1
|
Shadman J, Haghi-Aminjan H, Alipour MR, Panahpour H. The Neuroprotective Mechanisms of Kaempferol in Experimental Ischemic Stroke: A Preclinical Systematic Review. Mol Neurobiol 2025:10.1007/s12035-025-04848-y. [PMID: 40120044 DOI: 10.1007/s12035-025-04848-y] [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: 07/23/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Ischemic stroke represents a critical global health challenge, resulting in significant mortality and disability worldwide, yet there are limited effective treatment options currently available. While the intricate molecular pathways underlying the onset and progression of ischemic stroke are multifaceted, relying on a single therapeutic approach is unlikely to yield effective treatment for this complex disease. Therefore, it is crucial to explore efficient strategies that employ multifaceted targeting and address the multifarious pathological processes to overcome the challenges associated with ischemic brain injury. In recent times, natural plant-derived compounds have garnered significant interest as promising neuroprotective agents for the management of neurological conditions, including ischemic stroke. This study investigates the possible neuroprotective properties of kaempferol, a naturally occurring flavonoid compound, in mitigating the detrimental consequences of cerebral ischemic events. The findings from the reviewed preclinical studies suggest that kaempferol exhibits significant neuroprotective potential as a multifaceted therapeutic agent for ischemic stroke. Its efficacy stems from a combination of antioxidant, anti-inflammatory, and anti-apoptotic properties, which collectively mitigate ischemic stroke-induced brain injury. While these results are promising, clinical studies are essential to validate kaempferol's therapeutic viability for ischemic stroke patients.
Collapse
Affiliation(s)
- Javad Shadman
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamed Haghi-Aminjan
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | | | - Hamdollah Panahpour
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
2
|
Liu X, Huang Y, Mu L, Friedman V, Kelly TJ, Hu Y, Yuan D, Liu QS. Epac2-mediated synaptic insertion of Ca 2+-permeable AMPARs in the nucleus accumbens contributes to incubation of cocaine craving. Neuropsychopharmacology 2025; 50:620-629. [PMID: 39702576 PMCID: PMC11845495 DOI: 10.1038/s41386-024-02030-x] [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: 09/03/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024]
Abstract
The accumulation of GluA2-lacking Ca2+-permeable AMPARs (CP-AMPARs) in the medium spiny neurons (MSNs) of the nucleus accumbens (NAc) is required for the expression of incubation of cocaine craving. The exchange protein directly activated by cAMP (Epac) is an intracellular effector of cAMP and a guanine nucleotide exchange factor for the small GTPase Rap1. Epac2 has been implicated in the trafficking of AMPA receptors at central synapses. We tested the hypothesis that Epac2 activation contributes to the accumulation of CP-AMPARs in NAc MSNs and incubation of cocaine craving. Here we demonstrate that the selective Epac2 agonist S-220 facilitated the synaptic insertion of GluA2-lacking CP-AMPARs at excitatory synapses onto NAc MSNs. In addition, prolonged abstinence from cocaine self-administration in rats resulted in elevated Rap1-GTP levels in the NAc, implying that Epac2 is activated during incubation. Importantly, we show that AAV-mediated shRNA knockdown of Epac2 in the NAc core attenuated the accumulation of CP-AMPARs and cue-induced drug-seeking behavior after prolonged abstinence from cocaine self-administration. In contrast, acute pharmacological inhibition of Epac2 with the selective Epac2 inhibitor ESI-05 did not alter CP-AMPARs that had already accumulated during incubation, and intra-NAc application of ESI-05 did not significantly affect cue-induced drug seeking following prolonged abstinence. Taken together, these results suggest that Epac2 activation during the period of incubation, but not during cue-induced drug seeking, leads to the accumulation of CP-AMPARs in NAc MSNs, which in turn contributes to incubation of cocaine craving.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Yao Huang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lianwei Mu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Vladislav Friedman
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Thomas J Kelly
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ying Hu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Dong Yuan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
3
|
Luppi AI, Liu ZQ, Hansen JY, Cofre R, Niu M, Kuzmin E, Froudist-Walsh S, Palomero-Gallagher N, Misic B. Benchmarking macaque brain gene expression for horizontal and vertical translation. SCIENCE ADVANCES 2025; 11:eads6967. [PMID: 40020056 PMCID: PMC11870082 DOI: 10.1126/sciadv.ads6967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 01/27/2025] [Indexed: 03/03/2025]
Abstract
The spatial patterning of gene expression shapes cortical organization and function. The macaque is a fundamental model organism in neuroscience, but the translational potential of macaque gene expression rests on the assumption that it is a good proxy for patterns of corresponding proteins (vertical translation) and for patterns of orthologous human genes (horizontal translation). Here, we systematically benchmark regional gene expression in macaque cortex against (i) macaque cortical receptor density and in vivo and ex vivo microstructure and (ii) human cortical gene expression. We find moderate cortex-wide correspondence between macaque gene expression and protein density, which improves by considering layer-specific gene expression. Half of the examined genes exhibit significant correlation between macaque and human across the cortex. Interspecies correspondence of gene expression is greater in unimodal than in transmodal cortex, recapitulating evolutionary cortical expansion and gene-protein correspondence in the macaque. These results showcase the potential and limitations of macaque cortical transcriptomics for translational discovery within and across species.
Collapse
Affiliation(s)
- Andrea I. Luppi
- Montréal Neurological Institute, McGill University, Montréal, QC, Canada
- Department of Psychiatry, University of Oxford, Oxford, UK
- St John’s College, University of Cambridge, Cambridge, UK
| | - Zhen-Qi Liu
- Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Justine Y. Hansen
- Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Rodrigo Cofre
- Paris-Saclay University, CNRS, Paris-Saclay Institute for Neuroscience (NeuroPSI), Saclay, France
| | - Meiqi Niu
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Elena Kuzmin
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, Canada
- Department of Human Genetics, Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | | | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- C. and O. Vogt Institute for Brain Research, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Bratislav Misic
- Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| |
Collapse
|
4
|
Zhao L, Witter MP, Palomero-Gallagher N. Cyto-, gene, and multireceptor architecture of the early postnatal mouse hippocampal complex. Prog Neurobiol 2025; 245:102704. [PMID: 39709019 DOI: 10.1016/j.pneurobio.2024.102704] [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: 10/13/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Neurotransmitter receptors are key molecules in signal transmission in the adult brain, and their precise spatial and temporal balance expressions also play a critical role in normal brain development. However, the specific balance expression of multiple receptors during hippocampal development is not well characterized. In this study, we used quantitative in vivo receptor autoradiography to measure the distributions and densities of 18 neurotransmitter receptor types in the mouse hippocampal complex at postnatal day 7, and compared them with the expressions of their corresponding encoding genes. We provide a novel and comprehensive characterization of the cyto-, gene, and multireceptor architecture of the developing mouse hippocampal and subicular regions during the developmental period, which typically differs from that in the adult brain. High-density receptor expressions with distinct regional and laminar distributions were observed for AMPA, Kainate, mGluR2/3, GABAA, GABAA/BZ, α2, and A1 receptors during this specific period, whereas NMDA, GABAB, α1, M1, M2, M3, nicotinic α4β2, 5-HT1A, 5-HT2, D1 and D2/D3 receptors exhibited relatively low and homogeneous expressions. This specific balance of multiple receptors aligns with regional cytoarchitecture, neurotransmitter distributions, and gene expressions. Moreover, contrasting with previous findings, we detected a high α2 receptor density, with distinct regional and laminar distribution patterns. A non-covariation differentiation phenomenon between α2 receptor distributions and corresponding gene expressions is also demonstrated in this early developmental period. The multimodal data provides new insights into understanding the hippocampal development from the perspective of cell, gene, and multireceptor levels, and contributes important resources for further interdisciplinary analyses.
Collapse
Affiliation(s)
- Ling Zhao
- Department of Psychology, School of Public Policy and Management, Nanchang University, Nanchang 330000, China; Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich 52425, Germany.
| | - Menno P Witter
- Kavli Institute for Systems Neuroscience, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich 52425, Germany; C. & O. Vogt Institute for Brain Research, Heinrich-Heine-University, Dusseldorf 40225, Germany
| |
Collapse
|
5
|
Hoisington ZW, Gangal H, Phamluong K, Shukla C, Ehinger Y, Moffat JJ, Homanics GE, Wang J, Ron D. Prosapip1 in the dorsal hippocampus mediates synaptic protein composition, long-term potentiation, and spatial memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.13.597459. [PMID: 38915579 PMCID: PMC11195216 DOI: 10.1101/2024.06.13.597459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Prosapip1 is a brain-specific protein localized to the postsynaptic density, where it promotes dendritic spine maturation in primary hippocampal neurons. However, nothing is known about the role of Prosapip1 in vivo. To examine this, we utilized the Cre-loxP system to develop a Prosapip1 neuronal knockout mouse. We found that Prosapip1 controls the synaptic localization of its binding partner SPAR, along with PSD-95 and the GluN2B subunit of the NMDA receptor (NMDAR) in the dorsal hippocampus (dHP). We next sought to identify the potential contribution of Prosapip1 to the activity and function of the NMDAR and found that Prosapip1 plays an important role in NMDAR-mediated transmission and long-term potentiation (LTP) in the CA1 region of the dHP. As LTP is the cellular hallmark of learning and memory, we examined the consequences of neuronal knockout of Prosapip1 on dHP-dependent memory. We found that global or dHP-specific neuronal knockout of Prosapip1 caused a deficit in learning and memory whereas developmental, locomotor, and anxiety phenotypes were normal. Taken together, Prosapip1 in the dHP promotes the proper localization of synaptic proteins which, in turn, facilitates LTP driving recognition, social, and spatial learning and memory.
Collapse
|
6
|
Saccenti D, Lauro LJR, Crespi SA, Moro AS, Vergallito A, Grgič RG, Pretti N, Lamanna J, Ferro M. Boosting Psychotherapy With Noninvasive Brain Stimulation: The Whys and Wherefores of Modulating Neural Plasticity to Promote Therapeutic Change. Neural Plast 2024; 2024:7853199. [PMID: 39723244 PMCID: PMC11669434 DOI: 10.1155/np/7853199] [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: 05/03/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024] Open
Abstract
The phenomenon of neural plasticity pertains to the intrinsic capacity of neurons to undergo structural and functional reconfiguration through learning and experiential interaction with the environment. These changes could manifest themselves not only as a consequence of various life experiences but also following therapeutic interventions, including the application of noninvasive brain stimulation (NIBS) and psychotherapy. As standalone therapies, both NIBS and psychotherapy have demonstrated their efficacy in the amelioration of psychiatric disorders' symptoms, with a certain variability in terms of effect sizes and duration. Consequently, scholars suggested the convenience of integrating the two interventions into a multimodal treatment to boost and prolong the therapeutic outcomes. Such an approach is still in its infancy, and the physiological underpinnings substantiating the effectiveness and utility of combined interventions are still to be clarified. Therefore, this opinion paper aims to provide a theoretical framework consisting of compelling arguments as to why adding NIBS to psychotherapy can promote therapeutic change. Namely, we will discuss the physiological effects of the two interventions, thus providing a rationale to explain the potential advantages of a combined approach.
Collapse
Affiliation(s)
- Daniele Saccenti
- Department of Psychology, Sigmund Freud University, Milan, Italy
| | - Leonor J. Romero Lauro
- Department of Psychology and NeuroMi, University of Milano-Bicocca, Milan, Italy
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
| | - Sofia A. Crespi
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea S. Moro
- Department of Psychology, Sigmund Freud University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| | | | | | - Novella Pretti
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
- Clinical Psychology Center, Division of Neurology, Galliera Hospital, Genoa, Italy
| | - Jacopo Lamanna
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| | - Mattia Ferro
- Department of Psychology, Sigmund Freud University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
7
|
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
|
8
|
Larose A, Miller CCJ, Mórotz GM. The lemur tail kinase family in neuronal function and disfunction in neurodegenerative diseases. Cell Mol Life Sci 2024; 81:447. [PMID: 39520508 PMCID: PMC11550312 DOI: 10.1007/s00018-024-05480-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024]
Abstract
The complex neuronal architecture and the long distance of synapses from the cell body require precisely orchestrated axonal and dendritic transport processes to support key neuronal functions including synaptic signalling, learning and memory formation. Protein phosphorylation is a major regulator of both intracellular transport and synaptic functions. Some kinases and phosphatases such as cyclin dependent kinase-5 (cdk5)/p35, glycogen synthase kinase-3β (GSK3β) and protein phosphatase-1 (PP1) are strongly involved in these processes. A primary pathological hallmark of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis/frontotemporal dementia, is synaptic degeneration together with disrupted intracellular transport. One attractive possibility is that alterations to key kinases and phosphatases may underlie both synaptic and axonal transport damages. The brain enriched lemur tail kinases (LMTKs, formerly known as lemur tyrosine kinases) are involved in intracellular transport and synaptic functions, and are also centrally placed in cdk5/p35, GSK3β and PP1 signalling pathways. Loss of LMTKs is documented in major neurodegenerative diseases and thus can contribute to pathological defects in these disorders. However, whilst function of their signalling partners became clearer in modulating both synaptic signalling and axonal transport progress has only recently been made around LMTKs. In this review, we describe this progress with a special focus on intracellular transport, synaptic functions and neurodegenerative diseases.
Collapse
Affiliation(s)
- Angelique Larose
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, H-1089, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Christopher C J Miller
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane Camberwell, London, SE5 9RX, UK.
| | - Gábor M Mórotz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, H-1089, Hungary.
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
9
|
Bin-Alamer O, Abou-Al-Shaar H, Efrati S, Hadanny A, Beckman RL, Elamir M, Sussman E, Maroon JC. Hyperbaric oxygen therapy as a neuromodulatory technique: a review of the recent evidence. Front Neurol 2024; 15:1450134. [PMID: 39445195 PMCID: PMC11496187 DOI: 10.3389/fneur.2024.1450134] [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: 06/17/2024] [Accepted: 08/23/2024] [Indexed: 10/25/2024] Open
Abstract
Hyperbaric oxygen therapy (HBOT) has recently emerged as a promising neuromodulatory modality for treating several neurological and psychological disorders. Various studies indicate that HBOT can promote brain recovery and neuroplasticity through the modulation of key cellular and molecular mechanisms. HBOT affects multiple primary pathways and cellular functions including mitochondrial biogenesis and function (increased Bcl-2, reduced Bax, and enhanced ATP production), neurogenesis (upregulation of Wnt-3 and VEGF/ERK signaling), synaptogenesis (elevated GAP43 and synaptophysin expression), and anti-inflammatory responses (reduced TNF-α and IL-6). These mechanisms contribute to significant clinical benefits, such as enhanced cognitive function, improved recovery from traumatic brain injury and post-concussion syndrome, and symptom reduction in conditions like post-traumatic stress disorder and fibromyalgia. By influencing these molecular targets, HBOT offers a novel approach to neuromodulation that warrants further exploration. This review discusses the representative mechanisms of action of HBOT and highlights its therapeutic neuromodulatory effects and potential clinical applications across various neurological and psychiatric conditions.
Collapse
Affiliation(s)
- Othman Bin-Alamer
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hussam Abou-Al-Shaar
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shai Efrati
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be'er Ya'akov, Israel
| | - Amir Hadanny
- Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf Harofeh) Medical Center, Be'er Ya'akov, Israel
| | - Robert L. Beckman
- Foundation for the Study of Inflammatory Disease, Bethesda, MD, United States
| | | | | | - Joseph C. Maroon
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
10
|
Bogaciu CA, Rizzoli SO. Membrane trafficking of synaptic adhesion molecules. J Physiol 2024. [PMID: 39322997 DOI: 10.1113/jp286401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/02/2024] [Indexed: 09/27/2024] Open
Abstract
Synapse formation and stabilization are aided by several families of adhesion molecules, which are generally seen as specialized surface receptors. The function of most surface receptors, including adhesion molecules, is modulated in non-neuronal cells by the processes of endocytosis and recycling, which control the number of active receptors found on the cell surface. These processes have not been investigated extensively at the synapse. This review focuses on the current status of this topic, summarizing general findings on the membrane trafficking of the most prominent synaptic adhesion molecules. Remarkably, evidence for endocytosis processes has been obtained for many synaptic adhesion proteins, including dystroglycans, latrophilins, calsyntenins, netrins, teneurins, neurexins, neuroligins and neuronal pentraxins. Less evidence has been obtained on their recycling, possibly because of the lack of specific assays. We conclude that the trafficking of the synaptic adhesion molecules is an important topic, which should receive more attention in the future.
Collapse
Affiliation(s)
- Cristian A Bogaciu
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| | - Silvio O Rizzoli
- Institute for Neuro- and Sensory Physiology and Biostructural Imaging of Neurodegeneration (BIN) Center, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
11
|
Wang G, Qi W, Liu QH, Guan W. GluN2A: A Promising Target for Developing Novel Antidepressants. Int J Neuropsychopharmacol 2024; 27:pyae037. [PMID: 39185814 DOI: 10.1093/ijnp/pyae037] [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] [Accepted: 08/23/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Depression is a heterogeneous disorder with high morbidity and disability rates that poses serious problems regarding mental health care. It is now well established that N-methyl D-aspartate receptor (NMDAR) modulators are being increasingly explored as potential therapeutic options for treating depression, although relatively little is known about their mechanisms of action. NMDARs are glutamate-gated ion channels that are ubiquitously expressed in the central nervous system (CNS), and they have been shown to play key roles in excitatory synaptic transmission. GluN2A, the predominant Glu2N subunit of functional NMDARs in neurons, is involved in various physiological processes in the CNS and is associated with diseases such as anxiety, depression, and schizophrenia. However, the role of GluN2A in the pathophysiology of depression has not yet been elucidated. METHODS We reviewed several past studies to better understand the function of GluN2A in depression. Additionally, we also summarized the pathogenesis of depression based on the regulation of GluN2A expression, particularly its interaction with neuroinflammation and neurogenesis, which has received considerable critical attention and is highly implicated in the onset of depression. RESULTS These evidence suggests that GluN2A overexpression impairs structural and functional synaptic plasticity, which contributes to the development of depression. Consequently, this knowledge is vital for the development of selective antagonists targeting GluN2A subunits using pharmacological and molecular methods. CONCLUSIONS Specific inhibition of the GluN2A NMDAR subunit is resistant to chronic stress-induced depressive-like behaviors, making them promising targets for the development of novel antidepressants.
Collapse
Affiliation(s)
- Gang Wang
- Department of Hepatobiliary Surgery, Zhangjiagang Hospital affiliated to Soochow University/The First People's Hospital of Zhangjiagang City, Zhangjiagang, China
| | - Wang Qi
- Department of Pharmacology, The First People's Hospital of Yancheng, Yancheng, China
| | - Qiu-Hua Liu
- Department of Hepatobiliary Surgery, Zhangjiagang Hospital affiliated to Soochow University/The First People's Hospital of Zhangjiagang City, Zhangjiagang, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, China
| |
Collapse
|
12
|
Miranda-Riestra A, Cercós MG, Trueta C, Oikawa-Sala J, Argueta J, Constantino-Jonapa LA, Cruz-Garduño R, Benítez-King G, Estrada-Reyes R. Participation of Ca 2+-Calmodulin-Dependent Protein Kinase II in the Antidepressant-Like Effects of Melatonin. Mol Pharmacol 2024; 106:107-116. [PMID: 39079719 DOI: 10.1124/molpharm.124.000890] [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: 02/08/2024] [Accepted: 06/26/2024] [Indexed: 08/18/2024] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) is an indoleamine secreted by the pineal gland during the dark phase of the photoperiod. Its main function is the synchronization of different body rhythms with the dark-light cycle. Research on melatonin has significantly advanced since its discovery and we now know that it has considerable significance in various physiological processes, including immunity, aging, and reproduction. Moreover, in recent years evidence of the pharmacological possibilities of melatonin has increased. Indoleamine, on the other hand, has antidepressant-like effects in rodents, which may be mediated by the activation of calcium-calmodulin-dependent kinase II (CaMKII) and are also related to the regulation of neuroplasticity processes, including neurogenesis, synaptic maintenance, and long-term potentiation. Remarkably, patients with major depression show decreased levels of circulating melatonin in plasma. This review presents evidence of the antidepressant-like effects of melatonin in preclinical models and the participation of CaMKII in these actions. CaMKII's role in cognition and memory processes, which are altered in depressive states, are part of the review, and the effects of melatonin in these processes are also reviewed. Furthermore, participation of CaMKII on structural and synaptic plasticity and the effects of melatonin are also described. Finally, the advantages of using melatonin in combination with other antidepressants such as ketamine for neuroplasticity are described. Evidence supports that CaMKII is activated by melatonin and downstream melatonin receptors and may be the common effector in the synergistic effects of melatonin with other antidepressants. SIGNIFICANCE STATEMENT: This review compiled evidence supporting that melatonin causes antidepressant-like effects in mice through calmodulin kinase II stimulation of downstream melatonin receptors as well as the participation of this enzyme in neuroplasticity, memory, and cognition. Finally, we describe evidence about the effectiveness of antidepressant-like effects of melatonin in combination with ketamine.
Collapse
Affiliation(s)
- Armida Miranda-Riestra
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Montserrat G Cercós
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Citlali Trueta
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Julián Oikawa-Sala
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Jesús Argueta
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Luis A Constantino-Jonapa
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Ricardo Cruz-Garduño
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Gloria Benítez-King
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| | - Rosa Estrada-Reyes
- Laboratorio de Neurofarmacología (A.M.-R., J.O.-S., J.A., L.A.C.-J., G.B.-K.), Departamento de Neurofisiología, Dirección de Investigaciones en Neurociencias (M.G.C., C.T., R.C.-G.), and Laboratorio de Fitofarmacología, Dirección de Investigaciones en Neurociencias (R.E.-R.), Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, Mexico
| |
Collapse
|
13
|
Gong R, Qin L, Chen L, Wang N, Bao Y, Lu W. Myosin Va-dependent Transport of NMDA Receptors in Hippocampal Neurons. Neurosci Bull 2024; 40:1053-1075. [PMID: 38291290 PMCID: PMC11306496 DOI: 10.1007/s12264-023-01174-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/03/2023] [Indexed: 02/01/2024] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR) trafficking is a key process in the regulation of synaptic efficacy and brain function. However, the molecular mechanism underlying the surface transport of NMDARs is largely unknown. Here we identified myosin Va (MyoVa) as the specific motor protein that traffics NMDARs in hippocampal neurons. We found that MyoVa associates with NMDARs through its cargo binding domain. This association was increased during NMDAR surface transport. Knockdown of MyoVa suppressed NMDAR transport. We further demonstrated that Ca2+/calmodulin-dependent protein kinase II (CaMKII) regulates NMDAR transport through its direct interaction with MyoVa. Furthermore, MyoVa employed Rab11 family-interacting protein 3 (Rab11/FIP3) as the adaptor proteins to couple themselves with NMDARs during their transport. Accordingly, the knockdown of FIP3 impairs hippocampal memory. Together, we conclude that in hippocampal neurons, MyoVa conducts active transport of NMDARs in a CaMKII-dependent manner.
Collapse
Affiliation(s)
- Ru Gong
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Linwei Qin
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Linlin Chen
- Department of Neurobiology, Nanjing Medical University, Nanjing, 210096, China
| | - Ning Wang
- Department of Neurobiology, Nanjing Medical University, Nanjing, 210096, China
| | - Yifei Bao
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China
| | - Wei Lu
- Ministry of Education (MOE) Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, 210096, China.
- Department of Neurosurgery, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China.
- Department of Neurobiology, Nanjing Medical University, Nanjing, 210096, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
14
|
Liu J, Niethard N, Lun Y, Dimitrov S, Ehrlich I, Born J, Hallschmid M. Slow-wave sleep drives sleep-dependent renormalization of synaptic AMPA receptor levels in the hypothalamus. PLoS Biol 2024; 22:e3002768. [PMID: 39163472 PMCID: PMC11364421 DOI: 10.1371/journal.pbio.3002768] [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: 02/28/2024] [Revised: 08/30/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
According to the synaptic homeostasis hypothesis (SHY), sleep serves to renormalize synaptic connections that have been potentiated during the prior wake phase due to ongoing encoding of information. SHY focuses on glutamatergic synaptic strength and has been supported by numerous studies examining synaptic structure and function in neocortical and hippocampal networks. However, it is unknown whether synaptic down-regulation during sleep occurs in the hypothalamus, i.e., a pivotal center of homeostatic regulation of bodily functions including sleep itself. We show that sleep, in parallel with the synaptic down-regulation in neocortical networks, down-regulates the levels of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) in the hypothalamus of rats. Most robust decreases after sleep were observed at both sites for AMPARs containing the GluA1 subunit. Comparing the effects of selective rapid eye movement (REM) sleep and total sleep deprivation, we moreover provide experimental evidence that slow-wave sleep (SWS) is the driving force of the down-regulation of AMPARs in hypothalamus and neocortex, with no additional contributions of REM sleep or the circadian rhythm. SWS-dependent synaptic down-regulation was not linked to EEG slow-wave activity. However, spindle density during SWS predicted relatively increased GluA1 subunit levels in hypothalamic synapses, which is consistent with the role of spindles in the consolidation of memory. Our findings identify SWS as the main driver of the renormalization of synaptic strength during sleep and suggest that SWS-dependent synaptic renormalization is also implicated in homeostatic control processes in the hypothalamus.
Collapse
Affiliation(s)
- Jianfeng Liu
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
| | - Niels Niethard
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
| | - Yu Lun
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
| | - Stoyan Dimitrov
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
| | - Ingrid Ehrlich
- Department of Neurobiology, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
- Center for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University Tübingen (IDM), Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| | - Manfred Hallschmid
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University Tübingen (IDM), Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| |
Collapse
|
15
|
Nowacka A, Getz AM, Bessa-Neto D, Choquet D. Activity-dependent diffusion trapping of AMPA receptors as a key step for expression of early LTP. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230220. [PMID: 38853553 PMCID: PMC11343219 DOI: 10.1098/rstb.2023.0220] [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: 02/24/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 06/11/2024] Open
Abstract
This review focuses on the activity-dependent diffusion trapping of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) as a crucial mechanism for the expression of early long-term potentiation (LTP), a process central to learning and memory. Despite decades of research, the precise mechanisms by which LTP induction leads to an increase in AMPAR responses at synapses have been elusive. We review the different hypotheses that have been put forward to explain the increased AMPAR responsiveness during LTP. We discuss the dynamic nature of AMPAR complexes, including their constant turnover and activity-dependent modifications that affect their synaptic accumulation. We highlight a hypothesis suggesting that AMPARs are diffusively trapped at synapses through activity-dependent interactions with protein-based binding slots in the post-synaptic density (PSD), offering a potential explanation for the increased synaptic strength during LTP. Furthermore, we outline the challenges still to be addressed before we fully understand the functional roles and molecular mechanisms of AMPAR dynamic nanoscale organization in LTP. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Agata Nowacka
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
| | - Angela M. Getz
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, BordeauxF-33000, France
| | - Diogo Bessa-Neto
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
| | - Daniel Choquet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, BordeauxF-33000, France
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, BordeauxF-33000, France
| |
Collapse
|
16
|
Yamamoto K, Chen QY, Zhou Z, Kobayashi M, Zhuo M. Cortical nitric oxide required for presynaptic long-term potentiation in the insular cortex. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230475. [PMID: 38853563 PMCID: PMC11343264 DOI: 10.1098/rstb.2023.0475] [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: 10/25/2023] [Revised: 03/07/2024] [Accepted: 03/28/2024] [Indexed: 06/11/2024] Open
Abstract
Nitric oxide (NO) is a key diffusible messenger in the mammalian brain. It has been proposed that NO may diffuse retrogradely into presynaptic terminals, contributing to the induction of hippocampal long-term potentiation (LTP). Here, we present novel evidence that NO is required for kainate receptor (KAR)-dependent presynaptic form of LTP (pre-LTP) in the adult insular cortex (IC). In the IC, we found that inhibition of NO synthase erased the maintenance of pre-LTP, while the induction of pre-LTP required the activation of KAR. Furthermore, NO is essential for pre-LTP induced between two pyramidal cells in the IC using the double patch-clamp recording. These results suggest that NO is required for homosynaptic pre-LTP in the IC. Our results present strong evidence for the critical roles of NO in pre-LTP in the IC. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Kiyofumi Yamamoto
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo101-8310, Japan
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, OntarioM5S 1A8, Canada
| | - Qi-Yu Chen
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, OntarioM5S 1A8, Canada
- Zhuomin Institute for Brain Research, Qingdao266000, People's Republic of China
- CAS Key Laboratory of Brain Connectome and Manipulation, Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen Institute of Advanced Technology, Shenzhen518055, People's Republic of China
| | - Zhaoxiang Zhou
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, OntarioM5S 1A8, Canada
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou510130, People's Republic of China
| | - Masayuki Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo101-8310, Japan
| | - Min Zhuo
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, OntarioM5S 1A8, Canada
- Zhuomin Institute for Brain Research, Qingdao266000, People's Republic of China
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou510130, People's Republic of China
| |
Collapse
|
17
|
Wei H, Gong J, Liu J, He G, Ni Y, Fu C, Yang L, Guo J, Xu Z, Xu W. Thermally and Mechanically Stable Perovskite Artificial Synapse as Tuned by Phase Engineering for Efferent Neuromuscular Control. NANO LETTERS 2024. [PMID: 39023921 DOI: 10.1021/acs.nanolett.4c02240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The doping of perovskites with mixed cations and mixed halides is an effective strategy to optimize phase stability. In this study, we introduce a cubic black phase perovskite CsyFA(1-y)Pb(BrxI(1-x))3 artificial synapse, using phase engineering by adjusting the cesium-bromide content. Low-bromine mixed perovskites are suitable to improve the electric pulse excitation sensitivity and stability of the device. Specifically, the low-bromine and low-cesium mixed perovskite (x = 0.15, y = 0.22) annealed at 373 K allows the device to maintain logic response even after 1000 mechanical flex/flat cycles. The device also shows good thermal stability up to temperatures of 333 K. We have demonstrated reflex-arc behavior with MCMHP synaptic units, capable of making sensory warnings at high frequency. This compositionally engineered, dual-mixed perovskite synaptic device provides significant potential for perceptual soft neurorobotic systems and prostheses.
Collapse
Affiliation(s)
| | - Jiangdong Gong
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
- Center for Intelligent Medical Equipment and Devices, Institute for Innovative Medical Devices, Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu 215123, People's Republic of China
| | - Jiaqi Liu
- Institute of Photoelectronic Thin Film Devices and Technology, Nankai University, Tianjin 300350, People's Republic of China
| | | | - Yao Ni
- School of Integrated Circuits, Guangdong University of Technology, Guangzhou, Guangdong 510006, People's Republic of China
| | | | - Lu Yang
- Institute of Photoelectronic Thin Film Devices and Technology, Nankai University, Tianjin 300350, People's Republic of China
| | - Jiahao Guo
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, People's Republic of China
- Center for Intelligent Medical Equipment and Devices, Institute for Innovative Medical Devices, Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu 215123, People's Republic of China
| | - Zhipeng Xu
- Institute of Photoelectronic Thin Film Devices and Technology, Nankai University, Tianjin 300350, People's Republic of China
| | - Wentao Xu
- Institute of Photoelectronic Thin Film Devices and Technology, Nankai University, Tianjin 300350, People's Republic of China
| |
Collapse
|
18
|
Jung JTK, Marques LS, Brambila CA, da Cruz Weber Fulco B, Nogueira CW, Zeni G. Social-Single Prolonged Stress affects contextual fear conditioning in male and female Wistar rats: Molecular insights in the amygdala. Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:111021. [PMID: 38692472 DOI: 10.1016/j.pnpbp.2024.111021] [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: 12/30/2023] [Revised: 04/08/2024] [Accepted: 04/28/2024] [Indexed: 05/03/2024]
Abstract
Stress exposure can lead to post-traumatic stress disorder (PTSD) in male and female rats. Social-Single Prolonged Stress (SPS) protocol has been considered a potential PTSD model. This study aimed to pharmacologically validate the Social-SPS as a PTSD model in male and female rats. Male and female Wistar rats (60-day-old) were exposed to Social-SPS protocol and treated with fluoxetine (10 mg/Kg) or saline solution intraperitoneally 24 h before euthanasia. Two cohorts of animals were used; for cohort 1, male and female rats were still undisturbed until day 7 post-Social-SPS exposure, underwent locomotor and conditioned fear behaviors, and were euthanized on day 9. Animals of cohort 2 were subjected to the same protocol but were re-exposed to contextual fear behavior on day 14. Results showed that fluoxetine-treated rats gained less body weight than control and Social-SPS in both sexes. Social-SPS effectively increased the freezing time in male and female rats on day eight but not on day fourteen. Fluoxetine blocked the increase of freezing in male and female rats on day 8. Different mechanisms for fear behavior were observed in males, such as Social-SPS increased levels of glucocorticoid receptors and Beclin-1 in the amygdala. Social-SPS was shown to increase the levels of NMDA2A, GluR-1, PSD-95, and CAMKII in the amygdala of female rats. No alterations were observed in the amygdala of rats on day fourteen. The study revealed that Social-SPS is a potential PTSD protocol applicable to both male and female rats.
Collapse
MESH Headings
- Animals
- Male
- Female
- Fear/drug effects
- Fear/physiology
- Rats, Wistar
- Fluoxetine/pharmacology
- Amygdala/drug effects
- Amygdala/metabolism
- Stress, Psychological/metabolism
- Rats
- Disease Models, Animal
- Stress Disorders, Post-Traumatic/metabolism
- Stress Disorders, Post-Traumatic/psychology
- Conditioning, Classical/drug effects
- Conditioning, Classical/physiology
- Conditioning, Psychological/drug effects
- Conditioning, Psychological/physiology
- Selective Serotonin Reuptake Inhibitors/pharmacology
- Disks Large Homolog 4 Protein
- Receptors, AMPA
Collapse
Affiliation(s)
- Juliano Ten Kathen Jung
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Luiza Souza Marques
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Carlos Alexandre Brambila
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Bruna da Cruz Weber Fulco
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Cristina Wayne Nogueira
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Gilson Zeni
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
19
|
Harvey J. Novel Leptin-Based Therapeutic Strategies to Limit Synaptic Dysfunction in Alzheimer's Disease. Int J Mol Sci 2024; 25:7352. [PMID: 39000459 PMCID: PMC11242278 DOI: 10.3390/ijms25137352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Accumulation of hyper-phosphorylated tau and amyloid beta (Aβ) are key pathological hallmarks of Alzheimer's disease (AD). Increasing evidence indicates that in the early pre-clinical stages of AD, phosphorylation and build-up of tau drives impairments in hippocampal excitatory synaptic function, which ultimately leads to cognitive deficits. Consequently, limiting tau-related synaptic abnormalities may have beneficial effects in AD. There is now significant evidence that the hippocampus is an important brain target for the endocrine hormone leptin and that leptin has pro-cognitive properties, as activation of synaptic leptin receptors markedly influences higher cognitive processes including learning and memory. Clinical studies have identified a link between the circulating leptin levels and the risk of AD, such that AD risk is elevated when leptin levels fall outwith the physiological range. This has fuelled interest in targeting the leptin system therapeutically. Accumulating evidence supports this possibility, as numerous studies have shown that leptin has protective effects in a variety of models of AD. Recent findings have demonstrated that leptin has beneficial effects in the preclinical stages of AD, as leptin prevents the early synaptic impairments driven by tau protein and amyloid β. Here we review recent findings that implicate the leptin system as a potential novel therapeutic target in AD.
Collapse
Affiliation(s)
- Jenni Harvey
- Department of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
20
|
Inagaki R, Yamakuni T, Saito T, Saido TC, Moriguchi S. Preventive effect of propolis on cognitive decline in Alzheimer's disease model mice. Neurobiol Aging 2024; 139:20-29. [PMID: 38583392 DOI: 10.1016/j.neurobiolaging.2024.03.002] [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: 12/05/2023] [Revised: 02/20/2024] [Accepted: 03/11/2024] [Indexed: 04/09/2024]
Abstract
Brazilian green propolis (propolis) is a chemically complex resinous substance that is a potentially viable therapeutic agent for Alzheimer's disease. Herein, propolis induced a transient increase in intracellular Ca2+ concentration ([Ca2+]i) in Neuro-2A cells; moreover, propolis-induced [Ca2+]i elevations were suppressed prior to 24-h pretreatment with amyloid-β. To reveal the effect of [Ca2+]i elevation on impaired cognition, we performed memory-related behavioral tasks in APP-KI mice relative to WT mice at 4 and 12 months of age. Propolis, at 300-1000 mg/kg/d for 8 wk, significantly ameliorated cognitive deficits in APP-KI mice at 4 months, but not at 12 months of age. Consistent with behavioral observations, injured hippocampal long-term potentiation was markedly ameliorated in APP-KI mice at 4 months of age following repeated propolis administration. In addition, repeated administration of propolis significantly activated intracellular calcium signaling pathway in the CA1 region of APP-KI mice. These results suggest a preventive effect of propolis on cognitive decline through the activation of intracellular calcium signaling pathways in CA1 region of AD mice model.
Collapse
Affiliation(s)
- Ryo Inagaki
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Tohru Yamakuni
- Research Center of Supercritical Fluid Technology, Graduate School of Engineering, Tohoku University, Sendai, Japan; New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan
| | - Shigeki Moriguchi
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
21
|
Reddien PW. The purpose and ubiquity of turnover. Cell 2024; 187:2657-2681. [PMID: 38788689 DOI: 10.1016/j.cell.2024.04.034] [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: 01/31/2024] [Revised: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024]
Abstract
Turnover-constant component production and destruction-is ubiquitous in biology. Turnover occurs across organisms and scales, including for RNAs, proteins, membranes, macromolecular structures, organelles, cells, hair, feathers, nails, antlers, and teeth. For many systems, turnover might seem wasteful when degraded components are often fully functional. Some components turn over with shockingly high rates and others do not turn over at all, further making this process enigmatic. However, turnover can address fundamental problems by yielding powerful properties, including regeneration, rapid repair onset, clearance of unpredictable damage and errors, maintenance of low constitutive levels of disrepair, prevention of stable hazards, and transitions. I argue that trade-offs between turnover benefits and metabolic costs, combined with constraints on turnover, determine its presence and rates across distinct contexts. I suggest that the limits of turnover help explain aging and that turnover properties and the basis for its levels underlie this fundamental component of life.
Collapse
Affiliation(s)
- Peter W Reddien
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, MIT, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
22
|
Zhou H, Bi GQ, Liu G. Intracellular magnesium optimizes transmission efficiency and plasticity of hippocampal synapses by reconfiguring their connectivity. Nat Commun 2024; 15:3406. [PMID: 38649706 PMCID: PMC11035601 DOI: 10.1038/s41467-024-47571-3] [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: 07/24/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Synapses at dendritic branches exhibit specific properties for information processing. However, how the synapses are orchestrated to dynamically modify their properties, thus optimizing information processing, remains elusive. Here, we observed at hippocampal dendritic branches diverse configurations of synaptic connectivity, two extremes of which are characterized by low transmission efficiency, high plasticity and coding capacity, or inversely. The former favors information encoding, pertinent to learning, while the latter prefers information storage, relevant to memory. Presynaptic intracellular Mg2+ crucially mediates the dynamic transition continuously between the two extreme configurations. Consequently, varying intracellular Mg2+ levels endow individual branches with diverse synaptic computations, thus modulating their ability to process information. Notably, elevating brain Mg2+ levels in aging animals restores synaptic configuration resembling that of young animals, coincident with improved learning and memory. These findings establish intracellular Mg2+ as a crucial factor reconfiguring synaptic connectivity at dendrites, thus optimizing their branch-specific properties in information processing.
Collapse
Affiliation(s)
- Hang Zhou
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518107, China.
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Guo-Qiang Bi
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518107, China
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518055, China
- Hefei National Laboratory for Physical Sciences at the Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei, 230031, China
| | - Guosong Liu
- School of Medicine, Tsinghua University, Beijing, 100084, China.
- NeuroCentria Inc., Walnut Creek, CA, 94596, USA.
| |
Collapse
|
23
|
Chen Y, Liu S, Jacobi AA, Jeng G, Ulrich JD, Stein IS, Patriarchi T, Hell JW. Rapid sequential clustering of NMDARs, CaMKII, and AMPARs upon activation of NMDARs at developing synapses. Front Synaptic Neurosci 2024; 16:1291262. [PMID: 38660466 PMCID: PMC11039796 DOI: 10.3389/fnsyn.2024.1291262] [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: 09/08/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Rapid, synapse-specific neurotransmission requires the precise alignment of presynaptic neurotransmitter release and postsynaptic receptors. How postsynaptic glutamate receptor accumulation is induced during maturation is not well understood. We find that in cultures of dissociated hippocampal neurons at 11 days in vitro (DIV) numerous synaptic contacts already exhibit pronounced accumulations of the pre- and postsynaptic markers synaptotagmin, synaptophysin, synapsin, bassoon, VGluT1, PSD-95, and Shank. The presence of an initial set of AMPARs and NMDARs is indicated by miniature excitatory postsynaptic currents (mEPSCs). However, AMPAR and NMDAR immunostainings reveal rather smooth distributions throughout dendrites and synaptic enrichment is not obvious. We found that brief periods of Ca2+ influx through NMDARs induced a surprisingly rapid accumulation of NMDARs within 1 min, followed by accumulation of CaMKII and then AMPARs within 2-5 min. Postsynaptic clustering of NMDARs and AMPARs was paralleled by an increase in their mEPSC amplitudes. A peptide that blocked the interaction of NMDAR subunits with PSD-95 prevented the NMDAR clustering. NMDAR clustering persisted for 3 days indicating that brief periods of elevated glutamate fosters permanent accumulation of NMDARs at postsynaptic sites in maturing synapses. These data support the model that strong glutamatergic stimulation of immature glutamatergic synapses results in a fast and substantial increase in postsynaptic NMDAR content that required NMDAR binding to PSD-95 or its homologues and is followed by recruitment of CaMKII and subsequently AMPARs.
Collapse
Affiliation(s)
- Yucui Chen
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
| | - Shangming Liu
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Ariel A. Jacobi
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Grace Jeng
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Jason D. Ulrich
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
| | - Ivar S. Stein
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Tommaso Patriarchi
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Johannes W. Hell
- Department of Pharmacology, University of Iowa, Iowa City, IA, United States
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
24
|
Odriozola A, González A, Álvarez-Herms J, Corbi F. Sleep regulation and host genetics. ADVANCES IN GENETICS 2024; 111:497-535. [PMID: 38908905 DOI: 10.1016/bs.adgen.2024.02.002] [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: 06/24/2024]
Abstract
Due to the multifactorial and complex nature of rest, we focus on phenotypes related to sleep. Sleep regulation is a multifactorial process. In this chapter, we focus on those phenotypes inherent to sleep that are highly prevalent in the population, and that can be modulated by lifestyle, such as sleep quality and duration, insomnia, restless leg syndrome and daytime sleepiness. We, therefore, leave in the background those phenotypes that constitute infrequent pathologies or for which the current level of scientific evidence does not favour the implementation of practical approaches of this type. Similarly, the regulation of sleep quality is intimately linked to the regulation of the circadian rhythm. Although this relationship is discussed in the sections that require it, the in-depth study of circadian rhythm regulation at the molecular level deserves a separate chapter, and this is how it is dealt with in this volume.
Collapse
Affiliation(s)
- Adrián Odriozola
- Hologenomiks Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Adriana González
- Hologenomiks Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jesús Álvarez-Herms
- Phymo® Lab, Physiology, and Molecular Laboratory, Collado Hermoso, Segovia, Spain
| | - Francesc Corbi
- Institut Nacional d'Educació Física de Catalunya (INEFC), Centre de Lleida, Universitat de Lleida (UdL), Lleida, Spain
| |
Collapse
|
25
|
Khodaie B, Edelmann E, Leßmann V. Distinct GABAergic modulation of timing-dependent LTP in CA1 pyramidal neurons along the longitudinal axis of the mouse hippocampus. iScience 2024; 27:109320. [PMID: 38487018 PMCID: PMC10937841 DOI: 10.1016/j.isci.2024.109320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 01/25/2024] [Accepted: 02/20/2024] [Indexed: 03/17/2024] Open
Abstract
Synaptic plasticity in the hippocampus underlies episodic memory formation, with dorsal hippocampus being instrumental for spatial memory whereas ventral hippocampus is crucial for emotional learning. Here, we studied how GABAergic inhibition regulates physiologically relevant low repeat spike timing-dependent LTP (t-LTP) at Schaffer collateral-CA1 synapses along the dorsoventral hippocampal axis. We used two t-LTP protocols relying on only 6 repeats of paired spike-firing in pre- and postsynaptic cells within 10 s that differ in postsynaptic firing patterns. GABAA receptor mechanisms played a greater role in blocking 6× 1:1 t-LTP that recruits single postsynaptic action potentials. 6× 1:4 t-LTP that depends on postsynaptic burst-firing unexpectedly required intact GABAB receptor signaling. The magnitude of both t-LTP-forms decreased along the dorsoventral axis, despite increasing excitability and basal synaptic strength in this direction. This suggests that GABAergic inhibition contributes to the distinct roles of dorsal and ventral hippocampus in memory formation.
Collapse
Affiliation(s)
- Babak Khodaie
- Institut für Physiologie, Otto-von-Guericke-Universität (OVGU), Medizinische Fakultät, 39120 Magdeburg, Germany
- OVGU International ESF-funded Graduate School ABINEP, 39104 Magdeburg, Germany
| | - Elke Edelmann
- Institut für Physiologie, Otto-von-Guericke-Universität (OVGU), Medizinische Fakultät, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences, 39104 Magdeburg, Germany
- OVGU International ESF-funded Graduate School ABINEP, 39104 Magdeburg, Germany
| | - Volkmar Leßmann
- Institut für Physiologie, Otto-von-Guericke-Universität (OVGU), Medizinische Fakultät, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences, 39104 Magdeburg, Germany
- OVGU International ESF-funded Graduate School ABINEP, 39104 Magdeburg, Germany
- DZPG (German Center of Mental Health), partner site Halle/Jena/Magdeburg (CIRC), Magdeburg, Germany
| |
Collapse
|
26
|
He W, Shi X, Dong Z. The roles of RACK1 in the pathogenesis of Alzheimer's disease. J Biomed Res 2024; 38:137-148. [PMID: 38410996 PMCID: PMC11001590 DOI: 10.7555/jbr.37.20220259] [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: 12/20/2022] [Revised: 04/15/2023] [Accepted: 04/24/2023] [Indexed: 02/28/2024] Open
Abstract
The receptor for activated C kinase 1 (RACK1) is a protein that plays a crucial role in various signaling pathways and is involved in the pathogenesis of Alzheimer's disease (AD), a prevalent neurodegenerative disease. RACK1 is highly expressed in neuronal cells of the central nervous system and regulates the pathogenesis of AD. Specifically, RACK1 is involved in regulation of the amyloid-β precursor protein processing through α- or β-secretase by binding to different protein kinase C isoforms. Additionally, RACK1 promotes synaptogenesis and synaptic plasticity by inhibiting N-methyl-D-aspartate receptors and activating gamma-aminobutyric acid A receptors, thereby preventing neuronal excitotoxicity. RACK1 also assembles inflammasomes that are involved in various neuroinflammatory pathways, such as nuclear factor-kappa B, tumor necrosis factor-alpha, and NOD-like receptor family pyrin domain-containing 3 pathways. The potential to design therapeutics that block amyloid-β accumulation and inflammation or precisely regulate synaptic plasticity represents an attractive therapeutic strategy, in which RACK1 is a potential target. In this review, we summarize the contribution of RACK1 to the pathogenesis of AD and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Wenting He
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xiuyu Shi
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Zhifang Dong
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| |
Collapse
|
27
|
Basoglu H, Degirmencioglu İ, Ozturk H, Yorulmaz N, Aydin‐Abidin S, Abidin I. Title: A Boron‐Chelating Piperazine‐Tethered Schiff Base Can Modulate Excitability in Brain Slices in a Specific Frequency Range. ChemistrySelect 2023; 8. [DOI: 10.1002/slct.202303410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2025]
Abstract
AbstractThe ability of boron‐containing compounds to make various bonds with biological targets draws attention in their use as new therapeutic agents. In this study, the effect of a newly synthesized molecule with the short name fmpemboron, “Difluoro [2‐[([2‐[4‐(2,3,4‐trimethoxybenzyl)piperazin‐1‐yl]ethyl] imino‐κN)methyl]phenolato‐κO]boron (5)”on the excitability of neurons in the brain was investigated for the first time. First of all, fmpemboron was synthesized and characterized. Secondly, virtual scanning of the molecule was performed using ADME and molecular docking methods. Then, epileptiform activities were induced in mouse brain slices using Mg‐free or 4AP methods, and the effect of fmpemboron (5) at different concentrations was examined. The absorbance peak wavelength of fmpemboron (5) is between 330–340 nm and the fluorescence emission peak is 435 nm. According to ADME and molecular docking results, fmpemboron (5) can cross the blood‐brain barrier (BBB) and has affinity for the NMDA receptor. It also significantly reduces the power of oscillations between 8–12 Hz and 13–29 Hz in epileptiform activities generated by the Mg‐free method. Administration of 10 μM fmpemboron (5) had a modulatory effect on epileptiform activities, indicating that fmpemboron (5) can affect various neurological functions through NMDA channels. However, in‐vivo dose‐dependent studies are required to further investigate the effects of this novel molecule.
Collapse
Affiliation(s)
- Harun Basoglu
- Department of Biophysics Faculty of Medicine Karadeniz Technical University Trabzon Turkey
| | - İsmail Degirmencioglu
- Department of Chemistry Faculty of Science Karadeniz Technical University Trabzon Turkey
| | - Hilal Ozturk
- Department of Biophysics Faculty of Medicine Karadeniz Technical University Trabzon Turkey
| | - Nuri Yorulmaz
- Department of Physics Faculty of Science Harran University, Sanliurfa, Turkey Department of Biophysics Faculty of Medicine Karadeniz Technical University Trabzon Turkey
| | - Selcen Aydin‐Abidin
- Department of Biophysics Faculty of Medicine Karadeniz Technical University Trabzon Turkey
| | - Ismail Abidin
- Department of Biophysics Faculty of Medicine Karadeniz Technical University Trabzon Turkey
| |
Collapse
|
28
|
Bliznyuk A, Grossman Y. Role of NMDA Receptor in High-Pressure Neurological Syndrome and Hyperbaric Oxygen Toxicity. Biomolecules 2023; 13:1786. [PMID: 38136657 PMCID: PMC10742241 DOI: 10.3390/biom13121786] [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: 10/27/2023] [Revised: 11/26/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Professional divers exposed to pressures greater than 11 ATA (1.1 MPa) may suffer from high-pressure neurological syndrome (HPNS). Divers who use closed-circuit breathing apparatus and patients and medical attendants undergoing hyperbaric oxygen therapy (HBOT) face the risk of CNS hyperbaric oxygen toxicity (HBOTx) at oxygen pressure above 2 ATA (0.2 MPa). Both syndromes are characterized by reversible CNS hyperexcitability, accompanied by cognitive and motor deficits, and N-methyl-D-aspartate receptor (NMDAR) plays a crucial role in provoking them. Various NMDAR subtypes respond differently under hyperbaric conditions. The augmented currents observed only in NMDAR containing GluN2A subunit increase glutamatergic synaptic activity and cause dendritic hyperexcitability and abnormal neuronal activity. Removal of the resting Zn2+ voltage-independent inhibition exerted by GluN2A present in the NMDAR is the major candidate for the mechanism underlying the increase in receptor conductance. Therefore, this process should be the main target for future research aiming at developing neuroprotection against HPNS and HBOTx.
Collapse
Affiliation(s)
- Alice Bliznyuk
- Ilse Katz Institute for Nanoscale Science and Technology (IKI), Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Yoram Grossman
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel;
| |
Collapse
|
29
|
Zernov N, Popugaeva E. Role of Neuronal TRPC6 Channels in Synapse Development, Memory Formation and Animal Behavior. Int J Mol Sci 2023; 24:15415. [PMID: 37895105 PMCID: PMC10607207 DOI: 10.3390/ijms242015415] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
The transient receptor potential cation channel, subfamily C, member 6 (TRPC6), has been believed to adjust the formation of an excitatory synapse. The positive regulation of TRPC6 engenders synapse enlargement and improved learning and memory in animal models. TRPC6 is involved in different synaptoprotective signaling pathways, including antagonism of N-methyl-D-aspartate receptor (NMDAR), activation of brain-derived neurotrophic factor (BDNF) and postsynaptic store-operated calcium entry. Positive regulation of TRPC6 channels has been repeatedly shown to be good for memory formation and storage. TRPC6 is mainly expressed in the hippocampus, particularly in the dentate granule cells, cornu Ammonis 3 (CA3) pyramidal cells and gamma-aminobutyric acid (GABA)ergic interneurons. It has been observed that TRPC6 agonists have a great influence on animal behavior including memory formation and storage The purpose of this review is to collect the available information on the role of TRPC6 in memory formation in various parts of the brain to understand how TRPC6-specific pharmaceutical agents will affect memory in distinct parts of the central nervous system (CNS).
Collapse
Affiliation(s)
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| |
Collapse
|
30
|
Nicoll RA, Schulman H. Synaptic memory and CaMKII. Physiol Rev 2023; 103:2877-2925. [PMID: 37290118 PMCID: PMC10642921 DOI: 10.1152/physrev.00034.2022] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 06/10/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) and long-term potentiation (LTP) were discovered within a decade of each other and have been inextricably intertwined ever since. However, like many marriages, it has had its up and downs. Based on the unique biochemical properties of CaMKII, it was proposed as a memory molecule before any physiological linkage was made to LTP. However, as reviewed here, the convincing linkage of CaMKII to synaptic physiology and behavior took many decades. New technologies were critical in this journey, including in vitro brain slices, mouse genetics, single-cell molecular genetics, pharmacological reagents, protein structure, and two-photon microscopy, as were new investigators attracted by the exciting challenge. This review tracks this journey and assesses the state of this marriage 40 years on. The collective literature impels us to propose a relatively simple model for synaptic memory involving the following steps that drive the process: 1) Ca2+ entry through N-methyl-d-aspartate (NMDA) receptors activates CaMKII. 2) CaMKII undergoes autophosphorylation resulting in constitutive, Ca2+-independent activity and exposure of a binding site for the NMDA receptor subunit GluN2B. 3) Active CaMKII translocates to the postsynaptic density (PSD) and binds to the cytoplasmic C-tail of GluN2B. 4) The CaMKII-GluN2B complex initiates a structural rearrangement of the PSD that may involve liquid-liquid phase separation. 5) This rearrangement involves the PSD-95 scaffolding protein, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), and their transmembrane AMPAR-regulatory protein (TARP) auxiliary subunits, resulting in an accumulation of AMPARs in the PSD that underlies synaptic potentiation. 6) The stability of the modified PSD is maintained by the stability of the CaMKII-GluN2B complex. 7) By a process of subunit exchange or interholoenzyme phosphorylation CaMKII maintains synaptic potentiation in the face of CaMKII protein turnover. There are many other important proteins that participate in enlargement of the synaptic spine or modulation of the steps that drive and maintain the potentiation. In this review we critically discuss the data underlying each of the steps. As will become clear, some of these steps are more firmly grounded than others, and we provide suggestions as to how the evidence supporting these steps can be strengthened or, based on the new data, be replaced. Although the journey has been a long one, the prospect of having a detailed cellular and molecular understanding of learning and memory is at hand.
Collapse
Affiliation(s)
- Roger A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California, United States
| | - Howard Schulman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California, United States
- Panorama Research Institute, Sunnyvale, California, United States
| |
Collapse
|
31
|
He J, Lian Y. Clinical study of autonomic dysfunction in patients with autoimmune encephalitis. Immunobiology 2023; 228:152711. [PMID: 37543010 DOI: 10.1016/j.imbio.2023.152711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/25/2023] [Accepted: 07/15/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND Autoimmune encephalitis (AE) is a collective name, covering an emerging spectrum of autoimmune-mediated neurological diseases related to antibodies and synaptic or intracellular proteins. Anti-NMDAR, anti-LGI1, and anti-GABABR are three types of neuronal cell surface antibodies. Autonomic dysfunction represents a frequently occurring clinical manifestation. This observational study purposes to investigate comparisons between two groups with or without autonomic dysfunction and detect the autonomic dysfunction and other indexes in anti-NMDAR, anti-LGI1, and anti-GABABR cohorts. METHODS Patients with anti-NMDAR, anti-LGI1 and anti-GABABR encephalitis were recruited from the May 2017 to the April 2022. The following information was recorded: age, age at onset, tumor presence, gender, prodromal symptoms, clinical manifestations, cranial magnetic resonance imaging, cerebrospinal fluid and blood examinations, and immunotherapy. RESULTS There were totally 161 patients enrolled in this study. Among these participants, 104 individuals (64.6%) presented autonomic dysfunction and the remaining 57 (35.4%) were free of autonomic dysfunction. Sinus tachycardia was the most common autonomic dysfunction, followed by pollakiuria/uroclepsia, feverscence, central hypoventilation, sinus bradycardia, constipation, uroschesis, hyperhidrosis, hypersalivation, hypotension, and early satiety/emesis. Compared to patients without autonomic dysfunction, those with autonomic dysfunction had a higher incidence of central hypoventilation and ICU admissions. Meanwhile, in both groups with or without autonomic dysfunction, meatal behavior disorder, cognitive impairment, and epileptic seizure were three most common clinical manifestations. There were no significant differences in cranial magnetic resonance imaging (MRI), cerebrospinal fluid (CSF) examination, antibody titers and number of immunotherapy types. Further analysis of AE mediated by distinct neuronal surface antibodies demonstrated that there were 85 anti-NMDAR, 56 anti-LGI1, and 20 anti-GABABR encephalitis patients. The significant differences between these three cohorts appeared in age, tumor presence, fervescence presence and antibody titers. CONCLUSION This study demonstrated the comparisons between autonomic dysfunction group and autonomic dysfunction-free group and provided insights into better diagnosis and treatment.
Collapse
Affiliation(s)
- Jiao He
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yajun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| |
Collapse
|
32
|
Parvez S, Ramachandran B, Kaushik M, Tabassum H, Frey JU. Long-term depression induction and maintenance across regions of the apical branch of CA1 dendrites. Hippocampus 2023; 33:1058-1066. [PMID: 37254828 DOI: 10.1002/hipo.23553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 05/10/2023] [Accepted: 05/10/2023] [Indexed: 06/01/2023]
Abstract
Well known as the center for learning and memory, hippocampus is the crucial brain region to study synaptic plasticity in the context of cellular fundamental mechanisms such as long-term depression (LTD) and long-term potentiation (LTP). However, despite years of extensive research, the key to our LTD queries and their induction mechanisms has not been fully understood. Previously, we reported the induction of late-LTD (L-LTD) in the distally located synapses of apical branch of hippocampal CA1 dendrites using strong low-frequency stimulation (SLFS). In contrast synapses at the proximal site could not express L-LTD. Thus, in the present study, we wanted to investigate whether or not synapses of apical dendritic branch at the proximal location could induce and maintain LTD and its related properties in in vitro rat hippocampal slices. Results indicated that the SLFS in the distal and proximal region triggered the plasticity related proteins (PRP) synthesis in both regions, as evident by the induction and maintenance of L-LTD in the distal region by virtue of synaptic and cross-tagging. In addition, the application of emetine at the time of proximal input stimulation prevented the transition of early-LTD (E-LTD) into L-LTD at the distal region, proving PRP synthesis at the proximal site. Further, it was observed that weak low-frequency stimulation (WLFS) could induce E-LTD in the proximal region along with LTD-specific tag-setting at the synapses. In conclusion, the current study suggests unique findings that the synaptic and cross-tagging mediate L-LTD expression is maintained in the proximal location of hippocampus apical CA1 dendrites.
Collapse
Affiliation(s)
- Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
- Department of Neurophysiology, Leibniz-Institute for Neurobiology, Magdeburg, Germany
| | - Binu Ramachandran
- Department of Neurophysiology, Leibniz-Institute for Neurobiology, Magdeburg, Germany
- Neuronal Plasticity Group, Department of Zoology, University of Calicut, Malappuram, Kerala, India
| | - Medha Kaushik
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Heena Tabassum
- Department of Neurophysiology, Leibniz-Institute for Neurobiology, Magdeburg, Germany
- Division of Basic Medical Sciences, Indian Council of Medical Research, New Delhi, India
| | - Julietta U Frey
- Department of Neuroloy, Medical College of Georgia, Brain & Behavior Discovery Institute, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
33
|
Melanis K, Stefanou MI, Themistoklis KM, Papasilekas T. mTOR pathway - a potential therapeutic target in stroke. Ther Adv Neurol Disord 2023; 16:17562864231187770. [PMID: 37576547 PMCID: PMC10413897 DOI: 10.1177/17562864231187770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/27/2023] [Indexed: 08/15/2023] Open
Abstract
Stroke is ranked as the second leading cause of death worldwide and a major cause of long-term disability. A potential therapeutic target that could offer favorable outcomes in stroke is the mammalian target of rapamycin (mTOR) pathway. mTOR is a serine/threonine kinase that composes two protein complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), and is regulated by other proteins such as the tuberous sclerosis complex. Through a significant number of signaling pathways, the mTOR pathway can modulate the processes of post-ischemic inflammation and autophagy, both of which play an integral part in the pathophysiological cascade of stroke. Promoting or inhibiting such processes under ischemic conditions can lead to apoptosis or instead sustained viability of neurons. The purpose of this review is to examine the pathophysiological role of mTOR in acute ischemic stroke, while highlighting promising neuroprotective agents such as hamartin for therapeutic modulation of this pathway. The therapeutic potential of mTOR is also discussed, with emphasis on implicated molecules and pathway steps that warrant further elucidation in order for their neuroprotective properties to be efficiently tested in future clinical trials.
Collapse
Affiliation(s)
- Konstantinos Melanis
- Second Department of Neurology, School of Medicine and ‘Attikon’ University Hospital, National and Kapodistrian University of Athens, Rimini 1 Chaidari, Athens 12462, Greece
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maria-Ioanna Stefanou
- Second Department of Neurology, School of Medicine and ‘Attikon’ University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos M. Themistoklis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Neurosurgery, ‘Korgialenio, Benakio, H.R.C’. General Hospital of Athens, Athens, Greece
| | - Themistoklis Papasilekas
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Neurosurgery, ‘Korgialenio, Benakio, H.R.C’. General Hospital of Athens, Athens, Greece
| |
Collapse
|
34
|
Ge Y, Wang YT. GluN2B-containing NMDARs in the mammalian brain: pharmacology, physiology, and pathology. Front Mol Neurosci 2023; 16:1190324. [PMID: 37324591 PMCID: PMC10264587 DOI: 10.3389/fnmol.2023.1190324] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/24/2023] [Indexed: 06/17/2023] Open
Abstract
Glutamate N-methyl-D-aspartate receptor (NMDAR) is critical for promoting physiological synaptic plasticity and neuronal viability. As a major subpopulation of the NMDAR, the GluN2B subunit-containing NMDARs have distinct pharmacological properties, physiological functions, and pathological relevance to neurological diseases compared with other NMDAR subtypes. In mature neurons, GluN2B-containing NMDARs are likely expressed as both diheteromeric and triheteromeric receptors, though the functional importance of each subpopulation has yet to be disentangled. Moreover, the C-terminal region of the GluN2B subunit forms structural complexes with multiple intracellular signaling proteins. These protein complexes play critical roles in both activity-dependent synaptic plasticity and neuronal survival and death signaling, thus serving as the molecular substrates underlying multiple physiological functions. Accordingly, dysregulation of GluN2B-containing NMDARs and/or their downstream signaling pathways has been implicated in neurological diseases, and various strategies to reverse these deficits have been investigated. In this article, we provide an overview of GluN2B-containing NMDAR pharmacology and its key physiological functions, highlighting the importance of this receptor subtype during both health and disease states.
Collapse
Affiliation(s)
- Yang Ge
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yu Tian Wang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
35
|
Huang M, Bin NR, Rai J, Ma K, Chow CH, Eide S, Harada H, Xiao J, Feng D, Sun HS, Feng ZP, Gaisano HY, Pessin JE, Monnier PP, Okamoto K, Zhang L, Sugita S. Neuronal SNAP-23 is critical for synaptic plasticity and spatial memory independently of NMDA receptor regulation. iScience 2023; 26:106664. [PMID: 37168570 PMCID: PMC10165271 DOI: 10.1016/j.isci.2023.106664] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/30/2023] [Accepted: 04/06/2023] [Indexed: 05/13/2023] Open
Abstract
SNARE-mediated membrane fusion plays a crucial role in presynaptic vesicle exocytosis and also in postsynaptic receptor delivery. The latter is considered particularly important for synaptic plasticity and learning and memory, yet the identity of the key SNARE proteins remains elusive. Here, we investigate the role of neuronal synaptosomal-associated protein-23 (SNAP-23) by analyzing pyramidal-neuron specific SNAP-23 conditional knockout (cKO) mice. Electrophysiological analysis of SNAP-23 deficient neurons using acute hippocampal slices showed normal basal neurotransmission in CA3-CA1 synapses with unchanged AMPA and NMDA currents. Nevertheless, we found theta-burst stimulation-induced long-term potentiation (LTP) was vastly diminished in SNAP-23 cKO slices. Moreover, unlike syntaxin-4 cKO mice where both basal neurotransmission and LTP decrease manifested changes in a broad set of behavioral tasks, deficits of SNAP-23 cKO are more limited to spatial memory. Our data reveal that neuronal SNAP-23 is selectively crucial for synaptic plasticity and spatial memory without affecting basal glutamate receptor function.
Collapse
Affiliation(s)
- Mengjia Huang
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Na-Ryum Bin
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jayant Rai
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G1X5, Canada
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ke Ma
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Pediatrics, The First Hospital of Jilin University, Changchun 130021, China
| | - Chun Hin Chow
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sarah Eide
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Hidekiyo Harada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Jianbing Xiao
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Anatomy, Harbin Medical University, Harbin 150081, China
| | - Daorong Feng
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hong-Shuo Sun
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Anatomy, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Herbert Y. Gaisano
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jeffrey E. Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Philippe P. Monnier
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Donald K. Johnson Eye Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Ophthalmology & Vision Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kenichi Okamoto
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G1X5, Canada
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Liang Zhang
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shuzo Sugita
- Division of Experimental & Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
36
|
Zhao L, Mühleisen TW, Pelzer DI, Burger B, Beins EC, Forstner AJ, Herms S, Hoffmann P, Amunts K, Palomero-Gallagher N, Cichon S. Relationships between neurotransmitter receptor densities and expression levels of their corresponding genes in the human hippocampus. Neuroimage 2023; 273:120095. [PMID: 37030412 PMCID: PMC10167541 DOI: 10.1016/j.neuroimage.2023.120095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/02/2023] [Accepted: 04/05/2023] [Indexed: 04/08/2023] Open
Abstract
Neurotransmitter receptors are key molecules in signal transmission, their alterations are associated with brain dysfunction. Relationships between receptors and their corresponding genes are poorly understood, especially in humans. We combined in vitro receptor autoradiography and RNA sequencing to quantify, in the same tissue samples (7 subjects), the densities of 14 receptors and expression levels of their corresponding 43 genes in the Cornu Ammonis (CA) and dentate gyrus (DG) of human hippocampus. Significant differences in receptor densities between both structures were found only for metabotropic receptors, whereas significant differences in RNA expression levels mostly pertained ionotropic receptors. Receptor fingerprints of CA and DG differ in shapes but have similar sizes; the opposite holds true for their "RNA fingerprints", which represent the expression levels of multiple genes in a single area. In addition, the correlation coefficients between receptor densities and corresponding gene expression levels vary widely and the mean correlation strength was weak-to-moderate. Our results suggest that receptor densities are not only controlled by corresponding RNA expression levels, but also by multiple regionally specific post-translational factors.
Collapse
|
37
|
Zhang J, Liu Z, Liu X, Wang X, Yu L. Intravenous Injection of GluR2-3Y Inhibits Repeated Morphine-Primed Reinstatement of Drug Seeking in Rats. Brain Sci 2023; 13:brainsci13040590. [PMID: 37190555 DOI: 10.3390/brainsci13040590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Studies have demonstrated that the α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor is essential to drug addiction. In this study, we explored the influence of GluR2-3Y, an interfering peptide to prevent the endocytosis of AMPA receptors containing the GluR2 subunit, on morphine-seeking behavior in the rat self-administration model. After self-administration was established, the rats received intravenous injections of GluR2-3Y during the extinction sessions. There were no significant differences in both active and inactive pokes compared to the control group of rats that received GluR2-3S, indicating that GluR2-3Y has no significant influences on the extinction of morphine self-administration. The other two groups of rats were trained, extinguished, and reinstated by repeated morphine priming (respectively, called Prime 1, Prime 2, and Prime 3). Only one intravenous injection of GluR2-3Y was performed before Prime 1. Compared to the control group, GluR2-3Y did not affect Prime 1, but significantly attenuated the morphine-seeking behavior during repeated morphine-primed reinstatement, indicating an inhibitory after effect of GluR2-3Y on morphine-seeking behavior in rats. The long-term depression (LTD) in the nucleus accumbens (NAc) shell was also assessed. Pretreatment with GluR2-3Y altered the ability of LTD induction to the level of that in the naive group, while pretreatment with GluR2-3S had no effects on LTD. Our results demonstrated that the intravenous injection of GluR2-3Y, to block the endocytosis of AMPA receptors, inhibited the reinstatement of morphine-seeking behavior, which may be induced by modulating the neuronal plasticity in the NAc shell of rats.
Collapse
Affiliation(s)
- Jianjun Zhang
- College of Basic Medical, Shanxi University of Chinese Medicine, Jinzhong 030619, China
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong 030619, China
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China
| | - Zhuo Liu
- School of Crime Investigation, People’s Public Security University of China, Beijing 100038, China
| | - Xiaodong Liu
- Beijing Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoqian Wang
- College of Basic Medical, Shanxi University of Chinese Medicine, Jinzhong 030619, China
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong 030619, China
| | - Longchuan Yu
- School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
38
|
Damiani F, Cornuti S, Tognini P. The gut-brain connection: Exploring the influence of the gut microbiota on neuroplasticity and neurodevelopmental disorders. Neuropharmacology 2023; 231:109491. [PMID: 36924923 DOI: 10.1016/j.neuropharm.2023.109491] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/22/2023] [Accepted: 03/05/2023] [Indexed: 03/17/2023]
Abstract
Neuroplasticity refers to the ability of brain circuits to reorganize and change the properties of the network, resulting in alterations in brain function and behavior. It is traditionally believed that neuroplasticity is influenced by external stimuli, learning, and experience. Intriguingly, there is new evidence suggesting that endogenous signals from the body's periphery may play a role. The gut microbiota, a diverse community of microorganisms living in harmony with their host, may be able to influence plasticity through its modulation of the gut-brain axis. Interestingly, the maturation of the gut microbiota coincides with critical periods of neurodevelopment, during which neural circuits are highly plastic and potentially vulnerable. As such, dysbiosis (an imbalance in the gut microbiota composition) during early life may contribute to the disruption of normal developmental trajectories, leading to neurodevelopmental disorders. This review aims to examine the ways in which the gut microbiota can affect neuroplasticity. It will also discuss recent research linking gastrointestinal issues and bacterial dysbiosis to various neurodevelopmental disorders and their potential impact on neurological outcomes.
Collapse
Affiliation(s)
| | - Sara Cornuti
- Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Paola Tognini
- Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.
| |
Collapse
|
39
|
Naylor DE. In the fast lane: Receptor trafficking during status epilepticus. Epilepsia Open 2023; 8 Suppl 1:S35-S65. [PMID: 36861477 PMCID: PMC10173858 DOI: 10.1002/epi4.12718] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Status epilepticus (SE) remains a significant cause of morbidity and mortality and often is refractory to standard first-line treatments. A rapid loss of synaptic inhibition and development of pharmacoresistance to benzodiazepines (BZDs) occurs early during SE, while NMDA and AMPA receptor antagonists remain effective treatments after BZDs have failed. Multimodal and subunit-selective receptor trafficking within minutes to an hour of SE involves GABA-A, NMDA, and AMPA receptors and contributes to shifts in the number and subunit composition of surface receptors with differential impacts on the physiology, pharmacology, and strength of GABAergic and glutamatergic currents at synaptic and extrasynaptic sites. During the first hour of SE, synaptic GABA-A receptors containing γ2 subunits move to the cell interior while extrasynaptic GABA-A receptors with δ subunits are preserved. Conversely, NMDA receptors containing N2B subunits are increased at synaptic and extrasynaptic sites, and homomeric GluA1 ("GluA2-lacking") calcium permeant AMPA receptor surface expression also is increased. Molecular mechanisms, largely driven by NMDA receptor or calcium permeant AMPA receptor activation early during circuit hyperactivity, regulate subunit-specific interactions with proteins involved with synaptic scaffolding, adaptin-AP2/clathrin-dependent endocytosis, endoplasmic reticulum (ER) retention, and endosomal recycling. Reviewed here is how SE-induced shifts in receptor subunit composition and surface representation increase the excitatory to inhibitory imbalance that sustains seizures and fuels excitotoxicity contributing to chronic sequela such as "spontaneous recurrent seizures" (SRS). A role for early multimodal therapy is suggested both for treatment of SE and for prevention of long-term comorbidities.
Collapse
Affiliation(s)
- David E Naylor
- VA Greater Los Angeles Healthcare System, Department of Neurology, David Geffen School of Medicine at UCLA, and The Lundquist Institute at Harbor-UCLA Medical Center, Los Angeles, California, USA
| |
Collapse
|
40
|
Montanez-Miranda C, Bramlett SN, Hepler JR. RGS14 expression in CA2 hippocampus, amygdala, and basal ganglia: Implications for human brain physiology and disease. Hippocampus 2023; 33:166-181. [PMID: 36541898 PMCID: PMC9974931 DOI: 10.1002/hipo.23492] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/09/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
RGS14 is a multifunctional scaffolding protein that is highly expressed within postsynaptic spines of pyramidal neurons in hippocampal area CA2. Known roles of RGS14 in CA2 include regulating G protein, H-Ras/ERK, and calcium signaling pathways to serve as a natural suppressor of synaptic plasticity and postsynaptic signaling. RGS14 also shows marked postsynaptic expression in major structures of the limbic system and basal ganglia, including the amygdala and both the ventral and dorsal subdivisions of the striatum. In this review, we discuss the signaling functions of RGS14 and its role in postsynaptic strength (long-term potentiation) and spine structural plasticity in CA2 hippocampal neurons, and how RGS14 suppression of plasticity impacts linked behaviors such as spatial learning, object memory, and fear conditioning. We also review RGS14 expression in the limbic system and basal ganglia and speculate on its possible roles in regulating plasticity in these regions, with a focus on behaviors related to emotion and motivation. Finally, we explore the functional implications of RGS14 in various brain circuits and speculate on its possible roles in certain disease states such as hippocampal seizures, addiction, and anxiety disorders.
Collapse
Affiliation(s)
| | | | - John R. Hepler
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322-3090
| |
Collapse
|
41
|
Non-competitive AMPA glutamate receptors antagonism by perampanel as a strategy to counteract hippocampal hyper-excitability and cognitive deficits in cerebral amyloidosis. Neuropharmacology 2023; 225:109373. [PMID: 36502868 DOI: 10.1016/j.neuropharm.2022.109373] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 11/26/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Pathological accumulation of Aβ oligomers has been linked to neuronal networks hyperexcitability, potentially underpinned by glutamatergic AMPA receptors (AMPARs) dysfunction. We aimed to investigate whether the non-competitive block of AMPARs was able to counteract the alteration of hippocampal epileptic threshold, and of synaptic plasticity linked to Aβ oligomers accumulation, being this glutamate receptor a valuable specific therapeutic target. In this work, we showed that the non-competitive AMPARs antagonist perampanel (PER) which, per se, did not affect physiological synaptic transmission, was able to counteract Aβ-induced hyperexcitability. Moreover, AMPAR antagonism was able to counteract Aβ-induced hippocampal LTP impairment and hippocampal-based cognitive deficits in Aβ oligomers-injected mice, while retaining antiseizure efficacy. Beside this, AMPAR antagonism was also able to reduce the increased expression of proinflammatory cytokines in this mice model, also suggesting the presence of an anti-inflammatory activity. Thus, targeting AMPARs might be a valuable strategy to reduce both hippocampal networks hyperexcitability and synaptic plasticity deficits induced by Aβ oligomers accumulation.
Collapse
|
42
|
NMDARs antagonist MK801 suppresses LPS-induced apoptosis and mitochondrial dysfunction by regulating subunits of NMDARs via the CaM/CaMKII/ERK pathway. Cell Death Discov 2023; 9:59. [PMID: 36774369 PMCID: PMC9922289 DOI: 10.1038/s41420-023-01362-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/13/2023] Open
Abstract
Lipopolysaccharide (LPS) displays a robust immunostimulatory ability upon Toll-like receptor 4 (TLR4) recognition. N-methyl-D-aspartate receptors (NMDARs) are highly compartmentalized in most cells and implicated in various inflammatory disorders. However, the relationship between TLR4 and NMDARs has not been explored deeply. This study aimed to examine the role of NMDARs and its specific inhibitor MK801 in LPS-treated endothelial cell dysfunction and the related mechanism in vivo and in vitro. The results showed that pre-treatment with MK801 significantly decreased LPS-induced cell death, cellular Ca2+, cellular reactive oxygen species, and glutamate efflux. Moreover, MK801 restrained LPS-induced mitochondrial dysfunction by regulating mitochondrial membrane potential and mitochondrial Ca2+ uptake. The oxygen consumption, basal and maximal respiration rate, and ATP production in LPS-treated HUVECs were reversed by MK801 via regulating ATP synthesis-related protein SDHB2, MTCO1, and ATP5A. The molecular pathway involved in MK801-regulated LPS injury was mediated by phosphorylation of CaMKII and ERK and the expression of MCU, MCUR1, and TLR4. LPS-decreased permeability in HUVECs was improved by MK801 via the Erk/ZO-1/occluding/Cx43 axis. Co-immunoprecipitation assay and western blotting showed three subtypes of NMDARs, NMDAζ1, NMDAε2, and NMDAε4 were bound explicitly to TLR4, suppressed by LPS, and promoted by MK801. Deficiency of NMDAζ1, NMDAε2, or NMDAε4 induced cell apoptosis, Ca2+ uptake, ROS production, and decreased basal and maximal respiration rate, and ATP production, suggesting that NMDARs integrity is vital for cell and mitochondrial function. In vivo investigation showed MK801 improved impairment of vascular permeability, especially in the lung and mesentery in LPS-injured mice. Our study displayed a novel mechanism and utilization of MK801 in LPS-induced ECs injury and permeability.
Collapse
|
43
|
G-protein coupled estrogen receptor (GPER1) activation promotes synaptic insertion of AMPA receptors and induction of chemical LTP at hippocampal temporoammonic-CA1 synapses. Mol Brain 2023; 16:16. [PMID: 36709268 PMCID: PMC9883958 DOI: 10.1186/s13041-023-01003-3] [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: 09/12/2022] [Accepted: 01/12/2023] [Indexed: 01/30/2023] Open
Abstract
It is well documented that 17β estradiol (E2) regulates excitatory synaptic transmission at hippocampal Shaffer-collateral (SC)-CA1 synapses, via activation of the classical estrogen receptors (ERα and ERβ). Hippocampal CA1 pyramidal neurons are also innervated by the temporoammonic (TA) pathway, and excitatory TA-CA1 synapses are reported to be regulated by E2. Recent studies suggest a role for the novel G-protein coupled estrogen receptor (GPER1) at SC-CA1 synapses, however, the role of GPER1 in mediating the effects of E2 at juvenile TA-CA1 synapses is unclear. Here we demonstrate that the GPER1 agonist, G1 induces a persistent, concentration-dependent (1-10 nM) increase in excitatory synaptic transmission at TA-CA1 synapses and this effect is blocked by selective GPER1 antagonists. The ability of GPER1 to induce this novel form of chemical long-term potentiation (cLTP) was prevented following blockade of N-methyl-D-aspartate (NMDA) receptors, and it was not accompanied by any change in paired pulse facilitation ratio (PPR). GPER1-induced cLTP involved activation of ERK but was independent of phosphoinositide 3-kinase (PI3K) signalling. Prior treatment with philanthotoxin prevented the effects of G1, indicating that synaptic insertion of GluA2-lacking α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors underlies GPER1-induced cLTP. Furthermore, activity-dependent LTP occluded G1-induced cLTP and vice versa, indicating that these processes have overlapping expression mechanisms. Activity-dependent LTP was blocked by the GPER1 antagonist, G15, suggesting that GPER1 plays a role in NMDA-dependent LTP at juvenile TA-CA1 synapses. These findings add a new dimension to our understanding of GPER1 in modulating neuronal plasticity with relevance to age-related neurodegenerative conditions.
Collapse
|
44
|
Aldossary HS, Nieves DJ, Kavanagh DM, Owen D, Ray CJ, Kumar P, Coney AM, Holmes AP. Analyzing Angiotensin II Receptor Type 1 Clustering in PC12 Cells in Response to Hypoxia Using Direct Stochastic Optical Reconstruction Microscopy (dSTORM). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1427:175-184. [PMID: 37322348 DOI: 10.1007/978-3-031-32371-3_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Angiotensin II (Ang II) is a hormone that plays a major role in maintaining homeostasis. The Ang II receptor type 1 (AT1R) is expressed in acute O2 sensitive cells, including carotid body (CB) type I cells and pheochromocytoma 12 (PC12) cells, and Ang II increases cell activity. While a functional role for Ang II and AT1Rs in increasing the activity of O2 sensitive cells has been established, the nanoscale distribution of AT1Rs has not. Furthermore, it is not known how exposure to hypoxia may alter the single-molecule arrangement and clustering of AT1Rs. In this study, the AT1R nanoscale distribution under control normoxic conditions in PC12 cells was determined using direct stochastic optical reconstruction microscopy (dSTORM). AT1Rs were arranged in distinct clusters with measurable parameters. Across the entire cell surface there averaged approximately 3 AT1R clusters/μm2 of cell membrane. Cluster area varied in size ranging from 1.1 × 10-4 to 3.9 × 10-2 μm2. Twenty-four hours of exposure to hypoxia (1% O2) altered clustering of AT1Rs, with notable increases in the maximum cluster area, suggestive of an increase in supercluster formation. These observations could aid in understanding mechanisms underlying augmented Ang II sensitivity in O2 sensitive cells in response to sustained hypoxia.
Collapse
Affiliation(s)
- Hayyaf S Aldossary
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK.
- College of Medicine, Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | - Daniel J Nieves
- Institute of Immunology and Immunotherapy and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | | | - Dylan Owen
- School of Mathematics, University of Birmingham, Birmingham, UK
| | - Clare J Ray
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
| | - Prem Kumar
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
| | - Andrew M Coney
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
| | - Andrew P Holmes
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
45
|
Maternal Hyperhomocysteinemia Produces Memory Deficits Associated with Impairment of Long-Term Synaptic Plasticity in Young Rats. Cells 2022; 12:cells12010058. [PMID: 36611852 PMCID: PMC9818716 DOI: 10.3390/cells12010058] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
Maternal hyperhomocysteinemia (HCY) is a common pregnancy complication caused by high levels of the homocysteine in maternal and fetal blood, which leads to the alterations of the cognitive functions, including learning and memory. In the present study, we investigated the mechanisms of these alterations in a rat model of maternal HCY. The behavioral tests confirmed the memory impairments in young and adult rats following the prenatal HCY exposure. Field potential recordings in hippocampal slices demonstrated that the long-term potentiation (LTP) was significantly reduced in HCY rats. The whole-cell patch-clamp recordings in hippocampal slices demonstrated that the magnitude of NMDA receptor-mediated currents did not change while their desensitization decreased in HCY rats. No significant alterations of glutamate receptor subunit expression except GluN1 were detected in the hippocampus of HCY rats using the quantitative real-time PCR and Western blot methods. The immunofluorescence microscopy revealed that the number of synaptopodin-positive spines is reduced, while the analysis of the ultrastructure of hippocampus using the electron microscopy revealed the indications of delayed hippocampal maturation in young HCY rats. Thus, the obtained results suggest that maternal HCY disturbs the maturation of hippocampus during the first month of life, which disrupts LTP formation and causes memory impairments.
Collapse
|
46
|
Kim SS, Kim JL, Hwang KS, Park HC, Bae MA, Kim KT, Cho SH. Mechanism of action and neurotoxic effects of chronic exposure to bisphenol F in adult zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 851:158258. [PMID: 36030852 DOI: 10.1016/j.scitotenv.2022.158258] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 06/15/2023]
Abstract
Although bisphenol F (BPF), the main replacement for bisphenol A, has been commonly used in polycarbonate production, its neurotoxicity and the underlying mechanisms remain poorly understood. To address this knowledge gap, this study aimed to assess the neurotoxicity caused by chronic exposure to BPF and to identify its underlying mechanisms. We exposed adult zebrafish chronically to BPF at environmentally relevant concentrations (0.001, 0.01, and 0.1 mg/L) for 4 weeks. The results revealed that with BPF crossing the blood-brain barrier and bioaccumulating in brain tissues, chronic exposure to BPF resulted in anxiety-like behaviors and disruptions in learning and memory function in adult zebrafish. Furthermore, BPF toxicity in the zebrafish brain involved the dysregulation of metabolic pathways for choline and kynurenine in neurotransmitter systems and for 17β-estradiol, cortisol, pregnenolone-sulfate, and Dehydroepiandrosterone (DHEA)-sulfate in neurosteroid systems. RNA-seq analysis revealed that BPF exposure affected metabolic pathways, calcium signaling pathways, neuroactive ligand-receptor interactions, tight junctions, gap junctions, and the gonadotropin-releasing hormone signaling pathway. Our results indicate that chronic exposure to BPF alters the neurochemical profile of the brain and causes neurobehavioral effects, such as anxiety and cognitive decline. Overall, the multimodal approach, including behavioral and neurochemical profiling technologies, has great potential for the comprehensive assessment of potential risks posed by environmental pollutants to human and ecosystem health.
Collapse
Affiliation(s)
- Seong Soon Kim
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Jiwon L Kim
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Kyu-Seok Hwang
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University, Ansan, Gyeonggido 425-707, Republic of Korea
| | - Myung Ae Bae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, University of Science & Technology, Daejeon 34113, Republic of Korea.
| | - Ki-Tae Kim
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea.
| | - Sung-Hee Cho
- Chemical Analysis Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
| |
Collapse
|
47
|
Prolonged contextual fear memory in AMPA receptor palmitoylation-deficient mice. Neuropsychopharmacology 2022; 47:2150-2159. [PMID: 35618841 PMCID: PMC9556755 DOI: 10.1038/s41386-022-01347-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/26/2022] [Accepted: 05/07/2022] [Indexed: 11/24/2022]
Abstract
Long-lasting fear-related disorders depend on the excessive retention of traumatic fear memory. We previously showed that the palmitoylation-dependent removal of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptors prevents hyperexcitation-based epileptic seizures and that AMPA receptor palmitoylation maintains neural network stability. In this study, AMPA receptor subunit GluA1 C-terminal palmitoylation-deficient (GluA1C811S) mice were subjected to comprehensive behavioral battery tests to further examine whether the mutation causes other neuropsychiatric disease-like symptoms. The behavioral analyses revealed that palmitoylation-deficiency in GluA1 is responsible for characteristic prolonged contextual fear memory formation, whereas GluA1C811S mice showed no impairment of anxiety-like behaviors at the basal state. In addition, fear generalization gradually increased in these mutant mice without affecting their cued fear. Furthermore, fear extinction training by repeated exposure of mice to conditioned stimuli had little effect on GluA1C811S mice, which is in line with augmentation of synaptic transmission in pyramidal neurons in the basolateral amygdala. In contrast, locomotion, sociability, depression-related behaviors, and spatial learning and memory were unaffected by the GluA1 non-palmitoylation mutation. These results indicate that impairment of AMPA receptor palmitoylation specifically causes posttraumatic stress disorder (PTSD)-like symptoms.
Collapse
|
48
|
Chan ES, Ge Y, So YW, Bai YF, Liu L, Wang YT. Allosteric potentiation of GABAA receptor single-channel conductance by netrin-1 during neuronal-excitation-induced inhibitory synaptic homeostasis. Cell Rep 2022; 41:111584. [DOI: 10.1016/j.celrep.2022.111584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/13/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
|
49
|
Hayashi T. Membrane lipid rafts are required for AMPA receptor tyrosine phosphorylation. Front Synaptic Neurosci 2022; 14:921772. [PMID: 36387774 PMCID: PMC9662747 DOI: 10.3389/fnsyn.2022.921772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 10/11/2022] [Indexed: 12/24/2023] Open
Abstract
Membrane lipid rafts are sphingolipids and cholesterol-enriched membrane microdomains, which form a center for the interaction or assembly of palmitoylated signaling molecules, including Src family non-receptor type protein tyrosine kinases. Lipid rafts abundantly exist in neurons and function in the maintenance of synapses. Excitatory synaptic strength is largely controlled by the surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptors in the mammalian brain. AMPA receptor endocytosis from the synaptic surface is regulated by phosphorylation of the GluA2 subunit at tyrosine 876 by Src family kinases. Here, I revealed that tyrosine phosphorylated GluA2 is concentrated in the lipid rafts fraction. Furthermore, stimulation-induced upregulation of GluA2 tyrosine phosphorylation is disrupted by the treatment of neurons with a cholesterol-depleting compound, filipin III. These results indicate the importance of lipid rafts as enzymatic reactive sites for AMPA receptor tyrosine phosphorylation and subsequent AMPA receptor internalization from the synaptic surface.
Collapse
Affiliation(s)
- Takashi Hayashi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
50
|
Jensen KL, Jensen SB, Madsen KL. A mechanistic overview of approaches for the treatment of psychostimulant dependence. Front Pharmacol 2022; 13:854176. [PMID: 36160447 PMCID: PMC9493975 DOI: 10.3389/fphar.2022.854176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Psychostimulant use disorder is a major health issue around the world with enormous individual, family-related and societal consequences, yet there are no effective pharmacological treatments available. In this review, a target-based overview of pharmacological treatments toward psychostimulant addiction will be presented. We will go through therapeutic approaches targeting different aspects of psychostimulant addiction with focus on three major areas; 1) drugs targeting signalling, and metabolism of the dopamine system, 2) drugs targeting either AMPA receptors or metabotropic glutamate receptors of the glutamate system and 3) drugs targeting the severe side-effects of quitting long-term psychostimulant use. For each of these major modes of intervention, findings from pre-clinical studies in rodents to clinical trials in humans will be listed, and future perspectives of the different treatment strategies as well as their potential side-effects will be discussed. Pharmaceuticals modulating the dopamine system, such as antipsychotics, DAT-inhibitors, and disulfiram, have shown some promising results. Cognitive enhancers have been found to increase aspects of behavioural control, and drugs targeting the glutamate system such as modulators of metabotropic glutamate receptors and AMPA receptors have provided interesting changes in relapse behaviour. Furthermore, CRF-antagonists directed toward alleviating the symptoms of the withdrawal stage have been examined with interesting resulting changes in behaviour. There are promising results investigating therapeutics for psychostimulant addiction, but further preclinical work and additional human studies with a more stratified patient selection are needed to prove sufficient evidence of efficacy and tolerability.
Collapse
|