematic review was conducted to locate and abstract all studies published from January 2000 to February 2023 that involved in vivo hippocampal local field potential recording in transgenic mouse models of AD. Results The presence of Aβ was associated with electrophysiological alterations that are mainly reflected in power increases, frequency decreases, and lower modulation index values. Concomitantly, pTau accumulation was associated with electrophysiological alterations that are mainly reflected in power decreases, frequency decreases, and no significant alterations in modulation index values. Conclusions In this study, we showed that electrophysiological parameters are altered from prodromal stages to the late stages of pathology. Thus, we found that Aβ deposition is associated with brain network hyperexcitability, whereas pTau deposition mainly leads to brain network hypoexcitability in transgenic models.
Collapse
Affiliation(s)
- Carlos Antonio García-Carlos
- UNAM Division of Neurosciences, Institute of Cellular Physiology, National Autonomous University of México, México City, México
| | | | - George Perry
- UTSA Neuroscience Institute and Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | - Siddhartha Mondragón-Rodríguez
- UAQ Centre for Applied Biomedical Research - CIBA, School of Medicine, Autonomous University of Querétaro, Querétaro, México
- CONAHCYT National Council for Science and Technology, México City, México
| |
Collapse
|
47
|
Lacagnina AF, Dong TN, Iyer RR, Khan S, Mohamed MK, Clem RL. Ventral hippocampal interneurons govern extinction and relapse of contextual associations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.28.568835. [PMID: 38077077 PMCID: PMC10705382 DOI: 10.1101/2023.11.28.568835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Contextual associations are critical for survival but must be extinguished when new conditions render them nonproductive. By most accounts, extinction forms a new memory that competes with the original association for control over behavior, but the mechanisms underlying this competition remain largely enigmatic. Here we find the retrieval of contextual fear conditioning and extinction yield contrasting patterns of activity in prefrontal cortex and ventral hippocampus. Within ventral CA1, activation of somatostatin-expressing interneurons (SST-INs) occurs preferentially during extinction retrieval and correlates with differences in input synaptic transmission. Optogenetic manipulation of these cells but not parvalbumin interneurons (PV-INs) elicits bidirectional changes in fear expression following extinction, and the ability of SST-INs to gate fear is specific to the context in which extinction was acquired. A similar pattern of results was obtained following reward-based extinction. These data show that ventral hippocampal SST-INs are critical for extinguishing prior associations and thereby gate relapse of both aversive and appetitive responses.
Collapse
|