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
|
Chu H, Sun N, Tang Q, Yu X, Li L, Chang H. Deciphering the polypharmacology of Dingzhi Xiaowan against comorbid depression: Integrated metabolomics of brain tissue and network pharmacology analysis in chronic restraint stress (CRS)-LPS model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119960. [PMID: 40350046 DOI: 10.1016/j.jep.2025.119960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Depression represents a prevalent and recurrent neuropsychiatric condition, with emerging evidence implicating glutamate (Glu) receptor dysfunction in the physiological causes of depression. Previous studies have highlighted Dingzhi Xiaowan (DZXW)'s antidepressant properties, attributing them to mechanisms like enhancing monoamine neurotransmitter release and reducing neuroinflammation. Nonetheless, a clearer understanding of how the glutamatergic system influences the antidepressant properties of DZXW remains to be explored. AIM OF THE STUDY This study aimed to elucidate the protective effects and mechanism of DZXW on depressive model mice by integrating metabolomics of brain tissue analysis and network pharmacology study. MATERIALS AND METHODS We initially assessed the therapeutic effects of DZXW on depression-like behaviors in mice using the OFT, FST, and SPT. Furthermore, we employed a variety of techniques, including HE and Nissl staining, ELISA, Golgi staining, immunofluorescence, and WB, to investigate how DZXW influences depression-like behaviors in mice. Additionally, we conducted metabolomic analyses and network pharmacology investigations to pinpoint the bioactive metabolites, essential components, and pathways linked to the therapeutic effects of DZXW in treating depression. Subsequently, after administering a TrkB receptor antagonist and an AMPA receptor antagonist, we replicated the behavioral evaluations, along with WB and immunofluorescence staining. RESULTS Behavioral assessments demonstrated that DZXW alleviated depression-like symptoms in the mouse CRS + LPS group. Notably, the antidepressant effects of DZXW were linked to a decrease in Glu levels and an improvement in neural synaptic plasticity. Metabolomics research identified 355 differential metabolites, of which DZXW significantly improved the levels of N-Acetylglutamine and N-Acetylaspartylglutamate. Combined with the network pharmacology indicating that components of DZXW, including Ginsenoside Rg1 and Sibiricose A1, may exert therapeutic effects via the glutamate metabolism and mTOR pathway. Furthermore, DZXW enhanced the protein levels of BDNF, TrkB, ERK, mTOR, and GluA1, while concurrently suppressing the expression of GluN1. Significantly, the introduction of TrkB and AMPA receptor antagonists effectively blocked the antidepressant properties of DZXW, indicating a potential crosstalk between the BDNF-TrkB-ERK-mTOR pathway and glutamatergic signaling. CONCLUSIONS The findings of this study support the idea that DZXW mitigates depression through the pathway associated with glutamate metabolism. Its antidepressant properties largely stem from enhancing the BDNF-TrkB-ERK-mTOR pathway, boosting the expression of AMPA receptors while concurrently inhibiting the expression of N-methyl-d-aspartate (NMDA) receptors. This research serves as a fresh reference point for identifying new antidepressant treatments.
Collapse
Affiliation(s)
- Haolin Chu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Nan Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Qin Tang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China; Pharmacy Department, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China.
| | - Xue Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Lina Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Hongsheng Chang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
3
|
Gong H, Xu X, Talifu Z, Zhang CJ, Sun YZ, Yue ZM, Rao JS, Du LJ, Du XX. Prospects and challenges in NMDAR signaling in spinal cord injury recovery and neural circuit remodeling. Regen Ther 2025; 29:381-389. [PMID: 40265135 PMCID: PMC12013404 DOI: 10.1016/j.reth.2025.03.008] [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: 01/22/2025] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/24/2025] Open
Abstract
N-methyl-d-aspartate receptors (NMDARs) are essential for excitatory synaptic transmission in the central nervous system, contributing to various physiological and pathological functions including learning, memory, neural development, synaptic transmission, and plasticity. NMDAR signaling plays a role in spinal cord injury outcomes, including restoring spinal circuits, modulating synaptic plasticity, reinstating synchronized functions, enhancing motor capabilities, and reducing neuropathic pain. Consequently, targeting NMDARs may serve as a promising approach to enhance axonal regeneration and reorganization of neural circuits following spinal injury.
Collapse
Affiliation(s)
- Han Gong
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Beijing International Cooperation Bases for Science and Technology on Biomaterials and Neural Regeneration, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
- University of Health and Rehabilitation Sciences, Qingdao City, 266113, China
- School of Rehabilitation, Capital Medical University, Beijing, 100086, China
- China Rehabilitation Research Center, Beijing, 100086, China
| | - Xin Xu
- University of Health and Rehabilitation Sciences, Qingdao City, 266113, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250100, China
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, 250100, China
| | - Zuliyaer Talifu
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Chun-Jia Zhang
- Department of Rehabilitation Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yu-Zhe Sun
- School of Rehabilitation, Capital Medical University, Beijing, 100086, China
- China Rehabilitation Research Center, Beijing, 100086, China
| | - Zhao-Ming Yue
- School of Rehabilitation, Capital Medical University, Beijing, 100086, China
- China Rehabilitation Research Center, Beijing, 100086, China
| | - Jia-Sheng Rao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Beijing International Cooperation Bases for Science and Technology on Biomaterials and Neural Regeneration, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| | - Liang-Jie Du
- School of Rehabilitation, Capital Medical University, Beijing, 100086, China
- China Rehabilitation Research Center, Beijing, 100086, China
| | - Xiao-Xia Du
- University of Health and Rehabilitation Sciences, Qingdao City, 266113, China
- School of Rehabilitation, Capital Medical University, Beijing, 100086, China
- China Rehabilitation Research Center, Beijing, 100086, China
| |
Collapse
|
4
|
Wosnitzka E, Gambarotto L, Nikoletopoulou V. Macroautophagy at the service of synapses. Curr Opin Neurobiol 2025; 93:103054. [PMID: 40414166 DOI: 10.1016/j.conb.2025.103054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/02/2025] [Accepted: 05/02/2025] [Indexed: 05/27/2025]
Abstract
Post-mitotic and highly polarized neurons are dependent on the fitness of their synapses, which are often found a long distance away from the soma. How the synaptic proteome is maintained, dynamically reshaped, and continuously turned over is a topic of intense investigation. Autophagy, a highly conserved, lysosome-mediated degradation pathway has emerged as a vital component of long-term neuronal maintenance, and now more specifically of synaptic homeostasis. Here, we review the most recent findings on how autophagy undergoes both dynamic and local regulation at the synapse, and how it contributes to pre- and post-synaptic proteostasis and function. We also discuss the insights and open questions that this new evidence brings.
Collapse
Affiliation(s)
- Erin Wosnitzka
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005, Lausanne, Switzerland
| | - Lisa Gambarotto
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005, Lausanne, Switzerland
| | - Vassiliki Nikoletopoulou
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005, Lausanne, Switzerland.
| |
Collapse
|
5
|
Sonoda T, Stephany CÉ, Kelley K, Kang D, Wu R, Uzgare MR, Fagiolini M, Greenberg ME, Chen C. Experience influences the refinement of feature selectivity in the mouse primary visual thalamus. Neuron 2025; 113:1352-1362.e4. [PMID: 40112812 DOI: 10.1016/j.neuron.2025.02.023] [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: 10/17/2023] [Revised: 01/15/2025] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Neurons exhibit selectivity for specific features: a property essential for extracting and encoding relevant information in the environment. This feature selectivity is thought to be modifiable by experience at the level of the cortex. Here, we demonstrate that selective exposure to a feature during development can alter the population representation of that feature in the primary visual thalamus. This thalamic plasticity is not due to changes in corticothalamic inputs and is blocked in mutant mice that exhibit deficits in retinogeniculate refinement, suggesting that plasticity is a direct result of changes in feedforward connectivity. Notably, experience-dependent changes in thalamic feature selectivity also occur in adult animals, although these changes are transient, unlike in juvenile animals, where they are long lasting. These results reveal an unexpected degree of plasticity in the visual thalamus and show that salient environmental features can be encoded in thalamic circuits during a discrete developmental window.
Collapse
Affiliation(s)
- Takuma Sonoda
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Céleste-Élise Stephany
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kaleb Kelley
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Di Kang
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Rui Wu
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Harvard-MIT Health Sciences and Technology Program, Harvard Medical School, Boston, MA 02115, USA
| | - Meghna R Uzgare
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Michela Fagiolini
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Chinfei Chen
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
6
|
Tu T, Cai XL, Sun ZP, Yang C, Jiang J, Wan L, Ai JQ, Wang Y, Coulibaly S, Tang BS, Wang J, Wang JH, Zhang Y, Wang XP, Peng Y, Wang H, Pan A, Yan XX, Tu E, Zhang QL. Mossy fiber expression of αSMA in human hippocampus and its relevance to brain evolution and neuronal development. Sci Rep 2025; 15:15834. [PMID: 40328887 PMCID: PMC12056149 DOI: 10.1038/s41598-025-00094-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
α-Smooth muscle actin (αSMA) is best characterized as the building block of thin filaments in smooth muscle cells. We observed a clear αSMA immunolabeling in adult human hippocampal mossy fibers (MF), prompting us to explore this novel pattern in phylogenic and ontogenic perspectives in the present study. αSMA immunolabeling occurred distinctively at the hippocampal MF terminals in humans from infancy to elderly. Hippocampal MF αSMA immunolabeling was not observed in mice and rats, visible in CA3 in guinea pigs and cats, and prominent in CA3 and dentate hilus in Rhesus monkeys. MF αSMA immunolabeling in human hippocampus emerged and refined from the last gestational trimester to early infancy. A transient overall neuronal labeling of ɑSMA was observed in prenatal human brains. ɑSMA expression was detected in human and rat primary neuronal cultures. The specificity of ɑSMA antibodies was confirmed by ACTA2 small interfering RNA (siRNA) silencing in SH-SY5Y cells. With this validation, we detected a higher αSMA protein level in dentate gyrus lysates relative to other human brain areas. Taken together, αSMA is distinctly expressed in human hippocampal mossy fibers. This pattern is related to hippocampal evolution among mammals and involves a refinement of neuronal αSMA expression during brain development.
Collapse
Affiliation(s)
- Tian Tu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Xiao-Lu Cai
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Zhong-Ping Sun
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Chen Yang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Juan Jiang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Lily Wan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Jia-Qi Ai
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Yan Wang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Sidiki Coulibaly
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jian Wang
- National Engineering and Research Center of Human Stem Cells, Changsha, 410119, Hunan, China
| | - Jian-Hong Wang
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, Yunnan, China
| | - Yan Zhang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Xiao-Ping Wang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Ying Peng
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410028, Hunan, China
| | - Hua Wang
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410028, Hunan, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China
| | - Ewen Tu
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, 410007, Hunan, China.
| | - Qi-Lei Zhang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, 410013, Hunan, China.
| |
Collapse
|
7
|
Miguez-Cabello F, Wang XT, Yan Y, Brake N, Alexander RPD, Perozzo AM, Khadra A, Bowie D. GluA2-containing AMPA receptors form a continuum of Ca 2+-permeable channels. Nature 2025; 641:537-544. [PMID: 40108453 DOI: 10.1038/s41586-025-08736-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025]
Abstract
Fast excitatory neurotransmission in the mammalian brain is mediated by cation-selective AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors (AMPARs)1. AMPARs are critical for the learning and memory mechanisms of Hebbian plasticity2 and glutamatergic synapse homeostasis3, with recent work establishing that AMPAR missense mutations can cause autism and intellectual disability4-7. AMPARs have been grouped into two functionally distinct tetrameric assemblies based on the inclusion or exclusion of the GluA2 subunit that determines Ca2+ permeability through RNA editing8,9. GluA2-containing AMPARs are the most abundant in the central nervous system and considered to be Ca2+ impermeable10. Here we show this is not the case. Contrary to conventional understanding, GluA2-containing AMPARs form a continuum of polyamine-insensitive ion channels with varying degrees of Ca2+ permeability. Their ability to transport Ca2+ is shaped by the subunit composition of AMPAR tetramers as well as the spatial orientation of transmembrane AMPAR regulatory proteins and cornichon auxiliary subunits. Ca2+ crosses the ion-conduction pathway by docking to an extracellular binding site that helps funnel divalent ions into the pore selectivity filter. The dynamic range in Ca2+ permeability, however, arises because auxiliary subunits primarily modify the selectivity filter. Taken together, our work proposes a broader role for AMPARs in Ca2+ signalling in the mammalian brain and offers mechanistic insight into the pathogenic nature of missense mutations.
Collapse
Affiliation(s)
| | - Xin-Tong Wang
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Yuhao Yan
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Niklas Brake
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- Quantitative Life Sciences PhD program, McGill University, Montreal, Quebec, Canada
| | - Ryan P D Alexander
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Amanda M Perozzo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Anmar Khadra
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Derek Bowie
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
8
|
Martinez P, Dutschmann M, Epercieux V, Gourjon G, Joulia F. Breath-hold diving as a tool to harness a beneficial increase in cardiac vagal tone. Respir Physiol Neurobiol 2025; 334:104416. [PMID: 40096873 DOI: 10.1016/j.resp.2025.104416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
Here we review central mechanisms that mediate the diving bradycardia and propose that breath-hold diving (BH-D) is a powerful therapeutic tool to improve cardiac vagal tone (CVT). Physiological fluctuations in CVT are known as the respiratory heart rate variability (respirHRV) and involve two respiratory-related brainstem mechanisms. During inspiration pre-Bötzinger complex (pre-BötC) neurons inhibit cardiac vagal motor neurons to increase heart rate and subsequently cardiac vagal disinhibition and a decrease in heart rate is associated with a Kölliker-Fuse (KF) nucleus-mediated partial glottal constriction during early expiration. Both KF and pre-BötC receive direct descending cortical inputs that could mediate volitional glottal closure as critical anatomical framework to volitionally target brainstem circuits that generate CVT during BH-D. Accordingly we show that volitional and reflex glottal closure during BH-D appropriates the respirHRV core network to mediate the diving bradycardia via converging trigeminal afferents inputs from the nose and forehead. Additional sensory inputs linked to prolonged BH-D after regular training further increase CVT during the acute dive and can yield a long-term increase in CVT. Centrally, evidence of Hebbian plasticity within respirHRV/BH-D core circuit further support the notion that regular BH-D exercise can yield a permanent increase in CVT specifically via a sensitization of synapse involved in the generation of the respirHRV. Contrary to other regular physical activity, BH-D reportedly does not cause structural remodeling of the heart and therefore we suggest that regular BH-D exercise could be employed as a save and non-invasive approach to treat sympathetic hyperactivity, particularly in elderly patients with cardio-vascular predispositions.
Collapse
Affiliation(s)
- Pierrick Martinez
- Laboratory « Jeunesse - Activité Physique et Sportive, Santé »(J-AP2S), University of Toulon, Toulon, France.
| | - Mathias Dutschmann
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University Hospitals Cleveland Medical Center and Case Western Reserve University, Cleveland, OH 44106, USA; Center for Sleep Disorders Research, Louis Stokes Cleveland VA Medical Center and Case Western Reserve University, Cleveland, OH 44106, USA
| | - Vincent Epercieux
- Aix Marseille Univ., CNRS, IBDM, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Géraud Gourjon
- Scientific and Osteopathic Research Department, Institut de Formation en Ostéopathie du Grand Avignon, 403 Rue Marcel Demonque, Avignon, France
| | - Fabrice Joulia
- Laboratory « Jeunesse - Activité Physique et Sportive, Santé »(J-AP2S), University of Toulon, Toulon, France; Center for Cardiovascular and Nutrition Research C2VN, INSERM 1263 INRAE 1260 Aix Marseille Université, Marseille, France
| |
Collapse
|
9
|
Zaater MA, El Kerdawy AM, Mahmoud WR, Abou-Seri SM. Going beyond ATP binding site as a novel inhibitor design strategy for tau protein kinases in the treatment of Alzheimer's disease: A review. Int J Biol Macromol 2025; 307:142141. [PMID: 40090653 DOI: 10.1016/j.ijbiomac.2025.142141] [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: 12/16/2024] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Alzheimer's disease (AD) is among the top mortality causing diseases worldwide. The presence of extracellular β-amyloidosis, as well as intraneuronal neurofibrillary aggregates of the abnormally hyperphosphorylated tau protein are two major characteristics of AD. Targeting protein kinases that are involved in the disease pathways has been a common approach in the fight against AD. Unfortunately, most kinase inhibitors currently available target the adenosine triphosphate (ATP)- binding site, which has proven unsuccessful due to issues with selectivity and resistance. As a result, a pressing need to find other alternative sites beyond the ATP- binding site has profoundly evolved. In this review, we will showcase some case studies of inhibitors of tau protein kinases acting beyond ATP binding site which have shown promising results in alleviating AD.
Collapse
Affiliation(s)
- Marwa A Zaater
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt
| | - Ahmed M El Kerdawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt; School of Health and Care Sciences, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, United Kingdom.
| | - Walaa R Mahmoud
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt
| | - Sahar M Abou-Seri
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt
| |
Collapse
|
10
|
Hou Y, Zuo Y, Song S, Zhang T. Long-term variable photoperiod exposure impairs hippocampal synapse involving of the glutamate system and leads to memory deficits in male Wistar rats. Exp Neurol 2025; 387:115191. [PMID: 39971149 DOI: 10.1016/j.expneurol.2025.115191] [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: 02/07/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
Excessive artificial light at night can induce the human circadian misalignment, potentially impairing memory consolidation and the rhythms of hippocampal clock genes. To investigate the impact of circadian misalignment on hippocampal function, we measured various field excitatory postsynaptic potentials (fEPSP) and golgi staining in the CA1 and dentate gyrus (DG) regions in Wistar rats. Our findings revealed that circadian misalignment resulted in a leftward shift in the input-output (I-O) curve within the CA1 region, decreased long-term potentiation (LTP), multi-time interval paired-pulse ratio (PPR), as well as dendritic spines and complexity across both CA1 and DG regions. Additionally, magnetic resonance spectroscopy (MRS) showed that circadian misalignment downregulated glutamate-related neurotransmitters (Glu + Gln) in the hippocampus, contributing to impaired synaptic function. Furthermore, disruptions to glutamate receptor subunits due to circadian misalignment led to reduced expression of AMPA receptor and NMDA receptor subunits in the hippocampus. In summary, our results suggest that memory impairments resulting from circadian misalignment are associated with diminished functionality within the glutamatergic system; this includes reductions in both Glx levels and availability of glutamate receptor subunits-key factors contributing to compromised synaptic function within the hippocampus.
Collapse
Affiliation(s)
- Yuanyuan Hou
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Yao Zuo
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, 550004, China
| | - Shaofei Song
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, 610041, China
| | - Tong Zhang
- Department of Neurological Rehabilitation, China Rehabilitation Research Center, Beijing Boai Hospital, Beijing, China; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China; School of Rehabilitation, Capital Medical University, Beijing 100068, China.
| |
Collapse
|
11
|
Langlais VC, Mountadem S, Benazzouz I, Amadio A, Matos M, Jourdes A, Cannich A, Julio-Kalajzic F, Belluomo I, Matias I, Maitre M, Lesté-Lasserre T, Marais S, Avignone E, Marsicano G, Bellocchio L, Oliet SHR, Panatier A. Astrocytic EphB3 receptors regulate d-serine-gated synaptic plasticity and memory. Prog Neurobiol 2025; 248:102747. [PMID: 40081519 DOI: 10.1016/j.pneurobio.2025.102747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/04/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
The activation of classical NMDA receptors (NMDARs) requires the binding of a co-agonist in addition to glutamate. Whereas astrocytic-derived d-serine was shown to play such a role at CA3-CA1 hippocampal synapses, the exact mechanism by which neurons interact with neighboring astrocytes to regulate synaptic d-serine availability remains to be fully elucidated. Considering the close anatomical apposition of astrocytic and neuronal elements at synapses, the aforementioned process is likely to involve cells adhesion molecules. One very likely candidate could be the astrocytic EphB3 receptor and its neuronal partner, ephrinB3. Here, we first showed in acute hippocampal slices from adult mice that stimulation of EphB3 receptors with exogenous ephrinB3 increased d-serine availability at CA3-CA1 synapses, resulting in an increased NMDAR activity. Conversely, inhibiting endogenous EphB3 receptors caused an impairment of both synaptic NMDAR activity and NMDAR-dependent long-term synaptic potentiation (LTP), effects that could be rescued by exogenous d-serine. Most interestingly, knocking down EphB3 receptors specifically in astrocytes yielded a similar impairment in hippocampal plasticity and, most importantly, caused a deficit in novel object recognition memory. Altogether, our data thus indicate that EphB3 receptors in hippocampal astrocytes play a key role in regulating synaptic NMDAR function, activity-dependent plasticity and memory.
Collapse
Affiliation(s)
| | - Sarah Mountadem
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Ines Benazzouz
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Aurélie Amadio
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Marco Matos
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Aurélie Jourdes
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Astrid Cannich
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Francisca Julio-Kalajzic
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France; Univ. Bordeaux, CNRS, INSERM, BIC, US4, UAR 3420, Bordeaux F-33000, France; Lead contact, France
| | - Ilaria Belluomo
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Isabelle Matias
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Marlène Maitre
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | | | - Sébastien Marais
- Univ. Bordeaux, CNRS, INSERM, BIC, US4, UAR 3420, Bordeaux F-33000, France
| | - Elena Avignone
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Giovanni Marsicano
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Luigi Bellocchio
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | | | - Aude Panatier
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux F-33000, France; Lead contact, France.
| |
Collapse
|
12
|
Couch T, McCullock TW, MacLean DM. The impact of subunit type, alternative splicing, and auxiliary proteins on AMPA receptor trafficking. J Biol Chem 2025; 301:108569. [PMID: 40316023 DOI: 10.1016/j.jbc.2025.108569] [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: 12/20/2024] [Revised: 03/31/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025] Open
Abstract
AMPA receptors underlie fast excitatory synaptic transmission in the mammalian nervous system and are critical for the expression of synaptic plasticity. Four genes encode the AMPA receptor subunits, each subject to RNA editing and alternative splicing at multiple positions. In addition, each tetrameric AMPA receptor can harbor up to four auxiliary proteins of which there are multiple types. Subunit type, alternative splicing, and auxiliary proteins are all known to affect AMPA receptor gating and trafficking. However, determining which factors dominate AMPA receptor trafficking requires high-throughput assessment of trafficking across multiple conditions. Here, we deploy two such methods to assess the relative contribution of AMPA receptor subunit type (GluA1 versus GluA2), alternative splicing (flip versus flop), and various transmembrane AMPA receptor regulatory proteins (TARPs) to AMPA receptor trafficking. We find that subunit type is the most important factor, with GluA2 showing a much better surface expression than GluA1, and alternative splicing plays a secondary role, with flip subunits consistently outperforming flop variants in surface expression across all conditions. Type 1 TARPs (γ2-4 and γ8) enhance surface trafficking, while Type 2 TARPs (γ5 and γ7) reduce surface expression, although we could not detect differences within each type. These data will be a helpful resource in comparing surface expression across a variety of AMPA receptor compositions. Our assays will also enable high-throughput assessment of novel disease-associated mutations, chimeras, and auxiliary and chaperone proteins.
Collapse
Affiliation(s)
- Tyler Couch
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Tyler W McCullock
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | - David M MacLean
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA.
| |
Collapse
|
13
|
Li YL, Li Y, Tu H, Evans AJ, Patel TA, Zheng H, Patel KP. Stellate Ganglia: A Key Therapeutic Target for Malignant Ventricular Arrhythmia in Heart Disease. Circ Res 2025; 136:1049-1069. [PMID: 40273204 PMCID: PMC12026290 DOI: 10.1161/circresaha.124.325384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Malignant ventricular arrhythmias (VAs), such as ventricular tachycardia and ventricular fibrillation, are the cause of approximately half a million deaths per year in the United States, which is a common lethal event in heart disease, such as hypertension, catecholaminergic polymorphic ventricular tachycardia, takotsubo cardiomyopathy, long-QT syndrome, and progressing into advanced heart failure. A common characteristic of these heart diseases, and the subsequent development of VAs, is the overactivation of the sympathetic nervous system. Current treatments for VAs in these heart diseases, such as β-adrenergic receptor blockers and cardiac sympathetic ablation, aim at inhibiting cardiac sympathetic overactivation. However, these treatments do not translate into becoming efficacious as long-term suppressors of ventricular tachycardia/ventricular fibrillation events. As a key regulatory component in the heart, cardiac postganglionic sympathetic neurons residing in the stellate ganglia (SGs) release neurotransmitters (such as norepinephrine and NPY [neuropeptide Y]) to perform their regulatory role in dictating cardiac function. Growing evidence from animal experiments and clinical studies has demonstrated that the remodeling of the SG may be intimately involved in malignant arrhythmogenesis. This identifies the SG as a key potential therapeutic target for the treatment of malignant VAs in heart disease. Therefore, this review summarizes the role of SG in ventricular arrhythmogenesis and updates the novel targeting of SG for clinical treatment of VAs in heart disease.
Collapse
Affiliation(s)
- Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yu Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Huiyin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anthony J. Evans
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tapan A. Patel
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hong Zheng
- Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Kaushik P. Patel
- Department of Cellular and Integrated Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
14
|
Menarchek BJ, Bridi MCD. Latent mechanisms of plasticity are upregulated during sleep. Curr Opin Neurobiol 2025; 93:103029. [PMID: 40267630 DOI: 10.1016/j.conb.2025.103029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/25/2025]
Abstract
Sleep is thought to serve an important role in learning and memory, but the mechanisms by which sleep promotes plasticity remain unclear. Even in the absence of plastic changes in neuronal function, many molecular, cellular, and physiological processes linked to plasticity are upregulated during sleep. Therefore, sleep may be a state in which latent plasticity mechanisms are poised to respond following novel experiences during prior wake. Many of these plasticity-related processes can promote both synaptic strengthening and weakening. Signaling pathways activated during sleep may interact with complements of proteins, determined by the content of prior waking experience, to establish the polarity of plasticity. Furthermore, precise reactivation of neuronal spiking patterns during sleep may interact with ongoing neuromodulatory, dendritic, and network activity to strengthen and weaken synapses. In this review, we will discuss the idea that sleep elevates latent plasticity mechanisms, which drive bidirectional plasticity depending on prior waking experience.
Collapse
Affiliation(s)
- Benjamin J Menarchek
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| | - Michelle C D Bridi
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA.
| |
Collapse
|
15
|
Sun Y, An P, Cai Y, Yang W, Fang Y, Liu H, Zhang G, Shan Y, Wang J, Zhang Y, Zhou X. Environmental enrichment reverses noise induced impairments in learning and memory associated with the hippocampus in female rats. Sci Rep 2025; 15:11509. [PMID: 40181175 PMCID: PMC11968901 DOI: 10.1038/s41598-025-96119-y] [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: 11/29/2024] [Accepted: 03/26/2025] [Indexed: 04/05/2025] Open
Abstract
Environmental enrichment (EE) has positive effects on brain function and behavior in both healthy and behaviorally impaired animals. In earlier studies, we showed that rats exposed to noise during early development exhibited deficits in learning and memory associated with the hippocampus. In this study, we investigated whether EE provided during adulthood can reverse such noise-induced impairments. We found that four weeks of EE substantially improved learning and memory in adult female rats exposed to noise during early development. The behavioral changes observed after EE were accompanied by the restoration of parvalbumin-positive (PV+) inhibitory interneurons in the hippocampal subregions. EE also reversed noise-induced reductions in hippocampal long-term potentiation (LTP) of synaptic connections, a mechanism essential for learning and memory processing. However, an enriched environment that lacked social interaction had little effect on restoring LTP in noise-exposed rats. These findings suggest that EE effectively mitigates hippocampal impairments that stem from early noise exposure, with social interaction playing a crucial role in this recovery process.
Collapse
Affiliation(s)
- Yutian Sun
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
- Institute of Brain and Cognitive Science, New York University-East China Normal University (NYU-ECNU), NYU-Shanghai, Shanghai, 200062, China
| | - Pengying An
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
- Institute of Brain and Cognitive Science, New York University-East China Normal University (NYU-ECNU), NYU-Shanghai, Shanghai, 200062, China
| | - Yongjian Cai
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
- Institute of Brain and Cognitive Science, New York University-East China Normal University (NYU-ECNU), NYU-Shanghai, Shanghai, 200062, China
| | - Wenjing Yang
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
- Institute of Brain and Cognitive Science, New York University-East China Normal University (NYU-ECNU), NYU-Shanghai, Shanghai, 200062, China
| | - Yue Fang
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
- Institute of Brain and Cognitive Science, New York University-East China Normal University (NYU-ECNU), NYU-Shanghai, Shanghai, 200062, China
| | - Hui Liu
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
- Institute of Brain and Cognitive Science, New York University-East China Normal University (NYU-ECNU), NYU-Shanghai, Shanghai, 200062, China
| | - Guimin Zhang
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
- Institute of Brain and Cognitive Science, New York University-East China Normal University (NYU-ECNU), NYU-Shanghai, Shanghai, 200062, China
| | - Ye Shan
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
| | - Jie Wang
- Department of Otolaryngology-Head and Neck Surgery, Wuhu Hospital, East China Normal University, Wuhu, 241000, China
| | - Yifan Zhang
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China.
- Institute of Brain and Cognitive Science, New York University-East China Normal University (NYU-ECNU), NYU-Shanghai, Shanghai, 200062, China.
| | - Xiaoming Zhou
- Key Laboratory of Brain Functional Genomics of Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China.
- Institute of Brain and Cognitive Science, New York University-East China Normal University (NYU-ECNU), NYU-Shanghai, Shanghai, 200062, China.
| |
Collapse
|
16
|
Shi H, Li Q, Hu F, Liu Y, Wang K. A novel role of the antidepressant paroxetine in inhibiting neuronal Kv7/M channels to enhance neuronal excitability. Transl Psychiatry 2025; 15:116. [PMID: 40175331 PMCID: PMC11965407 DOI: 10.1038/s41398-025-03291-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/18/2025] [Accepted: 02/18/2025] [Indexed: 04/04/2025] Open
Abstract
The voltage-gated Kv7/KCNQ/M potassium channels exert inhibitory control over neuronal membrane excitability. The reduction of Kv7 channel function can improve neuronal excitability that defines the fundamental mechanism of learning and memory. This suggests that pharmacological inhibition of Kv7 channels may present a therapeutic strategy for cognitive improvement. Paroxetine, a selective serotonin reuptake inhibitor, is widely used in the treatment of various types of depression with reported improvements in memory and attention. However, the exact mechanism underlying cognitive improvement by paroxetine remains poorly understood. In this study, we demonstrate that paroxetine inhibits whole-cell Kv7.2/Kv7.3 channel currents in a concentration-dependent manner with an IC50 of 3.6 ± 0.2 μΜ. In single-channel recording assay, paroxetine significantly reduces the open probability of Kv7.2/Kv7.3 channels. Moreover, paroxetine exhibits an inhibition of the native M-current and an increase in the firing of action potentials in hippocampal neurons. Taken together, our findings unveil a novel role of the antidepressant paroxetine in inhibiting M-current, providing insights into its pharmacological effects on cognition enhancement.
Collapse
Affiliation(s)
- Huan Shi
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical college, Qingdao, China
| | - Qinqin Li
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical college, Qingdao, China
| | - Fang Hu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical college, Qingdao, China
- Institute of Innovative Drugs, Qingdao University, Qingdao, China
| | - Yani Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical college, Qingdao, China.
- Institute of Innovative Drugs, Qingdao University, Qingdao, China.
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical college, Qingdao, China.
- Institute of Innovative Drugs, Qingdao University, Qingdao, China.
| |
Collapse
|
17
|
Wang BY, Wang B, Cao B, Gu LL, Chen J, He H, Zhao Z, Chen F, Wang Z. Associative Learning-Induced Synaptic Potentiation at the Two Major Hippocampal CA1 Inputs for Cued Memory Acquisition. Neurosci Bull 2025; 41:649-664. [PMID: 39604622 PMCID: PMC11979062 DOI: 10.1007/s12264-024-01327-7] [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: 05/31/2024] [Accepted: 09/03/2024] [Indexed: 11/29/2024] Open
Abstract
Learning-associated functional plasticity at hippocampal synapses remains largely unexplored. Here, in a single session of reward-based trace conditioning, we examine learning-induced synaptic plasticity in the dorsal CA1 hippocampus (dCA1). Local field-potential recording combined with selective optogenetic inhibition first revealed an increase of dCA1 synaptic responses to the conditioned stimulus (CS) induced during conditioning at both Schaffer collaterals to the stratum radiatum (Rad) and temporoammonic input to the lacunosum moleculare (LMol). At these dCA1 inputs, synaptic potentiation of CS-responding excitatory synapses was further demonstrated by locally blocking NMDA receptors during conditioning and whole-cell recording sensory-evoked synaptic responses in dCA1 neurons from naive animals. An overall similar time course of the induction of synaptic potentiation was found in the Rad and LMol by multiple-site recording; this emerged later and saturated earlier than conditioned behavioral responses. Our experiments demonstrate a cued memory-associated dCA1 synaptic plasticity induced at both Schaffer collaterals and temporoammonic pathways.
Collapse
Affiliation(s)
- Bing-Ying Wang
- Institute and Key Laboratory of Brain Functional Genomics of Chinese Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
| | - Bo Wang
- Institute and Key Laboratory of Brain Functional Genomics of Chinese Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
| | - Bo Cao
- Institute and Key Laboratory of Brain Functional Genomics of Chinese Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
| | - Ling-Ling Gu
- Institute and Key Laboratory of Brain Functional Genomics of Chinese Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
| | - Jiayu Chen
- Institute and Key Laboratory of Brain Functional Genomics of Chinese Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China
| | - Hua He
- Department of Neurosurgery, Third Affiliated Hospital of Navy Military Medical University, Shanghai, 200438, China
| | - Zheng Zhao
- Institute and Key Laboratory of Brain Functional Genomics of Chinese Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China.
| | - Fujun Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Zhiru Wang
- Institute and Key Laboratory of Brain Functional Genomics of Chinese Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, School of Life Sciences, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
18
|
Dixit N, Pyari G, Bansal H, Roy S. Theoretical analysis of low power optogenetic control of synaptic plasticity with subcellular expression of CapChR2 at postsynaptic spine. Sci Rep 2025; 15:11166. [PMID: 40169824 PMCID: PMC11962105 DOI: 10.1038/s41598-025-95355-6] [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: 09/26/2024] [Accepted: 03/20/2025] [Indexed: 04/03/2025] Open
Abstract
Precise control of intracellular calcium ([Formula: see text]) concentration at the synaptic neuron terminal can unravel the mechanism behind computation, learning, and memory formation inside the brain. Recently, the discovery of [Formula: see text]-permeable channelrhodopsins (CapChRs) has opened the opportunity to effectively control the intracellular [Formula: see text] concentration using optogenetics. Here, we present a new theoretical model for precise optogenetic control with newly discovered CapChR2 at postsynaptic neuron. A detailed theoretical analysis of coincident stimulation of presynaptic terminal, postsynaptic spine and optogenetic activation of CapChR2-expressing postsynaptic spine shows different ways to control postsynaptic intracellular [Formula: see text] concentration. Irradiance-dependent [Formula: see text] flow is an additional advantage of this novel method. The minimum threshold of light irradiance and optimal ranges of time lag among different stimulations and stimulation frequencies have also been determined. It is shown that synaptic efficacy occurs at 20 µW/mm2 at coincident electrical stimulation of presynaptic terminal and postsynaptic spine with optogenetic activation of CapChR2-expressed postsynaptic spine. The analysis provides a new means of direct optogenetic control of [Formula: see text]-based synaptic plasticity, better understanding of learning and memory processes, and opens prospects for targeted therapeutic interventions to modulate synaptic function and address various neurological disorders.
Collapse
Affiliation(s)
- Nripesh Dixit
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, 282005, India
| | - Gur Pyari
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, 282005, India
| | - Himanshu Bansal
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, 282005, India
| | - Sukhdev Roy
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, 282005, India.
| |
Collapse
|
19
|
Yasmin F, Marwick KFM, Hunter DW, Nawaz S, Marshall GF, Booker SA, Hardingham GE, Kind PC, Wyllie DJA. Absence of GluN2A in hippocampal CA1 neurons leads to altered dendritic structure and reduced frequency of miniature excitatory synaptic events. Brain Commun 2025; 7:fcaf124. [PMID: 40226380 PMCID: PMC11986202 DOI: 10.1093/braincomms/fcaf124] [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: 08/27/2024] [Revised: 02/04/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
GluN2A is a NMDA receptor subunit postulated as important for learning and memory. In humans, heterozygous loss of function variants in the gene encoding it (GRIN2A) increase the risk of epilepsy, intellectual disability and schizophrenia. Haploinsufficient mouse models show electrophysiological abnormalities and thus to improve and widen understanding of the pathogenesis of GRIN2A-associated disorders in humans, this study aimed to assess the impact of Grin2a absence and haploinsufficiency on core neuronal and synaptic properties in genetically modified rats. Electrophysiological whole-cell current- and voltage-clamp recordings were made from CA1 pyramidal neurons in acute hippocampal slices from wild-type and Grin2a heterozygous (Grin2a+/- ) and homozygous (Grin2a-/- ) knock out rats aged postnatal day 27-34. While reduced levels or absence of GluN2A did not affect neuronal excitability or intrinsic membrane properties in both Grin2a+/- and Grin2a-/- rats, we found a reduced frequency of miniature excitatory post synaptic currents and a reduced density of proximal dendrites suggestive of a reduced number of excitatory synapses. Recordings from CA1 neurons in slices prepared from Grin2a+/- and Grin2a-/- rats revealed there was a reduced ratio of the current mediated by NMDA receptors compared to AMPA receptors, while in Grin2a-/- recordings, there was a slowing of the decay time-constant of the NMDA receptor-mediated excitatory postsynaptic currents. Moreover, neither summation of sub-threshold excitatory postsynaptic potentials nor summation of supra-threshold excitatory postsynaptic potentials to initiate action potential firing in CA1 pyramidal neurons indicated any dependence on GluN2A. We conclude that reduced levels of GluN2A alters the kinetics of NMDA receptor-mediated synaptic events and dendritic structure of CA1 neurons, but do not affect several other core neuronal functions. These relatively subtle changes are consistent with the largely intact neural functioning of the majority of humans carrying GRIN2A loss of function variants. Further research could explore whether the changes in synaptic properties we observed contribute to alterations in higher level circuit dynamics and computation, which may manifest as disorders of cognition and excitability in humans.
Collapse
Affiliation(s)
- Farhana Yasmin
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Katie F M Marwick
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Daniel W Hunter
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Sarfaraz Nawaz
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Grant F Marshall
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Sam A Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh EH16 4SB, UK
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - David J A Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| |
Collapse
|
20
|
Rodríguez-García ME, Carino-Escobar RI, Carrillo-Mora P, Hernandez-Arenas C, Ramirez-Nava AG, Pacheco-Gallegos MDR, Valdés-Cristerna R, Cantillo-Negrete J. Neuroplasticity changes in cortical activity, grey matter, and white matter of stroke patients after upper extremity motor rehabilitation via a brain-computer interface therapy program. J Neural Eng 2025; 22:026025. [PMID: 40064104 DOI: 10.1088/1741-2552/adbebf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 03/10/2025] [Indexed: 03/21/2025]
Abstract
Objective. Upper extremity (UE) motor function loss is one of the most impactful consequences of stroke. Recently, brain-computer interface (BCI) systems have been utilized in therapy programs to enhance UE motor recovery after stroke, widely attributed to neuroplasticity mechanisms. However, the effect that the BCI's closed-loop feedback can have in these programs is unclear. The aim of this study was to quantitatively assess and compare the neuroplasticity effects elicited in stroke patients by a UE motor rehabilitation BCI therapy and by its sham-BCI counterpart.Approach. Twenty patients were randomly assigned to either the experimental group (EG), who controlled the BCI system via UE motor intention, or the control group (CG), who received random feedback. The elicited neuroplasticity effects were quantified using asymmetry metrics derived from electroencephalography (EEG), functional magnetic resonance imaging (fMRI), and diffusion tensor imaging (DTI) data acquired before, at the middle, and at the end of the intervention, alongside UE sensorimotor function evaluations. These asymmetry indexes compare the affected and unaffected hemispheres and are robust to lesion location variability.Main results. Most patients from the EG presented brain activity lateralisation to one brain hemisphere, as described by EEG (8 patients) and fMRI (6 patients) metrics. Conversely, the CG showed less pronounced lateralisations, presenting primarily bilateral activity patterns. DTI metrics showed increased white matter integrity in half of the EG patients' unaffected hemisphere, and in all but 2 CG patients' affected hemisphere. Individual patient analysis suggested that lesion location was relevant since functional and structural lateralisations occurred towards different hemispheres depending on stroke site.Significance. This study shows that a BCI intervention can elicit more pronounced neuroplasticity-related lateralisations than a sham-BCI therapy. These findings could serve as future biomarkers, helping to better select patients and increasing the impact that a BCI intervention can achieve. Clinical trial: NCT04724824.
Collapse
Affiliation(s)
| | - Ruben I Carino-Escobar
- Division of Research in Clinical Neuroscience, Instituto Nacional de Rehabilitación 'Luis Guillermo Ibarra Ibarra', Mexico City 14389, Mexico
| | - Paul Carrillo-Mora
- Division of Research in Clinical Neuroscience, Instituto Nacional de Rehabilitación 'Luis Guillermo Ibarra Ibarra', Mexico City 14389, Mexico
| | - Claudia Hernandez-Arenas
- Division of Neurological Rehabilitation, Instituto Nacional de Rehabilitación 'Luis Guillermo Ibarra Ibarra', Mexico City 14389, Mexico
| | - Ana G Ramirez-Nava
- Division of Neurological Rehabilitation, Instituto Nacional de Rehabilitación 'Luis Guillermo Ibarra Ibarra', Mexico City 14389, Mexico
| | | | - Raquel Valdés-Cristerna
- Electrical Engineering Department, Universidad Autónoma Metropolitana Unidad Iztapalapa, Mexico City 09340, Mexico
| | - Jessica Cantillo-Negrete
- Technological Research Subdirection, Instituto Nacional de Rehabilitación 'Luis Guillermo Ibarra Ibarra', Mexico City 14389, Mexico
| |
Collapse
|
21
|
Suhaimi FW, Khari NHM, Hassan Z, Müller CP. Exploring the cognitive effects of kratom: A review. Behav Brain Res 2025; 480:115387. [PMID: 39643045 DOI: 10.1016/j.bbr.2024.115387] [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: 09/23/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Despite the strict kratom regulation in some regions, the demand for kratom products is still increasing worldwide. Kratom products are commonly consumed for their pain-relieving effect or as a self-treatment for opioid use disorder. Kratom is also taken as a recreational drug among youth and adults. Since substance abuse can cause cognitive impairment, many studies investigated the effects of kratom on cognition. The interaction of some kratom alkaloids with various receptors such as opioid, serotonergic, and adrenergic receptors further sparks the interest to investigate the effects of kratom on cognitive function. Hence, this review aims to provide an overview of the effects of kratom on cognitive behaviours and their underlying changes in neurobiological mechanisms. In conclusion, kratom, particularly its main alkaloid, mitragynine may adversely affect cognitive performances that may be attributed to the disruption in synaptic plasticity, brain activity as well as various proteins involved in synaptic transmission. The impact of kratom on cognitive functions could also shed light on its safety profile, which is essential for the therapeutic development of kratom, including its potential use in opioid substitution therapy.
Collapse
Affiliation(s)
| | | | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
22
|
Radin DP, Lippa A, Rana S, Fuller DD, Smith JL, Cerne R, Witkin JM. Amplification of the therapeutic potential of AMPA receptor potentiators from the nootropic era to today. Pharmacol Biochem Behav 2025; 248:173967. [PMID: 39894310 PMCID: PMC11849398 DOI: 10.1016/j.pbb.2025.173967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptors (AMPA receptors or AMPARs) are involved in fast excitatory neurotransmission and as such control multiple important physiological processes. AMPARs also are involved in the dynamics of synaptic plasticity in the nervous system where they impact neuroplastic responses such as long-term facilitation and long-term potentiation that regulate biological functions ranging from breathing to cognition. AMPARs also regulate neurotrophic factors that are strategically involved in neural plastic changes in the nervous system. As with other major ionotropic receptors, modulation of AMPARs can have prominent effects on biological systems that can include marked tolerability issues. AMPAR potentiators (AMPAkines) are positive allosteric modulators of AMPARs which have therapeutic potential. Medicinal chemistry combined with new pharmacological findings have defined AMPAkines with favorable oral bioavailability and pharmacological safety parameters that enable clinical advancement of their therapeutic utility. AMPAkines are being investigated in patients with diverse neurological and psychiatric disorders including spinal cord injury (breathing and bladder function), cognition, attention-deficit-hyperactivity disorder, and major depressive disorder. The present discussion of this class of compounds focuses on their general value as therapeutics through their impact on synaptic plasticity.
Collapse
Affiliation(s)
| | - Arnold Lippa
- RespireRx Pharmaceuticals Inc., Glen Rock, NJ, USA
| | - Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery St. Vincent's Hospital, Indianapolis, IN, USA
| | - Rok Cerne
- RespireRx Pharmaceuticals Inc., Glen Rock, NJ, USA; Laboratory of Antiepileptic Drug Discovery St. Vincent's Hospital, Indianapolis, IN, USA; Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, Ljubljana, Slovenia
| | - Jeffrey M Witkin
- RespireRx Pharmaceuticals Inc., Glen Rock, NJ, USA; Laboratory of Antiepileptic Drug Discovery St. Vincent's Hospital, Indianapolis, IN, USA; Department of Neurosciene and Trauma Research, Ascension St. Vincent Hospital, Indianapolis IN, USA.
| |
Collapse
|
23
|
Nojiri E, Takase K. Understanding Sensory-Motor Disorders in Autism Spectrum Disorders by Extending Hebbian Theory: Formation of a Rigid-Autonomous Phase Sequence. PERSPECTIVES ON PSYCHOLOGICAL SCIENCE 2025; 20:276-289. [PMID: 37910043 DOI: 10.1177/17456916231202674] [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: 11/03/2023]
Abstract
Autism spectrum disorder is a neuropsychiatric disorder characterized by persistent deficits in social communication and social interaction and restricted, repetitive patterns of behavior, interests, or activities. The symptoms invariably appear in early childhood and cause significant impairment in social, occupational, and other important functions. Various abnormalities in the genetic, neurological, and endocrine systems of patients with autism spectrum disorder have been reported as the etiology; however, no clear factor leading to the onset of the disease has been identified. Additionally, higher order cognitive dysfunctions, which are represented by a lack of theory of mind, sensorimotor disorders, and memory-related disorders (e.g., flashbacks), have been reported in recent years, but no theoretical framework has been proposed to explain these behavioral abnormalities. In this study, we extended Hebb's biopsychology theory to provide a theoretical framework that comprehensively explains the various behavioral abnormalities observed in autism spectrum disorder. Specifically, we propose that a wide range of symptoms in autism spectrum disorder may be caused by the formation of a rigid-autonomous phase sequence (RAPS) in the brain. Using the RAPS formation theory, we propose a biopsychological mechanism that could be a target for the treatment of autism spectrum disorders.
Collapse
|
24
|
Balmer GL, Guha S, Poll S. Engrams across diseases: Different pathologies - unifying mechanisms? Neurobiol Learn Mem 2025; 219:108036. [PMID: 40023216 DOI: 10.1016/j.nlm.2025.108036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Memories are our reservoir of knowledge and thus, are crucial for guiding decisions and defining our self. The physical correlate of a memory in the brain is termed an engram and since decades helps researchers to elucidate the intricate nature of our imprinted experiences and knowledge. Given the importance that memories have for our lives, their impairment can present a tremendous burden. In this review we aim to discuss engram malfunctioning across diseases, covering dementia-associated pathologies, epilepsy, chronic pain and psychiatric disorders. Current neuroscientific tools allow to witness the emergence and fate of engram cells and enable their manipulation. We further suggest that specific mechanisms of mnemonic malfunction can be derived from engram cell readouts. While depicting the way diseases act on the mnemonic component - specifically, on the cellular engram - we emphasize a differentiation between forms of amnesia and hypermnesia. Finally, we highlight commonalities and distinctions of engram impairments on the cellular level across diseases independent of their pathogenic origins and discuss prospective therapeutic measures.
Collapse
Affiliation(s)
- Greta Leonore Balmer
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany
| | - Shuvrangshu Guha
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany
| | - Stefanie Poll
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE) Bonn, Germany.
| |
Collapse
|
25
|
Pannoni KE, Fischer QS, Tarannum R, Cawley ML, Alsalman MM, Acosta N, Ezigbo C, Gil DV, Campbell LA, Farris S. MCU expression in hippocampal CA2 neurons modulates dendritic mitochondrial morphology and synaptic plasticity. Sci Rep 2025; 15:4540. [PMID: 39915602 PMCID: PMC11802895 DOI: 10.1038/s41598-025-85958-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
Neuronal mitochondria are diverse across cell types and subcellular compartments in order to meet unique energy demands. While mitochondria are essential for synaptic transmission and synaptic plasticity, the mechanisms regulating mitochondria to support normal synapse function are incompletely understood. The mitochondrial calcium uniporter (MCU) is proposed to couple neuronal activity to mitochondrial ATP production, which would allow neurons to rapidly adapt to changing energy demands. MCU is uniquely enriched in hippocampal CA2 distal dendrites compared to proximal dendrites, however, the functional significance of this layer-specific enrichment is not clear. Synapses onto CA2 distal dendrites readily express plasticity, unlike the plasticity-resistant synapses onto CA2 proximal dendrites, but the mechanisms underlying these different plasticity profiles are unknown. Using a CA2-specific MCU knockout (cKO) mouse, we found that MCU deletion impairs plasticity at distal dendrite synapses. However, mitochondria were more fragmented and spine head area was diminished throughout the dendritic layers of MCU cKO mice versus control mice. Fragmented mitochondria might have functional changes, such as altered ATP production, that could explain the structural and functional deficits at cKO synapses. Differences in MCU expression across cell types and circuits might be a general mechanism to tune mitochondrial function to meet distinct synaptic demands.
Collapse
Affiliation(s)
- Katy E Pannoni
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Quentin S Fischer
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Renesa Tarannum
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Mikel L Cawley
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Mayd M Alsalman
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Nicole Acosta
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Chisom Ezigbo
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Daniela V Gil
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Logan A Campbell
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Shannon Farris
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA.
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA.
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA.
| |
Collapse
|
26
|
Blanco FA, Saifullah MAB, Cheng JX, Abella C, Scala F, Firozi K, Niu S, Park J, Chin J, Tolias KF. Targeting Tiam1 Enhances Hippocampal-Dependent Learning and Memory in the Adult Brain and Promotes NMDA Receptor-Mediated Synaptic Plasticity and Function. J Neurosci 2025; 45:e0298242024. [PMID: 39725519 PMCID: PMC11800756 DOI: 10.1523/jneurosci.0298-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 10/18/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Excitatory synapses and the actin-rich dendritic spines on which they reside are indispensable for information processing and storage in the brain. In the adult hippocampus, excitatory synapses must balance plasticity and stability to support learning and memory. However, the mechanisms governing this balance remain poorly understood. Tiam1 is an actin cytoskeleton regulator prominently expressed in the dentate gyrus (DG) throughout life. Previously, we showed that Tiam1 promotes dentate granule cell synapse and spine stabilization during development, but its role in the adult hippocampus remains unclear. Here, we deleted Tiam1 from adult forebrain excitatory neurons (Tiam1fKO ) and assessed the effects on hippocampal-dependent behaviors. Adult male and female Tiam1fKO mice displayed enhanced contextual fear memory, fear extinction, and spatial discrimination. Investigation into underlying mechanisms revealed that dentate granule cells from Tiam1fKO brain slices exhibited augmented synaptic plasticity and N-methyl-D-aspartate-type glutamate receptor (NMDAR) function. Additionally, Tiam1 loss in primary hippocampal neurons blocked agonist-induced NMDAR internalization, reduced filamentous actin levels, and promoted activity-dependent spine remodeling. Notably, strong NMDAR activation in wild-type hippocampal neurons triggered Tiam1 loss from spines. Our results suggest that Tiam1 normally constrains hippocampal-dependent learning and memory in the adult brain by restricting NMDAR-mediated synaptic plasticity in the DG. We propose that Tiam1 achieves this by limiting NMDAR availability at synaptic membranes and stabilizing spine actin cytoskeleton and that these constraints can be alleviated by activity-dependent degradation of Tiam1. These findings reveal a previously unknown mechanism restricting hippocampal synaptic plasticity and highlight Tiam1 as a therapeutic target for enhancing cognitive function.
Collapse
Affiliation(s)
- Francisco A Blanco
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | | | - Jinxuan X Cheng
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Carlota Abella
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Federico Scala
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Sanyong Niu
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Jin Park
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Memory & Brain Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Jeannie Chin
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Memory & Brain Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
27
|
Jones TA, Nemchek V, Fracassi M. Experience-driven competition in neural reorganization after stroke. J Physiol 2025; 603:737-757. [PMID: 39476290 PMCID: PMC11785499 DOI: 10.1113/jp285565] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/27/2024] [Indexed: 02/01/2025] Open
Abstract
Behavioural experiences interact with regenerative responses to shape patterns of neural reorganization after stroke. This review is focused on the competitive nature of these behavioural experience effects. Interactions between learning-related plasticity and regenerative reactions have been found to underlie the establishment of new compensatory behaviours and the efficacy of motor rehabilitative training in rodent stroke models. Learning in intact brains depends on competitive and cooperative mechanisms of synaptic plasticity. Synapses are added in response to learning and selectively maintained and strengthened via activity-dependent competition. Long-term memories for experiences that occur closely in time can be weakened or enhanced by competitive or cooperative interactions in the time-dependent process of stabilizing synaptic changes. Rodent stroke model findings suggest that compensatory reliance on the non-paretic hand after stroke can shape and stabilize synaptic reorganization patterns in both hemispheres, to compete with the capacity for experiences of the paretic side to do so. However, the competitive edge of the non-paretic side can be countered by overlapping experiences of the paretic hand, and might even be shifted in a cooperative direction with skilfully coordinated bimanual experience. Advances in the basic understanding of learning-related synaptic competition are helping to inform the basis of experience-dependent variations in stroke outcome.
Collapse
Affiliation(s)
- Theresa A. Jones
- Psychology Department & Institute for Neuroscience, University of Texas at Austin
| | - Victoria Nemchek
- Psychology Department & Institute for Neuroscience, University of Texas at Austin
| | - Michela Fracassi
- Psychology Department & Institute for Neuroscience, University of Texas at Austin
| |
Collapse
|
28
|
Dow CT, Kidess Z. Proposing Bromo-epi-androsterone (BEA) for perioperative neurocognitive disorders with Interleukin-6 as a druggable target. J Clin Anesth 2025; 101:111736. [PMID: 39746239 DOI: 10.1016/j.jclinane.2024.111736] [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: 10/23/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
Cognitive impairment following surgery is a significant complication, affecting multiple neurocognitive domains. The term "perioperative neurocognitive disorders" (PND) is recommended to encompass this entity. Individuals who develop PND are typically older and have increases in serum and brain pro-inflammatory cytokines notwithstanding the type of surgery undergone. Surgical trauma induces production of small biomolecules, damage-associated molecular patterns (DAMP), particularly the DAMP known as high molecular group box 1 protein (HMGB1). Mechanistically, peripheral surgical trauma promotes pro-inflammatory cytokines that stimulate central nervous system (CNS) inflammation by disrupting the blood-brain barrier (BBB) causing functional neuronal disruption that leads to PND. PND is strongly linked to elevations in serum and CNS pro-inflammatory cytokines interleukin-1 beta (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor alpha (TNFα); these cytokines cause further release of HMGB1 creating a positive feedback loop that amplifies the inflammatory response. The cytokine IL-6 is necessary and sufficient for PND. Dehydroepiandrosterone (DHEA) is a principal component of the steroid metabolome and is involved in immune homeostasis. DHEA has been shown to suppress expression of several pro-inflammatory cytokines by regulation of the NF-kB pathway. Bromo-epi-androsterone (BEA) is a potent synthetic analog of DHEA; unlike DHEA, it is non-androgenic, non-anabolic and is an effective modulator of immune dysregulation. In a randomized, placebo-controlled clinical trial, BEA effected significant and sustained decreases in IL-1β, TNFα and IL-6. This article presents BEA as a potential candidate for clinical trials targeting PND and further suggests the use of BEA in elective total hip arthroplasty as a well-documented surgical entity relevant to the management of PND.
Collapse
Affiliation(s)
- Coad Thomas Dow
- McPherson Eye Research Institute, University of Wisconsin-Madison, 9431 Wisconsin Institutes for, Medical Research (WIMR), 1111 Highland Avenue, Madison, WI 53705, United States of America.
| | - Zade Kidess
- Department of Chemistry and Biochemistry, Creighton University, 2500 California Plaza, Omaha, NE 68178, United States of America.
| |
Collapse
|
29
|
Donello JE, McIntyre RS, Pickel DB, Stahl SM. Demystifying the Antidepressant Mechanism of Action of Stinels, a Novel Class of Neuroplastogens: Positive Allosteric Modulators of the NMDA Receptor. Pharmaceuticals (Basel) 2025; 18:157. [PMID: 40005971 PMCID: PMC11858332 DOI: 10.3390/ph18020157] [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: 11/06/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Plastogens are a class of therapeutics that function by rapidly promoting changes in neuroplasticity. A notable example, ketamine, is receiving great attention due to its combined rapid and long-term antidepressant effects. Ketamine is an N-methyl-D-aspartate receptor (NMDAR) antagonist, and, in addition to its therapeutic activity, it is associated with psychotomimetic and dissociative side effects. Stinels-rapastinel, apimostinel, and zelquistinel-are also plastogens not only with rapid and long-term antidepressant effects but also with improved safety and tolerability profiles compared to ketamine. Previous descriptions of the mechanism by which stinels modulate NMDAR activity have been inconsistent and, at times, contradictory. The purpose of this review is to clarify the mechanism of action and contextualize stinels within a broader class of NMDAR-targeting therapeutics. In this review, we present the rationale behind targeting NMDARs for treatment-resistant depression and other psychiatric conditions, describe the various mechanisms by which NMDAR activity is regulated by different classes of therapeutics, and present evidence for the stinel mechanism. In contrast with previous descriptions of glycine-like NMDAR partial agonists, we define stinels as positive allosteric modulators of NMDAR activity with a novel regulatory binding site.
Collapse
Affiliation(s)
| | - Roger S. McIntyre
- Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada
| | | | - Stephen M. Stahl
- Department of Psychiatry and Neuroscience, University of California, Riverside, Riverside, CA 92521, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Department of Psychiatry, University of Cambridge, Cambridge CB2 2QQ, UK
- California Department of State Hospitals, Sacramento, CA 95814, USA
| |
Collapse
|
30
|
Chen Z, Coste GI, Li E, Huganir RL, Graves AR, Charles AS. Automatic detection of fluorescently labeled synapses in volumetric in vivo imaging data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634278. [PMID: 39896629 PMCID: PMC11785214 DOI: 10.1101/2025.01.22.634278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Synapses are submicron structures that connect and enable communication between neurons. Many forms of learning are thought to be encoded by synaptic plasticity, wherein the strength of specific synapses is regulated by modulating expression of neurotransmitter receptors. For instance, regulation of AMPA-type glutamate receptors is a central mechanism controlling the formation and recollection of long-term memories. A critical step in understanding how synaptic plasticity drives behavior is thus to directly observe, i.e., image, fluorescently labeled synapses in living tissue. However, due to their small size and incredible density - with one ~0.5 μm diameter synapse every cubic micron - accurately detecting individual synapses and segmenting each from its closely abutting neighbors is challenging. To overcome this, we trained a convolutional neural network to simultaneously detect and separate densely labeled synapses. These tools significantly increased the accuracy and scale of synapse detection, enabling segmentation of hundreds of thousands of individual synapses imaged in living mice.
Collapse
Affiliation(s)
- Zhining Chen
- Department of Biomedical Engineering, Johns Hopkins University
| | | | - Evan Li
- Department of Neuroscience, Johns Hopkins University
| | - Richard L. Huganir
- Department of Neuroscience, Johns Hopkins University
- Kavli Neurodiscovery Institute, Johns Hopkins University
| | - Austin R. Graves
- Department of Biomedical Engineering, Johns Hopkins University
- Kavli Neurodiscovery Institute, Johns Hopkins University
| | - Adam S. Charles
- Department of Biomedical Engineering, Johns Hopkins University
- Department of Neuroscience, Johns Hopkins University
- Kavli Neurodiscovery Institute, Johns Hopkins University
- Center for Imaging Science, Johns Hopkins University
- Mathematical Institute for Data Science, Johns Hopkins University
- Lead contact
| |
Collapse
|
31
|
Lepagnol-Bestel AM, Haziza S, Viard J, Salin PA, Duchon A, Herault Y, Simonneau M. DYRK1A Up-Regulation Specifically Impairs a Presynaptic Form of Long-Term Potentiation. Life (Basel) 2025; 15:149. [PMID: 40003558 PMCID: PMC11856406 DOI: 10.3390/life15020149] [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: 12/22/2024] [Revised: 01/12/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Chromosome 21 DYRK1A kinase is associated with a variety of neuronal diseases including Down syndrome. However, the functional impact of this kinase at the synapse level remains unclear. We studied a mouse model that incorporated YAC 152F7 (570 kb), encoding six chromosome 21 genes including DYRK1A. The 152F7 mice displayed learning difficulties but their N-methyl-D-aspartate (NMDA)-dependent synaptic long-term potentiation is indistinguishable from non-transgenic animals. We have demonstrated that a presynaptic form of NMDA-independent long-term potentiation (LTP) at the hippocampal mossy fiber was impaired in the 152F7 animals. To obtain insights into the molecular mechanisms involved in such synaptic changes, we analyzed the Dyrk1a interactions with chromatin remodelers. We found that the number of DYRK1A-EP300 and DYRK1A-CREBPP increased in 152F7 mice. Moreover, we observed a transcriptional decrease in genes encoding presynaptic proteins involved in glutamate vesicle exocytosis, namely Rims1, Munc13-1, Syn2 and Rab3A.To refine our findings, we used a mouse BAC 189N3 (152 kb) line that only triplicates the gene Dyrk1a. Again, we found that this NMDA-independent form of LTP is impaired in this mouse line. Altogether, our results demonstrate that Dyrk1a up-regulation is sufficient to specifically inhibit the NMDA-independent form of LTP and suggest that this inhibition is linked to chromatin changes that deregulate genes encoding proteins involved in glutamate synaptic release.
Collapse
Affiliation(s)
| | - Simon Haziza
- Centre Psychiatrie & Neurosciences, INSERM U894, 75014 Paris, France; (A.-M.L.-B.); (S.H.); (J.V.)
- Centre National de la Recherche Scientifique, Université Paris-Saclay, CentraleSupélec, École Normale Supérieure Paris-Saclay, LuMIn, 91190 Gif-sur-Yvette, France
| | - Julia Viard
- Centre Psychiatrie & Neurosciences, INSERM U894, 75014 Paris, France; (A.-M.L.-B.); (S.H.); (J.V.)
| | - Paul A. Salin
- Centre de Recherche en Neuroscience de Lyon CRNL (INSERM U1028), Université Claude-Bernard Lyon 1, 69100 Lyon, France;
| | - Arnaud Duchon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR7104, INSERM, U964, 67404 Illkirch, France; (A.D.); (Y.H.)
- Phenomin, Institut Clinique de la Souris (ICS), GIE CERBM, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR7104, INSERM, U964, 67404 Illkirch, France; (A.D.); (Y.H.)
- Phenomin, Institut Clinique de la Souris (ICS), GIE CERBM, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Michel Simonneau
- Centre Psychiatrie & Neurosciences, INSERM U894, 75014 Paris, France; (A.-M.L.-B.); (S.H.); (J.V.)
- Centre National de la Recherche Scientifique, Université Paris-Saclay, CentraleSupélec, École Normale Supérieure Paris-Saclay, LuMIn, 91190 Gif-sur-Yvette, France
- Département d’Enseignement et de Recherche en Biologie, École Normale Supérieure Paris-Saclay, 91190 Gif-sur-Yvette, France
| |
Collapse
|
32
|
Ly AT, Freites JA, Bertaccini GA, Evans EL, Dickinson GD, Tobias DJ, Pathak MM. Single-particle tracking reveals heterogeneous PIEZO1 diffusion. Biophys J 2025:S0006-3495(25)00023-2. [PMID: 39844465 DOI: 10.1016/j.bpj.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/21/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
The mechanically activated ion channel PIEZO1 is critical to numerous physiological processes, and is activated by diverse mechanical cues. The channel is gated by membrane tension and has been found to be mobile in the plasma membrane. We employed single-particle tracking (SPT) of endogenous, tdTomato-tagged PIEZO1 using total internal reflection fluorescence microscopy in live cells. Application of SPT unveiled a surprising heterogeneity of diffusing PIEZO1 subpopulations, which we labeled "mobile" and "immobile." We sorted these trajectories into the two aforementioned categories using trajectory spread. To evaluate the effects of the plasma membrane composition on PIEZO1 diffusion, we manipulated membrane composition by depleting or supplementing cholesterol, or by adding margaric acid to stiffen the membrane. To examine effects of channel activation on PIEZO1 mobility, we treated cells with Yoda1, a PIEZO1 agonist, and GsMTx-4, a channel inhibitor. We collected thousands of trajectories for each condition, and found that cholesterol removal and Yoda1 incubation increased the channel's propensity for mobility. Conversely, we found that GsMTx-4 incubation and cholesterol supplementation resulted in a lower chance of mobile trajectories, whereas margaric acid incubation did not have a significant effect on PIEZO1 mobility. The mobile trajectories were analyzed further by fitting the time-averaged mean-squared displacement as a function of lag time to a power law model, revealing that mobile PIEZO1 puncta exhibit anomalous subdiffusion. These studies illuminate the fundamental properties governing PIEZO1 diffusion in the plasma membrane and set the stage to determine how cellular processes and interactions may influence channel activity and mobility.
Collapse
Affiliation(s)
- Alan T Ly
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | | | - Gabriella A Bertaccini
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | - Elizabeth L Evans
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | - George D Dickinson
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | - Douglas J Tobias
- Department of Chemistry, UC Irvine, Irvine, California; Center for Complex Biological Systems, UC Irvine, Irvine, California.
| | - Medha M Pathak
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Department of Biomedical Engineering, UC Irvine, Irvine, California; Center for Complex Biological Systems, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California.
| |
Collapse
|
33
|
Micheva KD, Simhal AK, Schardt J, Smith SJ, Weinberg RJ, Owen SF. Data-driven synapse classification reveals a logic of glutamate receptor diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.11.628056. [PMID: 39713368 PMCID: PMC11661198 DOI: 10.1101/2024.12.11.628056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The rich diversity of synapses facilitates the capacity of neural circuits to transmit, process and store information. We used multiplex super-resolution proteometric imaging through array tomography to define features of single synapses in mouse neocortex. We find that glutamatergic synapses cluster into subclasses that parallel the distinct biochemical and functional categories of receptor subunits: GluA1/4, GluA2/3 and GluN1/GluN2B. Two of these subclasses align with physiological expectations based on synaptic plasticity: large AMPAR-rich synapses may represent potentiated synapses, whereas small NMDAR-rich synapses suggest "silent" synapses. The NMDA receptor content of large synapses correlates with spine neck diameter, and thus the potential for coupling to the parent dendrite. Overall, ultrastructural features predict receptor content of synapses better than parent neuron identity does, suggesting synapse subclasses act as fundamental elements of neuronal circuits. No barriers prevent future generalization of this approach to other species, or to study of human disorders and therapeutics.
Collapse
Affiliation(s)
- Kristina D. Micheva
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Anish K. Simhal
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Jenna Schardt
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Stephen J Smith
- Allen Institute for Brain Science, Seattle, WA 98109
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Richard J. Weinberg
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27514
| | - Scott F. Owen
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305
- Lead contact
| |
Collapse
|
34
|
Strucinska K, Kneis P, Pennington T, Cizio K, Szybowska P, Morgan A, Weertman J, Lewis TL. Fis1 is required for the development of the dendritic mitochondrial network in pyramidal cortical neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631801. [PMID: 39829888 PMCID: PMC11741399 DOI: 10.1101/2025.01.07.631801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Mitochondrial ATP production and calcium buffering are critical for metabolic regulation and neurotransmission making the formation and maintenance of the mitochondrial network a critical component of neuronal health. Cortical pyramidal neurons contain compartment-specific mitochondrial morphologies that result from distinct axonal and dendritic mitochondrial fission and fusion profiles. We previously showed that axonal mitochondria are maintained at a small size as a result of high axonal mitochondrial fission factor (Mff) activity. However, loss of Mff activity had little effect on cortical dendritic mitochondria, raising the question of how fission/fusion balance is controlled in the dendrites. Thus, we sought to investigate the role of another fission factor, fission 1 (Fis1), on mitochondrial morphology, dynamics and function in cortical neurons. We knocked down Fis1 in cortical neurons both in primary culture and in vivo, and unexpectedly found that Fis1 depletion decreased mitochondrial length in the dendrites, without affecting mitochondrial size in the axon. Further, loss of Fis1 activity resulted in both increased mitochondrial motility and dynamics in the dendrites. These results argue Fis1 exhibits dendrite selectivity and plays a more complex role in neuronal mitochondrial dynamics than previously reported. Functionally, Fis1 loss resulted in reduced mitochondrial membrane potential, increased sensitivity to complex III blockade, and decreased mitochondrial calcium uptake during neuronal activity. The altered mitochondrial network culminated in elevated resting calcium levels that increased dendritic branching but reduced spine density. We conclude that Fis1 regulates morphological and functional mitochondrial characteristics that influence dendritic tree arborization and connectivity.
Collapse
Affiliation(s)
- Klaudia Strucinska
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Parker Kneis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Molecular Biology & Biochemistry Department, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
| | - Travis Pennington
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Neuroscience Program, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
| | - Katarzyna Cizio
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Neuroscience Program, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
| | - Patrycja Szybowska
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Abigail Morgan
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Neuroscience Program, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
| | - Joshua Weertman
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Tommy L Lewis
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Molecular Biology & Biochemistry Department, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
- Neuroscience Program, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
- Physiology Department, Oklahoma University Health Sciences Campus, Oklahoma City, OK 73104
| |
Collapse
|
35
|
Rana S, Fusco AF, Witkin JM, Radin DP, Cerne R, Lippa A, Fuller DD. Pharmacological modulation of respiratory control: Ampakines as a therapeutic strategy. Pharmacol Ther 2025; 265:108744. [PMID: 39521442 PMCID: PMC11849399 DOI: 10.1016/j.pharmthera.2024.108744] [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: 08/04/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Ampakines are a class of compounds that are positive allosteric modulators of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors and enhance glutamatergic neurotransmission. Glutamatergic synaptic transmission and AMPA receptor activation are fundamentally important to the genesis and propagation of the neural impulses driving breathing, including respiratory motoneuron depolarization. Ampakines therefore have the potential to modulate the neural control of breathing. In this paper, we describe the influence of ampakines on respiratory motor output in health and disease. We dissect the molecular mechanisms underlying ampakine action, delineate the diverse targets of ampakines along the respiratory neuraxis, survey the spectrum of respiratory disorders in which ampakines have been tested, and culminate with an examination of how ampakines modulate respiratory function after spinal cord injury. Collectively, the studies reviewed here indicate that ampakines may be a useful adjunctive strategy to pair with conventional respiratory rehabilitation approaches in conditions with impaired neural activation of the respiratory muscles.
Collapse
Affiliation(s)
- Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, United States of America; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States of America; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States of America.
| | - Anna F Fusco
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, United States of America; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States of America; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States of America
| | - Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, St. Vincent's Hospital, Indianapolis, IN, United States of America; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN, United States of America; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America
| | - Daniel P Radin
- RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America
| | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, St. Vincent's Hospital, Indianapolis, IN, United States of America; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America; Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, Ljubljana, Slovenia
| | - Arnold Lippa
- RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, United States of America; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States of America; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States of America
| |
Collapse
|
36
|
Lømo T. Long-Term Potentiation: The Accidental Discovery. Hippocampus 2025; 35:e23664. [PMID: 39648690 PMCID: PMC11626224 DOI: 10.1002/hipo.23664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/01/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024]
Abstract
Long-term potentiation (LTP), is a type of synaptic plasticity now considered essential for learning and memory. Here I tell the story of how I accidentally discovered in 1966 in the laboratory of Per Andersen in Oslo, Norway, because I was not looking for it. It just emerged. I recount how I came to work with Per and why my result was not immediately followed up. Then, in 1968 Tim Bliss joined the lab and, on his urging, from 1968 to 1969 we did the experiments that resulted in Bliss and Lømo, 1973. I explain why I think the experiments later failed in Oslo, and for a few years also in Tim's lab in London, before it became a readily observable phenomenon. I also describe how Tony Gardner-Medwin and I in 1971 failed to reproduce the results that Tim and I had obtained 2 years earlier in the same lab and the same type of anesthetized rabbit preparation. I tell how this failure caused me to leave the LTP field and, instead, continue exploring mechanisms of nerve-muscle interactions, which I had studied with much success during my postdoc period in London from 1969 to 1971. I reflect on Donald Hebb's influence on LTP studies and on my experience when after many years of neglect, I became interested in LTP and the hippocampus anew and started to write about it, though without doing lab experiments. Finally, I report briefly on the experiments I am doing now in retirement, studying how the nervous system regulates body temperature through varying amounts of muscle tone.
Collapse
Affiliation(s)
- Terje Lømo
- Institute of Basic Medical Sciences, University of OsloOsloNorway
| |
Collapse
|
37
|
Svejgaard B, Modrau B, Hernández-Gloria JJ, Wested CL, Dosen S, Stevenson AJT, Mrachacz-Kersting N. Associative brain-computer interface training increases wrist extensor corticospinal excitability in patients with subacute stroke. J Neurophysiol 2025; 133:333-341. [PMID: 39704693 DOI: 10.1152/jn.00452.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/19/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
In a recently developed associative rehabilitative brain-computer interface (BCI) system, electroencephalography (EEG) is used to identify the most active phase of the motor cortex during attempted movement and deliver precisely timed peripheral stimulation during training. This approach has been demonstrated to facilitate corticospinal excitability and functional recovery in patients with lower limb weakness following stroke. The current study expands those findings by investigating changes in corticospinal excitability following the associative BCI intervention in patients with post stroke with upper limb weakness. In a randomized controlled trial, 24 patients with subacute stroke, subdivided into an intervention group and a "sham" control group, performed 30 wrist extensions. The intervention comprised 30 pairings of single peripheral nerve stimulation at the motor threshold, timed so that the generated afferent volley arrived at the motor cortex during the peak negativity of the movement-related cortical potential (MRCP), which was identified with EEG. The sham group underwent the same intervention, though the intensity of the nerve stimulation was below the perception threshold. Immediately after training, patients in the associative group exhibited significantly larger amplitudes of muscular-evoked potentials, compared with pretraining measurements in response to transcranial magnetic stimulation. These changes persisted for at least 30 min and were not observed in the sham group. We demonstrate that motor-evoked potential amplitudes increased significantly following paired associative BCI training targeting upper limb muscles in patients with subacute stroke, which is in line with results from lower limb studies.NEW & NOTEWORTHY We have demonstrated that a single training session with an associative brain-computer interface increased corticospinal excitability in patients suffering from upper limb weakness following stroke. This is the first time such an effect is described in the upper limb, which paves the way for effect augmentation of existing upper limb rehabilitation protocols.
Collapse
Affiliation(s)
- Benjamin Svejgaard
- Center for Neurotechnology and Rehabilitation, Aalborg University, Aalborg, Denmark
- Department of Neurology, Aalborg University Hospital, Aalborg, Denmark
- Neuroenhed Nord, North Denmark Regional Hospital, Brønderslev, Denmark
| | - Boris Modrau
- Center for Neurotechnology and Rehabilitation, Aalborg University, Aalborg, Denmark
- Department of Neurology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Carina L Wested
- Neuroenhed Nord, North Denmark Regional Hospital, Brønderslev, Denmark
| | - Strahinja Dosen
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Andrew J T Stevenson
- Center for Neurotechnology and Rehabilitation, Aalborg University, Aalborg, Denmark
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Natalie Mrachacz-Kersting
- Institute of Sport and Sport Science, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- BrainLinks-BrainTools Center, IMBIT, Albert-Ludwigs University of Freiburg, Freiburg, Germany
| |
Collapse
|
38
|
Ma Y, Kyuchukova D, Jiao F, Batsikadze G, Nitsche MA, Yavari F. The impact of temporal distribution on fear extinction learning. Int J Clin Health Psychol 2025; 25:100536. [PMID: 39877888 PMCID: PMC11770545 DOI: 10.1016/j.ijchp.2024.100536] [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: 09/12/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025] Open
Abstract
Fear extinction is the foundation of exposure therapy for anxiety and phobias. However, the stability of extinction memory diminishes over time, coinciding with fear recovery. To augment long-term extinction retention, the temporal distribution of extinction learning sessions is critical. This study investigated the effects of massed and spaced training (with short and long intervals) on extinction retention compared to a classic protocol. 120 healthy participants were recruited and randomly divided to massed training, spaced training with 20-minutes or 3-hours intervals, and a control group. The control group completed half the number of extinction trials compared to the other groups. The fear conditioning/extinction paradigm consisted of three consecutive days of fear acquisition, extinction, and recall, followed by a second recall one week later. Skin conductance response (SCR) and self-rating questionnaires (ratings of valence, arousal, and fear) were recorded and analyzed using mixed model ANOVAs. The results revealed that during the extinction phase, both massed and spaced protocols showed significantly lower SCRs compared to the control group, with massed training resulting in the largest effects. In the second recall, only the massed extinction group showed no significant difference in SCRs between threat and safety cues. The self-report assessments indicated that the massed extinction group showed furthermore lower arousal than the control group in the first recall. These results suggest that both massed and spaced training promote fear extinction learning, but only massed training improves long-term extinction retention. This study highlights the impact of the temporal distribution and trial number of extinction learning on extinction retention, offering insights for future research on improving fear extinction efficacy.
Collapse
Affiliation(s)
- Yuanbo Ma
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- Department of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Dzheylyan Kyuchukova
- Department of Psychotherapy and Systems Neuroscience, Justus Liebig University of Giessen, Germany
| | - Fujia Jiao
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Giorgi Batsikadze
- Department of Neurology and Center for Translational Neuro and Behavioral Sciences (C-TNBS), Essen University Hospital, University of Duisburg-Essen, Hufelandstraße 55, Essen 45147, Germany
| | - Michael A. Nitsche
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- Bielefeld University, University Hospital OWL, Protestant Hospital of Bethel Foundation, University Clinic of Psychiatry and Psychotherapy, Bielefeld, Germany
- German Center for Mental Health (DZPG), Bochum, Germany
| | - Fatemeh Yavari
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| |
Collapse
|
39
|
Barrantes FJ. Nicotinic acetylcholine receptors in the brain. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:37-54. [PMID: 40340066 DOI: 10.1016/b978-0-443-19088-9.00004-4] [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: 05/10/2025]
Abstract
The nicotinic acetylcholine receptor (nAChR) is the archetypal neurotransmitter receptor within the superfamily of pentameric ligand-gated ion channels (pLGICs). Typically, it mediates fast synaptic transmission in response to its endogenous ligand, acetylcholine, and can also intervene in slower signaling mechanisms via intracellular metabolic cascades in association with G-protein-coupled receptors. This review covers the structural and functional aspects of the different neuronal nAChR subtypes and their cellular and anatomic distribution in the brain. The significant progress in our knowledge on the topic derives from the successful combination of biochemical, neuroanatomic, pharmacologic, and cell biology approaches, complemented by site-directed mutagenesis, single-channel electrophysiology, and structural biophysical studies. This multipronged approach provides a comprehensive description of nAChR in health and disease, offering improved chances of success in tackling neurologic and neuropsychiatric diseases involving phenotypic alterations of nAChRs, particularly in neurodegenerative diseases.
Collapse
Affiliation(s)
- Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Biomedical Research Institute, Faculty of Medical Sciences, Pontifical Catholic University of Argentina (UCA), Buenos Aires, Argentina; National Scientific & Technological Research Council (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
40
|
Leana-Sandoval G, Kolli AV, Sandoval MA, Saavedra E, Li KH, Chen LY, Burlingame AL, Ramírez-Franco J, Díaz-Alonso J. The VGCC auxiliary subunit α2δ1 is an extracellular GluA1 interactor and regulates LTP, spatial memory, and seizure susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626379. [PMID: 39677598 PMCID: PMC11642997 DOI: 10.1101/2024.12.02.626379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Activity-dependent synaptic accumulation of AMPA receptors (AMPARs) and subsequent long-term synaptic strengthening underlie different forms of learning and memory. The AMPAR subunit GluA1 amino-terminal domain is essential for synaptic docking of AMPAR during LTP, but the precise mechanisms involved are not fully understood. Using unbiased proteomics, we identified the epilepsy and intellectual disability-associated VGCC auxiliary subunit α2δ1 as a candidate extracellular AMPAR slot. Presynaptic α2δ1 deletion in CA3 affects synaptic AMPAR incorporation during long-term potentiation, but not basal synaptic transmission, at CA1 synapses. Consistently, mice lacking α2δ1 in CA3 display a specific impairment in CA1-dependent spatial memory, but not in memory tests involving other cortical regions. Decreased seizure susceptibility in mice lacking α2δ1 in CA3 suggests a regulation of circuit excitability by α2δ1/AMPAR interactions. Our study sheds light on the regulation of activity-dependent AMPAR trafficking, and highlights the synaptic organizing roles of α2δ1.
Collapse
Affiliation(s)
- Gerardo Leana-Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Ananth V. Kolli
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Matthew A. Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Emily Saavedra
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Kathy H. Li
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lulu Y. Chen
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Alma L. Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jorge Ramírez-Franco
- Institut de Neurosciences de la Timone, Aix-Marseille Université (AMU) & CNRS, UMR7289, 13005, Marseille, France
| | - Javier Díaz-Alonso
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| |
Collapse
|
41
|
Christopoulou E, Charrier C. Molecular mechanisms of the specialization of human synapses in the neocortex. Curr Opin Genet Dev 2024; 89:102258. [PMID: 39255688 DOI: 10.1016/j.gde.2024.102258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/05/2024] [Accepted: 08/18/2024] [Indexed: 09/12/2024]
Abstract
Synapses of the neocortex specialized during human evolution to develop over extended timescales, process vast amounts of information and increase connectivity, which is thought to underlie our advanced social and cognitive abilities. These features reflect species-specific regulations of neuron and synapse cell biology. However, despite growing understanding of the human genome and the brain transcriptome at the single-cell level, linking human-specific genetic changes to the specialization of human synapses has remained experimentally challenging. In this review, we describe recent progress in characterizing divergent morphofunctional and developmental properties of human synapses, and we discuss new insights into the underlying molecular mechanisms. We also highlight intersections between evolutionary innovations and disorder-related dysfunctions at the synapse.
Collapse
Affiliation(s)
- Eirini Christopoulou
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Cécile Charrier
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France.
| |
Collapse
|
42
|
Leana-Sandoval G, Kolli AV, Chinn CA, Madrid A, Lo I, Sandoval MA, Vera VA, Simms J, Wood MA, Diaz-Alonso J. The GluA1 cytoplasmic tail regulates intracellular AMPA receptor trafficking and synaptic transmission onto dentate gyrus GABAergic interneurons, gating response to novelty. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626277. [PMID: 39677714 PMCID: PMC11643017 DOI: 10.1101/2024.12.01.626277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The GluA1 subunit, encoded by the putative schizophrenia-associated gene GRIA1, is required for activity-regulated AMPA receptor (AMPAR) trafficking, and plays a key role in cognitive and affective function. The cytoplasmic, carboxy-terminal domain (CTD) is the most divergent region across AMPAR subunits. The GluA1 CTD has received considerable attention for its role during long-term potentiation (LTP) at CA1 pyramidal neuron synapses. However, its function at other synapses and, more broadly, its contribution to different GluA1-dependent processes, is poorly understood. Here, we used mice with a constitutive truncation of the GluA1 CTD to dissect its role regulating AMPAR localization and function as well as its contribution to cognitive and affective processes. We found that GluA1 CTD truncation affected AMPAR subunit levels and intracellular trafficking. ΔCTD GluA1 mice exhibited no memory deficits, but presented exacerbated novelty-induced hyperlocomotion and dentate gyrus granule cell (DG GC) hyperactivity, among other behavioral alterations. Mechanistically, we found that AMPAR EPSCs onto DG GABAergic interneurons were significantly reduced, presumably underlying, at least in part, the observed changes in neuronal activity and behavior. In summary, this study dissociates CTD-dependent from CTD-independent GluA1 functions, unveiling the GluA1 CTD as a crucial hub regulating AMPAR function in a cell type-specific manner.
Collapse
Affiliation(s)
- Gerardo Leana-Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Ananth V Kolli
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Carlene A Chinn
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
- Department of Neurobiology & Behavior, University of California at Irvine, CA, 92697, USA
| | - Alexis Madrid
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Iris Lo
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Matthew A Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Vanessa Alizo Vera
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
- Department of Neurobiology & Behavior, University of California at Irvine, CA, 92697, USA
| | - Jeffrey Simms
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Marcelo A Wood
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
- Department of Neurobiology & Behavior, University of California at Irvine, CA, 92697, USA
| | - Javier Diaz-Alonso
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| |
Collapse
|
43
|
Pan G, Chai L, Chen R, Yuan Q, Song Z, Feng W, Wei J, Yang Z, Zhang Y, Xie G, Yan A, Lv Q, Wang C, Zhao Y, Wang Y. Potential mechanism of Qinggong Shoutao pill alleviating age-associated memory decline based on integration strategy. PHARMACEUTICAL BIOLOGY 2024; 62:105-119. [PMID: 38145345 PMCID: PMC10763866 DOI: 10.1080/13880209.2023.2291689] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 11/30/2023] [Indexed: 12/26/2023]
Abstract
CONTEXT Qinggong Shoutao Wan (QGSTW) is a pill used as a traditional medicine to treat age-associated memory decline (AAMI). However, its potential mechanisms are unclear. OBJECTIVE This study elucidates the possible mechanisms of QGSTW in treating AAMI. MATERIALS AND METHODS Network pharmacology and molecular docking approaches were utilized to identify the potential pathway by which QGSTW alleviates AAMI. C57BL/6J mice were divided randomly into control, model, and QGSTW groups. A mouse model of AAMI was established by d-galactose, and the pathways that QGSTW acts on to ameliorate AAMI were determined by ELISA, immunofluorescence staining and Western blotting after treatment with d-gal (100 mg/kg) and QGSTW (20 mL/kg) for 12 weeks. RESULTS Network pharmacology demonstrated that the targets of the active components were significantly enriched in the cAMP signaling pathway. AKT1, FOS, GRIN2B, and GRIN1 were the core target proteins. QGSTW treatment increased the discrimination index from -16.92 ± 7.06 to 23.88 ± 15.94% in the novel location test and from -19.54 ± 5.71 to 17.55 ± 6.73% in the novel object recognition test. ELISA showed that QGSTW could increase the levels of cAMP. Western blot analysis revealed that QGSTW could upregulate the expression of PKA, CREB, c-Fos, GluN1, GluA1, CaMKII-α, and SYN. Immunostaining revealed that the expression of SYN was decreased in the CA1 and DG. DISCUSSION AND CONCLUSIONS This study not only provides new insights into the mechanism of QGSTW in the treatment of AAMI but also provides important information and new research ideas for the discovery of traditional Chinese medicine compounds that can treat AAMI.
Collapse
Affiliation(s)
- Guiyun Pan
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lijuan Chai
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing Yuan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Song
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wanying Feng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinna Wei
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihua Yang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhang Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guinan Xie
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - An Yan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qingbo Lv
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Caijun Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingqiang Zhao
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
44
|
Comai S, De Martin S, Mattarei A, Guidetti C, Pappagallo M, Folli F, Alimonti A, Manfredi PL. N-methyl-D-aspartate Receptors and Depression: Linking Psychopharmacology, Pathology and Physiology in a Unifying Hypothesis for the Epigenetic Code of Neural Plasticity. Pharmaceuticals (Basel) 2024; 17:1618. [PMID: 39770460 PMCID: PMC11728621 DOI: 10.3390/ph17121618] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/03/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Uncompetitive NMDAR (N-methyl-D-aspartate receptor) antagonists restore impaired neural plasticity, reverse depressive-like behavior in animal models, and relieve major depressive disorder (MDD) in humans. This review integrates recent findings from in silico, in vitro, in vivo, and human studies of uncompetitive NMDAR antagonists into the extensive body of knowledge on NMDARs and neural plasticity. Uncompetitive NMDAR antagonists are activity-dependent channel blockers that preferentially target hyperactive GluN2D subtypes because these subtypes are most sensitive to activation by low concentrations of extracellular glutamate and are more likely activated by certain pathological agonists and allosteric modulators. Hyperactivity of GluN2D subtypes in specific neural circuits may underlie the pathophysiology of MDD. We hypothesize that neural plasticity is epigenetically regulated by precise Ca2+ quanta entering cells via NMDARs. Stimuli reach receptor cells (specialized cells that detect specific types of stimuli and convert them into electrical signals) and change their membrane potential, regulating glutamate release in the synaptic cleft. Free glutamate binds ionotropic glutamatergic receptors regulating NMDAR-mediated Ca2+ influx. Quanta of Ca2+ via NMDARs activate enzymatic pathways, epigenetically regulating synaptic protein homeostasis and synaptic receptor expression; thereby, Ca2+ quanta via NMDARs control the balance between long-term potentiation and long-term depression. This NMDAR Ca2+ quantal hypothesis for the epigenetic code of neural plasticity integrates recent psychopharmacology findings into established physiological and pathological mechanisms of brain function.
Collapse
Affiliation(s)
- Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35121 Padua, Italy; (S.C.); (S.D.M.); (A.M.)
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
- Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada
- IRCSS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35121 Padua, Italy; (S.C.); (S.D.M.); (A.M.)
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35121 Padua, Italy; (S.C.); (S.D.M.); (A.M.)
| | - Clotilde Guidetti
- Child Neuropsychiatry Unit, Department of Neuroscience, IRCCS Bambino Gesù Pediatric Hospital, 00165 Rome, Italy;
- Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Marco Pappagallo
- Relmada Therapeutics, Inc., Coral Gables, FL 33134, USA;
- MGGM LLC, 85 Baker Road, Kerhonkson, NY 12446, USA
| | - Franco Folli
- Department of Health Sciences, University of Milan, 20141 Milan, Italy;
| | - Andrea Alimonti
- The Institute of Oncology Research, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland;
- Veneto Institute of Molecular Medicine, 35129 Padua, Italy
- Department of Medicine, Zurich University, 8006 Zurich, Switzerland
- Department of Medicine, University of Padua, 35122 Padua, Italy
| | - Paolo L. Manfredi
- Relmada Therapeutics, Inc., Coral Gables, FL 33134, USA;
- MGGM LLC, 85 Baker Road, Kerhonkson, NY 12446, USA
| |
Collapse
|
45
|
Benarroch E. What Is the Role of Cytokines in Synaptic Transmission? Neurology 2024; 103:e209928. [PMID: 39303183 DOI: 10.1212/wnl.0000000000209928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
|
46
|
Caneus J, Autar K, Akanda N, Grillo M, Long CJ, Jackson M, Lindquist S, Guo X, Morgan D, Hickman JJ. Validation of a functional human AD model with four AD therapeutics utilizing patterned ipsc-derived cortical neurons integrated with microelectrode arrays. Sci Rep 2024; 14:24875. [PMID: 39438515 PMCID: PMC11496884 DOI: 10.1038/s41598-024-73869-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Preclinical methods are needed for screening potential Alzheimer's disease (AD) therapeutics that recapitulate phenotypes found in the Mild Cognitive Impairment (MCI) stage or even before this stage of the disease. This would require a phenotypic system that reproduces cognitive deficits without significant neuronal cell death to mimic the clinical manifestations of AD during these stages. Long-term potentiation (LTP), which is a correlate of learning and memory, was induced in mature human iPSC-derived cortical neurons cultured on microelectrode arrays utilizing circuit patterns connecting two adjacent electrodes. We demonstrated an LTP system that modeled the MCI and pre-MCI stages of Alzheimer's and validated this functional system utilizing four AD therapeutics, which was also verified utilizing patch-clamp electrophysiology. LTP was induced by tetanic electrical stimulation, and LTP maintenance was significantly reduced in the presence of Amyloid-Beta 42 (Aβ42) oligomers compared to the controls, however, co-treatment with AD therapeutics (Donepezil, Memantine, Rolipram and Saracatinib) corrected Aβ42-induced LTP impairment. The results illustrate the utility of the system as a validated platform to model MCI AD pathology, and potentially for the pre-MCI phase before significant neuronal death. This system also has the potential to become an ideal platform for high-content therapeutic screening for other neurodegenerative diseases.
Collapse
Affiliation(s)
- Julbert Caneus
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA.
| | - Kaveena Autar
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Nesar Akanda
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Marcella Grillo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | | | - Max Jackson
- Hesperos Inc., 12501 Research Pkwy #100, Orlando, FL, USA
| | | | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Dave Morgan
- Department of Translational Neuroscience, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
- Hesperos Inc., 12501 Research Pkwy #100, Orlando, FL, USA
| |
Collapse
|
47
|
Tao-Cheng JH, Moreira SL, Winters CA. Ultrastructural characterization of hippocampal inhibitory synapses under resting and stimulated conditions. Mol Brain 2024; 17:76. [PMID: 39438991 PMCID: PMC11494804 DOI: 10.1186/s13041-024-01151-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024] Open
Abstract
The present study uses electron microscopy to document ultrastructural characteristics of hippocampal GABAergic inhibitory synapses under resting and stimulated conditions in three experimental systems. Synaptic profiles were sampled from stratum pyramidale and radiatum of the CA1 region from (1) perfusion fixed mouse brains, (2) immersion fixed rat organotypic slice cultures, and from (3) rat dissociated hippocampal cultures of mixed cell types. Synapses were stimulated in the brain by a 5 min delay in perfusion fixation to trigger an ischemia-like excitatory condition, and by treating the two culture systems with 90 mM high K+ for 2-3 min to depolarize the neurons. Upon such stimulation conditions, the presynaptic terminals of the inhibitory synapses exhibited similar structural changes to those seen in glutamatergic excitatory synapses, with depletion of synaptic vesicles, increase of clathrin-coated vesicles and appearance of synaptic spinules. However, in contrast to excitatory synapses, no structural differences were detected in the postsynaptic compartment of the inhibitory synapses upon stimulation. There were no changes in the appearance of material associated with the postsynaptic membrane or the length and curvature of the membrane. Also no change was detected in the labeling density of gephyrin, a GABAergic synaptic marker, lining the postsynaptic membrane. Furthermore, virtually all inhibitory synaptic clefts remained rigidly apposed, unlike in the case of excitatory synapses where ~ 20-30% of cleft edges were open upon stimulation, presumably to facilitate the clearance of neurotransmitters from the cleft. The fact that no open clefts were induced in inhibitory synapses upon stimulation suggests that inhibitory input may not need to be toned down under these conditions. On the other hand, similar to excitatory synapse, EGTA (a calcium chelator) induced open clefts in ~ 18% of inhibitory synaptic cleft edges, presumably disrupting similar calcium-dependent trans-synaptic bridges in both types of synapses.
Collapse
Affiliation(s)
- Jung-Hwa Tao-Cheng
- NINDS Electron Microscopy Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Sandra Lara Moreira
- NINDS Electron Microscopy Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Christine A Winters
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
48
|
Barrantes FJ. Cognitive synaptopathy: synaptic and dendritic spine dysfunction in age-related cognitive disorders. Front Aging Neurosci 2024; 16:1476909. [PMID: 39420927 PMCID: PMC11484076 DOI: 10.3389/fnagi.2024.1476909] [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: 08/06/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Cognitive impairment is a leading component of several neurodegenerative and neurodevelopmental diseases, profoundly impacting on the individual, the family, and society at large. Cognitive pathologies are driven by a multiplicity of factors, from genetic mutations and genetic risk factors, neurotransmitter-associated dysfunction, abnormal connectomics at the level of local neuronal circuits and broader brain networks, to environmental influences able to modulate some of the endogenous factors. Otherwise healthy older adults can be expected to experience some degree of mild cognitive impairment, some of which fall into the category of subjective cognitive deficits in clinical practice, while many neurodevelopmental and neurodegenerative diseases course with more profound alterations of cognition, particularly within the spectrum of the dementias. Our knowledge of the underlying neuropathological mechanisms at the root of this ample palette of clinical entities is far from complete. This review looks at current knowledge on synaptic modifications in the context of cognitive function along healthy ageing and cognitive dysfunction in disease, providing insight into differential diagnostic elements in the wide range of synapse alterations, from those associated with the mild cognitive changes of physiological senescence to the more profound abnormalities occurring at advanced clinical stages of dementia. I propose the term "cognitive synaptopathy" to encompass the wide spectrum of synaptic pathologies associated with higher brain function disorders.
Collapse
Affiliation(s)
- Francisco J. Barrantes
- Laboratory of Molecular Neurobiology, Biomedical Research Institute, Pontifical Catholic University of Argentina (UCA), Argentine Scientific and Technological Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
49
|
Zsarnovszky A, Alymbaeva D, Jocsak G, Szabo C, Mária Schilling-Tóth B, Sandor Kiss D. Endocrine disrupting effects on morphological synaptic plasticity. Front Neuroendocrinol 2024; 75:101157. [PMID: 39393417 DOI: 10.1016/j.yfrne.2024.101157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/13/2024]
Abstract
Neural regulation of the homeostasis depends on healthy synaptic function. Adaptation of synaptic functions to physiological needs manifests in various forms of synaptic plasticity (SP), regulated by the normal hormonal regulatory circuits. During the past several decades, the hormonal regulation of animal and human organisms have become targets of thousands of chemicals that have the potential to act as agonists or antagonists of the endogenous hormones. As the action mechanism of these endocrine disrupting chemicals (EDCs) came into the focus of research, a growing number of studies suggest that one of the regulatory avenues of hormones, the morphological form of SP, may well be a neural mechanism affected by EDCs. The present review discusses known and potential effects of some of the best known EDCs on morphological synaptic plasticity (MSP). We highlight molecular mechanisms altered by EDCs and indicate the growing need for more research in this area of neuroendocrinology.
Collapse
Affiliation(s)
- Attila Zsarnovszky
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary; Department of Physiology and Animal Health, Agrobiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, H-7400 Kaposvár, Hungary.
| | - Daiana Alymbaeva
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary.
| | - Gergely Jocsak
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary.
| | - Csaba Szabo
- Department of Physiology and Animal Health, Agrobiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Nutrition, Hungarian University of Agriculture and Life Sciences, H-7400 Kaposvár, Hungary
| | | | - David Sandor Kiss
- Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, Hungary.
| |
Collapse
|
50
|
Shouval HZ, Flores-Obando RE, Sacktor TC. Maintenance of synaptic plasticity by negative-feedback of synaptic protein elimination: Dynamic modeling of KIBRA- PKM ζ interactions in LTP and memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614943. [PMID: 39386672 PMCID: PMC11463625 DOI: 10.1101/2024.09.25.614943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Activity-dependent modifications of synaptic efficacies are a cellular substrate of learning and memory. Current theories propose that the long-term maintenance of synaptic efficacies and memory is accomplished via a positive-feedback loop at the level of production of a protein species or a protein state. Here we propose a qualitatively different theoretical framework based on negative-feedback at the level of protein elimination. This theory is motivated by recent experimental findings regarding the binding of P K M ζ and KIBRA, two synaptic proteins involved in maintenance of memory, and on how this binding affects the proteins' degradation. We demonstrate this theoretical framework with two different models, a simple abstract model to explore generic features of such a process, and an experimentally motivated phenomenological model. The results of these models are qualitatively consistent with existing data, and generate novel predictions that could be experimentally tested to further validate or reject the negative-feedback theory.
Collapse
Affiliation(s)
- Harel Z. Shouval
- Department of Neurobiology and Anatomy, University of Texas Medical School, Houston, TX 77030, USA
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| | - Rafael E. Flores-Obando
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY. 11203. USA
| | - Todd C. Sacktor
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY. 11203. USA
- Department of Physiology, Pharmacology, Anesthesiology, and Neurology, SUNY Downstate Health Sciences University, Brooklyn, NY. 11203. USA
| |
Collapse
|