1
|
Tseng KY, Molla HM. Cannabinoid CB1 receptor-sensitive neurodevelopmental processes and trajectories. Mol Psychiatry 2025:10.1038/s41380-025-03057-2. [PMID: 40389627 DOI: 10.1038/s41380-025-03057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 05/12/2025] [Indexed: 05/21/2025]
Abstract
As high-potency cannabis (with high Δ9-Tetrahydrocannabinol content) becomes easily accessible and widespread, it is of extreme importance for public health that a scientific platform is used to implement practical guidelines, particularly for at-risk populations. Many reviews have been written in the past decade summarizing the impact of cannabis in the developing brain. One critical concept frequently mentioned but not discussed in detail is whether there are sensitive neurodevelopmental events driving the age-specific sensitivity to cannabis, particularly those mediated by cannabinoid type 1 receptor signaling. By integrating available data from humans and animal models, the goal of the present expert review article is to provide a mechanistic overview on how cannabis exposure during sensitive periods of neural circuit plasticity and development can result in lasting consequences. Here we used the frontal cortex as a proxy to align the trajectory of the brain cannabinoid system between humans and rodents. Both the strengths and limitations of available mechanistic studies on the effects of cannabis and cannabinoids were discussed using a developmental framework from which neural circuit adaptations during sensitive periods are considered. Such an approach is needed to align key neurodevelopmental variables through the lifespan, which in turn will provide valuable insights applicable to the human brain by defining the underpinning mechanisms of sensitive periods and how the impact of cannabis changes from childhood to adolescence, and thereafter through young adulthood.
Collapse
Affiliation(s)
- Kuei Y Tseng
- Department of Anatomy & Cell Biology, University of Illinois at Chicago - College of Medicine, Chicago, IL, USA.
| | - Hanna M Molla
- Department of Psychiatry & Behavioral Neuroscience, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
2
|
Sullivan KR, Ravens A, Walker AC, Shepherd JD. "Arc - A viral vector of memory and synaptic plasticity". Curr Opin Neurobiol 2025; 91:102979. [PMID: 39956025 PMCID: PMC11938376 DOI: 10.1016/j.conb.2025.102979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 02/18/2025]
Abstract
Learning induces gene expression and memory consolidation requires new protein synthesis. Many of these activity-induced genes are transcription factors. One of the exceptions is a key immediate early gene, Arc, which has been implicated in several forms of synaptic plasticity and is critical for long-term memory formation. Recently, Arc was discovered to have retroviral properties, such as the ability to form virus-like capsids, that were repurposed from an ancient retrotransposon. Arc capsids are released in extracellular vesicles that mediate intercellular communication. Here, we review Arc's role in synaptic plasticity and propose a model for how Arc mediates memory consolidation via a novel intercellular non-cell autonomous form of long-term depression.
Collapse
Affiliation(s)
| | - Alicia Ravens
- Department of Neurobiology, University of Utah, United States
| | - Alicia C Walker
- Department of Neurobiology, University of Utah, United States
| | | |
Collapse
|
3
|
Petroccione MA, Melone M, Rathwell TJ, Dwivedi N, Grienberger C, Conti F, Scimemi A. An unsuspected physiological role for mGluRIII glutamate receptors in hippocampal area CA1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646479. [PMID: 40236245 PMCID: PMC11996470 DOI: 10.1101/2025.03.31.646479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Group III metabotropic glutamate receptors (mGluRIII) are expressed broadly throughout the neocortex and hippocampus but are thought to inhibit neurotransmitter release only at a subset of synapses and in a target cell- specific manner. Accordingly, previous slice physiology experiments in hippocampal area CA1 showed that mGluRIII receptors inhibit glutamate and GABA release only at excitatory and inhibitory synapses formed onto GABAergic interneurons, not onto pyramidal cells. Here, we show that the supposed target cell-specific modulation of GABA release only occurs when the extracellular calcium concentration in the recording solution is higher than its physiological concentration in the cerebrospinal fluid. Under more physiological conditions, mGluRIII receptors inhibit GABA release at synapses formed onto both interneurons and pyramidal cells but limit glutamate release only onto interneurons. This previously unrecognized form of mGluRIII-dependent, pre-synaptic modulation of inhibition onto pyramidal cells is accounted for by a reduction in the size of the readily releasable pool, mediated by protein kinase A and its vesicle-associated target proteins, synapsins. Using in vivo whole-cell recordings in behaving mice, we demonstrate that blocking mGluRIII activation in the intact CA1 network results in net effects consistent with decreased inhibition and significantly alters CA1 place cell activity. Together, these findings challenge our current understanding of the role of mGluRIII receptors in the control of synaptic transmission and encoding of spatial information in the hippocampus.
Collapse
|
4
|
From M, Crosby KM. Endocannabinoid and nitric oxide interactions in the brain. Neuroscience 2025; 569:267-276. [PMID: 39909337 DOI: 10.1016/j.neuroscience.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/16/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Endogenous cannabinoids (eCBs) and nitric oxide (NO) are classical retrograde transmitters that modulate synaptic function throughout the brain. Although much is known about how these signals individually control synaptic activity and behavior, accumulating evidence suggests that they can also interact in a multitude of ways in the brain and beyond. Here, we present evidence for interactions between endogenous cannabinoids and nitric oxide in the brain. Specifically, we describe the effects of eCBs on NO synthesis and downstream signaling and in turn, we discuss how NO alters eCB levels and signaling pathways. We also provide an overview on how these transmitters work together or in opposition at the same synapses. This information will further our understanding of how two important, ubiquitous signals interact in the brain to ultimately affect neural function and behavior. Because eCBs and NO are involved in many physiological and pathological phenomena, understanding how these transmitters interact in non-human animals could lead to important therapeutic interventions in humans that potentially target both systems.
Collapse
Affiliation(s)
- Mary From
- Biology Department, Mount Allison University, 63B York Street, Sackville, NB E4L1G7, Canada
| | - Karen M Crosby
- Biology Department, Mount Allison University, 63B York Street, Sackville, NB E4L1G7, Canada.
| |
Collapse
|
5
|
Ancatén-González C, Meza RC, Gonzalez-Sanabria N, Segura I, Alcaino A, Peña-Pichicoi A, Latorre R, Chiu CQ, Chávez AE. BK channels mediate a presynaptic form of mGluR-LTD in the neonatal hippocampus. Proc Natl Acad Sci U S A 2025; 122:e2411506122. [PMID: 39773031 PMCID: PMC11745352 DOI: 10.1073/pnas.2411506122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
BK channels can control neuronal function, but their functional relevance in activity-dependent changes of synaptic function remains elusive. Here, we report that repetitive low-frequency stimulation activates BK channels through 12(S)HPETE, an arachidonic acid metabolite, produced downstream of postsynaptic metabotropic glutamate receptors (mGluRs) to trigger long-term depression (LTD) at CA3-CA1 synapses in hippocampal slices from P7-P10 mice. Activation of BK channels is subunit specific, as paxilline but not iberiotoxin blocked mGluR-LTD. Also, 12(S)HPETE does not change the electrophysiological properties of the BK channel when the BKα subunit is expressed alone but increases the channel open probability when the BKα is coexpressed with the β4-subunit. Our findings reveal an interaction between 12(S)HPETE and BK channels to regulate synaptic strength at central synapses and increase our understanding of the mechanisms underlying mGluR-LTD in the neonatal hippocampus that likely contribute to circuit maturation necessary for learning.
Collapse
Affiliation(s)
- Carlos Ancatén-González
- Programa de Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Rodrigo C. Meza
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Naileth Gonzalez-Sanabria
- Programa de Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Ignacio Segura
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Alejandro Alcaino
- Programa de Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Antonio Peña-Pichicoi
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Ramón Latorre
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Chiayu Q. Chiu
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Andrés E. Chávez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| |
Collapse
|
6
|
Michetti C, Benfenati F. Homeostatic regulation of brain activity: from endogenous mechanisms to homeostatic nanomachines. Am J Physiol Cell Physiol 2024; 327:C1384-C1399. [PMID: 39401424 DOI: 10.1152/ajpcell.00470.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 11/12/2024]
Abstract
After the initial concepts of the constancy of the internal milieu or homeostasis, put forward by Claude Bernard and Walter Cannon, homeostasis emerged as a mechanism to control oscillations of biologically meaningful variables within narrow physiological ranges. This is a primary need in the central nervous system that is continuously subjected to a multitude of stimuli from the internal and external environments that affect its function and structure, allowing to adapt the individual to the ever-changing daily conditions. Preserving physiological levels of activity despite disturbances that could either depress neural computation or excessively stimulate neural activity is fundamental, and failure of these homeostatic mechanisms can lead to brain diseases. In this review, we cover the role and main mechanisms of homeostatic plasticity involving the regulation of excitability and synaptic strength from the single neuron to the network level. We analyze the relationships between homeostatic and Hebbian plasticity and the conditions under which the preservation of the excitatory/inhibitory balance fails, triggering epileptogenesis and eventually epilepsy. Several therapeutic strategies to cure epilepsy have been designed to strengthen homeostasis when endogenous homeostatic plasticity mechanisms have become insufficient or ineffective to contrast hyperactivity. We describe "on demand" gene therapy strategies, including optogenetics, chemogenetics, and chemo-optogenetics, and particularly focus on new closed loop sensor-actuator strategies mimicking homeostatic plasticity that can be endogenously expressed to strengthen the homeostatic defenses against brain diseases.
Collapse
Affiliation(s)
- Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
7
|
Yan T, Sun J, Zhang Y, Wen C, Yang J. Enteromorpha prolifera Polysaccharide Alleviates Acute Alcoholic Liver Injury in C57 BL/6 Mice through the Gut-Liver Axis and NF-κB Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23258-23270. [PMID: 39404145 DOI: 10.1021/acs.jafc.4c05262] [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: 10/25/2024]
Abstract
Enteromorpha prolifera polysaccharide (EP2) protection against acute alcoholic liver injury (AALI) in mice was investigated. By integration of physiological indicators, gut microbiota, and short-chain fatty acids (SCFAs), the mechanism of EP2 in alleviating AALI was disclosed. The results showed that EP2 significantly ameliorated alcohol-induced abnormal transaminase activities, liver and intestinal systemic inflammation, and intestinal environmental disorders. EP2 significantly reduces liver and serum LPS contents by 1.69-fold and 1.54-fold. Furthermore, inhibition of the NF-κB signaling pathway by EP2 reduced the production of proinflammatory cytokines such as TNF-α (1.83-fold), IL-6 (11.09-fold), and IL-1β (1.99-fold). EP2 restored SCFAs to normal levels by upregulating the abundance of beneficial bacteria (Colidextribacter, Ruminococcus, unclassified_Lachnospiraceae, and Akkermansia). The alleviation of AALI by EP2 occurs through protection of the intestinal mucosal barrier and reduction of LPS permeating in serum. The decrease in LPS inactivates the NF-κB signaling pathway and prevents inflammation. In short, EP2 regulates the gut-liver axis and inflammation, alleviating effects in AALI mice.
Collapse
Affiliation(s)
- Tingting Yan
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Jinghe Sun
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Yuying Zhang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Chengrong Wen
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Jingfeng Yang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
8
|
Kim HH, Jeong SH, Park MY, Bhosale PB, Abusaliya A, Lee SJ, Heo JD, Kim HW, Seong JK, Kim DI, Park KI, Kim GS. Binding affinity screening of polyphenolic compounds in Stachys affinis extract (SAE) for their potential antioxidant and anti-inflammatory effects. Sci Rep 2024; 14:18095. [PMID: 39103443 PMCID: PMC11300793 DOI: 10.1038/s41598-024-68880-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024] Open
Abstract
Free radical is a marker in various inflammatory diseases. The antioxidant effect protects us from this damage, which also plays an essential role in preventing inflammation. Inflammation protects the body from biological stimuli, and pro-inflammatory mediators are negatively affected in the immune system. Inflammation caused by LPS is an endotoxin found in the outer membrane of Gram-negative bacteria, which induces immune cells to produce inflammatory cytokines such as cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase. Based on this, the antioxidant and anti-inflammatory effects of plant extracts were investigated. First, the main phenolic compounds for the five peaks obtained from Stachys affinis extract (SAE) were identified. The antioxidant effect of each phenolic compound was confirmed through HPLC analysis before and after the competitive binding reaction between DPPH and the extract. Afterward, the anti-inflammatory effect of each phenolic compound was confirmed through competitive binding between COX2 and the extract in HPLC analysis. Lastly, the anti-inflammatory effect of SAE was confirmed through in vitro experiments and also confirmed in terms of structural binding through molecular docking. This study confirmed that phenolic compounds in SAE extract have potential antioxidant and anti-inflammatory effects, and may provide information for primary screening of medicinal plants.
Collapse
Affiliation(s)
- Hun Hwan Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Se Hyo Jeong
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Min Yeong Park
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Pritam Bhangwan Bhosale
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Abuyaseer Abusaliya
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Sang Joon Lee
- Gyeongnam Department of Environment Toxicology and Chemistry, Biological Resources Research Group, Korea Institute of Toxicology, 17 Jegok-gil, Jinju, 52834, Korea
| | - Jeong Doo Heo
- Gyeongnam Department of Environment Toxicology and Chemistry, Biological Resources Research Group, Korea Institute of Toxicology, 17 Jegok-gil, Jinju, 52834, Korea
| | - Hyun Wook Kim
- Division of Animal Bioscience and Intergrated Biotechnology, Jinju, 52725, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dong Il Kim
- Namhae Garlic Research Institute, 2465-8 Namhaedaero, Namhae, Gyeongsangnam-do, 52430, Republic of Korea
| | - Kwang Il Park
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Gon Sup Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
9
|
Nasrallah K, Berthoux C, Hashimotodani Y, Chávez AE, Gulfo MC, Luján R, Castillo PE. Retrograde adenosine/A 2A receptor signaling facilitates excitatory synaptic transmission and seizures. Cell Rep 2024; 43:114382. [PMID: 38905101 PMCID: PMC11286346 DOI: 10.1016/j.celrep.2024.114382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/12/2023] [Accepted: 06/03/2024] [Indexed: 06/23/2024] Open
Abstract
Retrograde signaling at the synapse is a fundamental way by which neurons communicate and neuronal circuit function is fine-tuned upon activity. While long-term changes in neurotransmitter release commonly rely on retrograde signaling, the mechanisms remain poorly understood. Here, we identified adenosine/A2A receptor (A2AR) as a retrograde signaling pathway underlying presynaptic long-term potentiation (LTP) at a hippocampal excitatory circuit critically involved in memory and epilepsy. Transient burst activity of a single dentate granule cell induced LTP of mossy cell synaptic inputs, a BDNF/TrkB-dependent form of plasticity that facilitates seizures. Postsynaptic TrkB activation released adenosine from granule cells, uncovering a non-conventional BDNF/TrkB signaling mechanism. Moreover, presynaptic A2ARs were necessary and sufficient for LTP. Lastly, seizure induction released adenosine in a TrkB-dependent manner, while removing A2ARs or TrkB from the dentate gyrus had anti-convulsant effects. By mediating presynaptic LTP, adenosine/A2AR retrograde signaling may modulate dentate gyrus-dependent learning and promote epileptic activity.
Collapse
Affiliation(s)
- Kaoutsar Nasrallah
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Coralie Berthoux
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yuki Hashimotodani
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andrés E Chávez
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Michelle C Gulfo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rafael Luján
- Instituto de Biomedicina de la UCLM (IB-UCLM), Facultad de Medicina, Universidad Castilla-La Mancha, 02008 Albacete, Spain
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Psychiatry & Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
10
|
Rangel Guerrero DK, Balueva K, Barayeu U, Baracskay P, Gridchyn I, Nardin M, Roth CN, Wulff P, Csicsvari J. Hippocampal cholecystokinin-expressing interneurons regulate temporal coding and contextual learning. Neuron 2024; 112:2045-2061.e10. [PMID: 38636524 DOI: 10.1016/j.neuron.2024.03.019] [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: 02/24/2023] [Revised: 10/03/2023] [Accepted: 03/18/2024] [Indexed: 04/20/2024]
Abstract
Cholecystokinin-expressing interneurons (CCKIs) are hypothesized to shape pyramidal cell-firing patterns and regulate network oscillations and related network state transitions. To directly probe their role in the CA1 region, we silenced their activity using optogenetic and chemogenetic tools in mice. Opto-tagged CCKIs revealed a heterogeneous population, and their optogenetic silencing triggered wide disinhibitory network changes affecting both pyramidal cells and other interneurons. CCKI silencing enhanced pyramidal cell burst firing and altered the temporal coding of place cells: theta phase precession was disrupted, whereas sequence reactivation was enhanced. Chemogenetic CCKI silencing did not alter the acquisition of spatial reference memories on the Morris water maze but enhanced the recall of contextual fear memories and enabled selective recall when similar environments were tested. This work suggests the key involvement of CCKIs in the control of place-cell temporal coding and the formation of contextual memories.
Collapse
Affiliation(s)
- Dámaris K Rangel Guerrero
- Information and Systems Sciences, Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria.
| | - Kira Balueva
- Institute of Physiology, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
| | - Uladzislau Barayeu
- Information and Systems Sciences, Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Peter Baracskay
- Information and Systems Sciences, Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Igor Gridchyn
- Information and Systems Sciences, Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Michele Nardin
- Information and Systems Sciences, Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Chiara Nina Roth
- Information and Systems Sciences, Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Peer Wulff
- Institute of Physiology, Christian-Albrechts-University Kiel, 24118 Kiel, Germany.
| | - Jozsef Csicsvari
- Information and Systems Sciences, Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria.
| |
Collapse
|
11
|
Zhang H, Lei M, Zhang Y, Li H, He Z, Xie S, Zhu L, Wang S, Liu J, Li Y, Lu Y, Ma C. Phosphorylation of Doc2 by EphB2 modulates Munc13-mediated SNARE complex assembly and neurotransmitter release. SCIENCE ADVANCES 2024; 10:eadi7024. [PMID: 38758791 PMCID: PMC11100570 DOI: 10.1126/sciadv.adi7024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 04/12/2024] [Indexed: 05/19/2024]
Abstract
At the synapse, presynaptic neurotransmitter release is tightly controlled by release machinery, involving the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins and Munc13. The Ca2+ sensor Doc2 cooperates with Munc13 to regulate neurotransmitter release, but the underlying mechanisms remain unclear. In our study, we have characterized the binding mode between Doc2 and Munc13 and found that Doc2 originally occludes Munc13 to inhibit SNARE complex assembly. Moreover, our investigation unveiled that EphB2, a presynaptic adhesion molecule (SAM) with inherent tyrosine kinase functionality, exhibits the capacity to phosphorylate Doc2. This phosphorylation attenuates Doc2 block on Munc13 to promote SNARE complex assembly, which functionally induces spontaneous release and synaptic augmentation. Consistently, application of a Doc2 peptide that interrupts Doc2-Munc13 interplay impairs excitatory synaptic transmission and leads to dysfunction in spatial learning and memory. These data provide evidence that SAMs modulate neurotransmitter release by controlling SNARE complex assembly.
Collapse
Affiliation(s)
- Hong Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Mengshi Lei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Yu Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Hao Li
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhen He
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Sheng Xie
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Le Zhu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Jianfeng Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Youming Lu
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
12
|
Mellott JG, Duncan S, Busby J, Almassri LS, Wawrzyniak A, Iafrate MC, Ohl AP, Slabinski EA, Beaver AM, Albaba D, Vega B, Mafi AM, Buerke M, Tokar NJ, Young JW. Age-related upregulation of dense core vesicles in the central inferior colliculus. Front Cell Neurosci 2024; 18:1396387. [PMID: 38774486 PMCID: PMC11107844 DOI: 10.3389/fncel.2024.1396387] [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: 03/05/2024] [Accepted: 04/11/2024] [Indexed: 05/24/2024] Open
Abstract
Presbycusis is one of the most prevalent disabilities in aged populations of industrialized countries. As we age less excitation reaches the central auditory system from the periphery. To compensate, the central auditory system [e.g., the inferior colliculus (IC)], downregulates GABAergic inhibition to maintain homeostatic balance. However, the continued downregulation of GABA in the IC causes a disruption in temporal precision related to presbycusis. Many studies of age-related changes to neurotransmission in the IC have therefore focused on GABAergic systems. However, we have discovered that dense core vesicles (DCVs) are significantly upregulated with age in the IC. DCVs can carry neuropeptides, co-transmitters, neurotrophic factors, and proteins destined for the presynaptic zone to participate in synaptogenesis. We used immuno transmission electron microscopy across four age groups (3-month; 19-month; 24-month; and 28-month) of Fisher Brown Norway rats to examine the ultrastructure of DCVs in the IC. Tissue was stained post-embedding for GABA immunoreactivity. DCVs were characterized by diameter and by the neurochemical profile (GABAergic/non-GABAergic) of their location (bouton, axon, soma, and dendrite). Our data was collected across the dorsolateral to ventromedial axis of the central IC. After quantification, we had three primary findings. First, the age-related increase of DCVs occurred most robustly in non-GABAergic dendrites in the middle and low frequency regions of the central IC during middle age. Second, the likelihood of a bouton having more than one DCV increased with age. Lastly, although there was an age-related loss of terminals throughout the IC, the proportion of terminals that contained at least one DCV did not decline. We interpret this finding to mean that terminals carrying proteins packaged in DCVs are spared with age. Several recent studies have demonstrated a role for neuropeptides in the IC in defining cell types and regulating inhibitory and excitatory neurotransmission. Given the age-related increase of DCVs in the IC, it will be critical that future studies determine whether (1) specific neuropeptides are altered with age in the IC and (2) if these neuropeptides contribute to the loss of inhibition and/or increase of excitability that occurs during presbycusis and tinnitus.
Collapse
Affiliation(s)
- Jeffrey G. Mellott
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
- University Hospitals Hearing Research Center, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Syllissa Duncan
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Justine Busby
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Laila S. Almassri
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
- University Hospitals Hearing Research Center, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Alexa Wawrzyniak
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Milena C. Iafrate
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Andrew P. Ohl
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Elizabeth A. Slabinski
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Abigail M. Beaver
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Diana Albaba
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Brenda Vega
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Amir M. Mafi
- The Ohio State University College of Medicine, Columbus, OH, United States
| | - Morgan Buerke
- Department of Psychology, Louisiana State University, Baton Rouge, LA, United States
| | - Nick J. Tokar
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Jesse W. Young
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| |
Collapse
|
13
|
Barlow BSM, Longtin A, Joós B. Impact on backpropagation of the spatial heterogeneity of sodium channel kinetics in the axon initial segment. PLoS Comput Biol 2024; 20:e1011846. [PMID: 38489374 PMCID: PMC10942053 DOI: 10.1371/journal.pcbi.1011846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/21/2024] [Indexed: 03/17/2024] Open
Abstract
In a variety of neurons, action potentials (APs) initiate at the proximal axon, within a region called the axon initial segment (AIS), which has a high density of voltage-gated sodium channels (NaVs) on its membrane. In pyramidal neurons, the proximal AIS has been reported to exhibit a higher proportion of NaVs with gating properties that are "right-shifted" to more depolarized voltages, compared to the distal AIS. Further, recent experiments have revealed that as neurons develop, the spatial distribution of NaV subtypes along the AIS can change substantially, suggesting that neurons tune their excitability by modifying said distribution. When neurons are stimulated axonally, computational modelling has shown that this spatial separation of gating properties in the AIS enhances the backpropagation of APs into the dendrites. In contrast, in the more natural scenario of somatic stimulation, our simulations show that the same distribution can impede backpropagation, suggesting that the choice of orthodromic versus antidromic stimulation can bias or even invert experimental findings regarding the role of NaV subtypes in the AIS. We implemented a range of hypothetical NaV distributions in the AIS of three multicompartmental pyramidal cell models and investigated the precise kinetic mechanisms underlying such effects, as the spatial distribution of NaV subtypes is varied. With axonal stimulation, proximal NaV availability dominates, such that concentrating right-shifted NaVs in the proximal AIS promotes backpropagation. However, with somatic stimulation, the models are insensitive to availability kinetics. Instead, the higher activation threshold of right-shifted NaVs in the AIS impedes backpropagation. Therefore, recently observed developmental changes to the spatial separation and relative proportions of NaV1.2 and NaV1.6 in the AIS differentially impact activation and availability. The observed effects on backpropagation, and potentially learning via its putative role in synaptic plasticity (e.g. through spike-timing-dependent plasticity), are opposite for orthodromic versus antidromic stimulation, which should inform hypotheses about the impact of the developmentally regulated subcellular localization of these NaV subtypes.
Collapse
Affiliation(s)
- Benjamin S. M. Barlow
- Department of Physics, University of Ottawa, STEM Complex, 150 Louis-Pasteur Pvt, Ottawa, Ontario, Canada
| | - André Longtin
- Department of Physics, University of Ottawa, STEM Complex, 150 Louis-Pasteur Pvt, Ottawa, Ontario, Canada
- Center for Neural Dynamics and AI, University of Ottawa, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Béla Joós
- Department of Physics, University of Ottawa, STEM Complex, 150 Louis-Pasteur Pvt, Ottawa, Ontario, Canada
- Center for Neural Dynamics and AI, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
14
|
Chapp AD, Shan Z, Chen QH. Acetic Acid: An Underestimated Metabolite in Ethanol-Induced Changes in Regulating Cardiovascular Function. Antioxidants (Basel) 2024; 13:139. [PMID: 38397737 PMCID: PMC10886048 DOI: 10.3390/antiox13020139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
Acetic acid is a bioactive short-chain fatty acid produced in large quantities from ethanol metabolism. In this review, we describe how acetic acid/acetate generates oxidative stress, alters the function of pre-sympathetic neurons, and can potentially influence cardiovascular function in both humans and rodents after ethanol consumption. Our recent findings from in vivo and in vitro studies support the notion that administration of acetic acid/acetate generates oxidative stress and increases sympathetic outflow, leading to alterations in arterial blood pressure. Real-time investigation of how ethanol and acetic acid/acetate modulate neural control of cardiovascular function can be conducted by microinjecting compounds into autonomic control centers of the brain and measuring changes in peripheral sympathetic nerve activity and blood pressure in response to these compounds.
Collapse
Affiliation(s)
- Andrew D. Chapp
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhiying Shan
- Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA;
| | - Qing-Hui Chen
- Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA;
| |
Collapse
|
15
|
Zhao J, Gao L, Nurrish S, Kaplan JM. Post-synaptic GABA A receptors potentiate transmission by recruiting CaV2 channels to their inputs. Cell Rep 2023; 42:113161. [PMID: 37742192 PMCID: PMC10873018 DOI: 10.1016/j.celrep.2023.113161] [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: 02/21/2023] [Revised: 07/05/2023] [Accepted: 09/07/2023] [Indexed: 09/26/2023] Open
Abstract
We describe a retrograde synaptic signal at the C. elegans GABAergic neuromuscular junction. At this synapse, GABA release is controlled by two voltage-activated calcium channels (UNC-2/CaV2 and EGL-19/CaV1), and muscle responses are mediated by a single GABA receptor (UNC-49/GABAA). Mutations inactivating UNC-49 or those preventing UNC-49 synaptic clustering cause retrograde defects in GABAergic motor neurons, whereby UNC-2/CaV2 levels at active zones, UNC-2 current, and pre-synaptic GABA release are decreased. Inactivating post-synaptic GABAA receptors has no effect on GABA neuron EGL-19/CaV1 levels nor on several other pre-synaptic markers. The effect of GABAA receptors on pre-synaptic strength is not a consequence of decreased GABA transmission and is input selective. Finally, pre-synaptic UNC-2/CaV2 levels are increased when post-synaptic GABAA receptors are increased but are unaffected by increased extra-synaptic receptors. Collectively, these results suggest that clustered post-synaptic GABAA receptors adjust the strength of their inputs by recruiting CaV2 to contacting active zones.
Collapse
Affiliation(s)
- Jian Zhao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Luna Gao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen Nurrish
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua M Kaplan
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Domijan D, Marić M. An interactive cortical architecture for perceptual organization by accentuation. Neural Netw 2023; 169:205-225. [PMID: 39491385 DOI: 10.1016/j.neunet.2023.10.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 11/05/2024]
Abstract
Accentuation has been proposed as a general principle of perceptual organization. Here, we have developed a neurodynamic architecture to explain how accentuation affects boundary segmentation and shape perception. The model consists of bottom-up and top-down pathways. Bottom-up processing involves a set of feature maps that compute bottom-up salience of surfaces, boundaries, boundary completions, and junctions. Then, a feature-based winner-take-all network selects the most salient locations. Top-down processing includes an object-based attention stage that allows enhanced neural activity to propagate from the most salient locations to all connected locations, and a visual segmentation stage that employs inhibitory connections to segregate boundaries into distinct maps. The model was tested on a series of computer simulations showing how the position of the accent affects boundary segregation in the square-diamond and the pointing illusion. The model was also tested on a variety of texture segregation tasks, showing that its performance was comparable to that of human observers. The model suggests that there is an intermediate stage of visual processing between perceptual grouping and object recognition that helps the visual system choose between competing percepts of the ambiguous stimulus.
Collapse
|
17
|
Samuel S, Michael M, Tadros M. Should gastroenterologists prescribe cannabis? The highs, the lows and the unknowns. World J Clin Cases 2023; 11:4210-4230. [PMID: 37449231 PMCID: PMC10336994 DOI: 10.12998/wjcc.v11.i18.4210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/31/2023] [Accepted: 04/14/2023] [Indexed: 06/26/2023] Open
Abstract
Cannabis, commonly known as marijuana, is a drug extracted from the Cannabis plant known for its psychotropic and medicinal properties. It has been used for healing purposes during ancient times, although its psychoactive components led to its restricted use in medicine. Nonetheless, cannabis is found to have modulatory effects on the endocannabinoid system exhibiting its medicinal role in the gastrointestinal (GI) system. Emerging animal and human studies demonstrate the influential effects of cannabis on a variety of GI diseases including inflammatory bowel disease, motility disorders and GI malignancies. It also has a regulatory role in GI symptoms including nausea and vomiting, anorexia, weight gain, abdominal pain, among others. However, both its acute and chronic use can lead to undesirable side effects such as dependency and addiction, cognitive impairment and cannabinoid hyperemesis syndrome. We will discuss the role of cannabis in the GI system as well as dosing strategies to help guide gastroenterologists to assess its efficacy and provide patient counseling before prescription of medical marijuana.
Collapse
Affiliation(s)
- Sonia Samuel
- Department of Internal Medicine, Albany Medical Center, Albany, NY 12208, United States
| | - Mark Michael
- Department of Internal Medicine, Albany Medical Center, Albany, NY 12208, United States
| | - Micheal Tadros
- Department of Gastroenterology and Hepatology, Albany Medical Center, Albany, NY 12208, United States
| |
Collapse
|
18
|
An Z, Yang J, Xiao F, Lv J, Xing X, Liu H, Wang L, Liu Y, Zhang Z, Guo H. Hippocampal Proteomics Reveals the Role of Glutamatergic Synapse Activation in the Depression Induced by Perfluorooctane Sulfonate. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:7866-7877. [PMID: 37191230 DOI: 10.1021/acs.jafc.3c01344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Perfluorooctane sulfonate (PFOS), a new type of persistent organic pollutant in the environment of water, has drawn significant attention in recent years due to its widespread prevalence and high toxicity. Neurotoxicity is regarded as one of the major toxic effects of PFOS, while research studies on PFOS-induced depression and the underlying mechanisms remain scarce. In this study, behavioral tests revealed the depressive-like behaviors in PFOS-exposed male mice. Neuron damages including pyknosis and staining deepening were identified through hematoxylin and eosin staining. Then, we noticed the elevation of glutamate and proline levels as well as the decline of glutamine and tryptophan levels. Proteomics analysis identified 105 differentially expressed proteins that change in a dose-dependent manner and revealed that PFOS exposure activated the glutamatergic synapse signaling pathway, which were further confirmed by Western blot, and the data were consistent with the findings of the proteomics analysis. Additionally, the downstream signaling cyclic AMP-responsive element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF) and synaptic plasticity-related postsynaptic density protein 95, synaptophysin, were downregulated. Our results highlight that PFOS exposure may inhibit the synaptic plasticity of the hippocampus via glutamatergic synapse and the CREB/BDNF signaling pathway to cause depressive-like behaviors in male mice.
Collapse
Affiliation(s)
- Ziwen An
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Jing Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Fang Xiao
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Junli Lv
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaoqing Xing
- Department of Pharmacy, Hebei General Hospital, Shijiazhuang 050017, China
| | - Heqiong Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Lei Wang
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| | - Yi Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
| | - Zhanchi Zhang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang 050017, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China
| | - Huicai Guo
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| |
Collapse
|
19
|
Liu GX, Li ZL, Lin SY, Wang Q, Luo ZY, Wu K, Zhou YL, Ning YP. Mapping metabolite change in the mouse brain after esketamine injection by ambient mass spectrometry imaging and metabolomics. Front Psychiatry 2023; 14:1109344. [PMID: 37234214 PMCID: PMC10206402 DOI: 10.3389/fpsyt.2023.1109344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 03/20/2023] [Indexed: 05/27/2023] Open
Abstract
Ketamine is a new, fast, and effective antidepression treatment method; however, the possible dissociation effects, sensory changes, abuse risk, and the inability to accurately identify whether patients have a significant response to ketamine limit its clinical use. Further exploration of the antidepressant mechanisms of ketamine will contribute to its safe and practical application. Metabolites, the products of upstream gene expression and protein regulatory networks, play an essential role in various physiological and pathophysiological processes. In traditional metabonomics it is difficult to achieve the spatial localization of metabolites, which limits the further analysis of brain metabonomics by researchers. Here, we used a metabolic network mapping method called ambient air flow-assisted desorption electrospray ionization (AFADESI)-mass spectrometry imaging (MSI). We found the main changes in glycerophospholipid metabolism around the brain and sphingolipid metabolism changed mainly in the globus pallidus, which showed the most significant metabolite change after esketamine injection. The spatial distribution of metabolic changes was evaluated in the whole brain, and the potential mechanism of esketamine's antidepressant effect was explored in this research.
Collapse
Affiliation(s)
- Guan-Xi Liu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, China
| | - Ze-Lin Li
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, China
| | - Su-Yan Lin
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Wang
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, China
| | - Zheng-Yi Luo
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, China
| | - Kai Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Yan-Lin Zhou
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, China
| | - Yu-Ping Ning
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, China
| |
Collapse
|
20
|
Azargoonjahromi A. Dual role of nitric oxide in Alzheimer's Disease. Nitric Oxide 2023; 134-135:23-37. [PMID: 37019299 DOI: 10.1016/j.niox.2023.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Nitric oxide (NO), an enzymatic product of nitric oxide synthase (NOS), has been associated with a variety of neurological diseases such as Alzheimer's disease (AD). NO has long been thought to contribute to neurotoxic insults caused by neuroinflammation in AD. This perception shifts as more attention is paid to the early stages before cognitive problems manifest. However, it has revealed a compensatory neuroprotective role for NO that protects synapses by increasing neuronal excitability. NO can positively affect neurons by inducing neuroplasticity, neuroprotection, and myelination, as well as having cytolytic activity to reduce inflammation. NO can also induce long-term potentiation (LTP), a process by which synaptic connections among neurons become more potent. Not to mention that such functions give rise to AD protection. Notably, it is unquestionably necessary to conduct more research to clarify NO pathways in neurodegenerative dementias because doing so could help us better understand their pathophysiology and develop more effective treatment options. All these findings bring us to the prevailing notion that NO can be used either as a therapeutic agent in patients afflicted with AD and other memory impairment disorders or as a contributor to the neurotoxic and aggressive factor in AD. In this review, after presenting a general background on AD and NO, various factors that have a pivotal role in both protecting and exacerbating AD and their correlation with NO will be elucidated. Following this, both the neuroprotective and neurotoxic effects of NO on neurons and glial cells among AD cases will be discussed in detail.
Collapse
|
21
|
Postsynaptic plasticity of Purkinje cells in mice is determined by molecular identity. Commun Biol 2022; 5:1328. [PMID: 36463347 PMCID: PMC9719509 DOI: 10.1038/s42003-022-04283-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/20/2022] [Indexed: 12/05/2022] Open
Abstract
Cerebellar learning is expressed as upbound or downbound changes in simple spike activity of Purkinje cell subpopulations, but the underlying mechanism remains enigmatic. By visualizing murine Purkinje cells with different molecular identities, we demonstrate that the potential for induction of long-term depression is prominent in downbound and minimal in the upbound subpopulation. These differential propensities depend on the expression profile, but not on the synaptic inputs, of the individual Purkinje cell involved, highlighting the functional relevance of intrinsic properties for memory formation.
Collapse
|
22
|
Gouws JM, Sherrington A, Zheng S, Kim JS, Iremonger KJ. Regulation of corticotropin-releasing hormone neuronal network activity by noradrenergic stress signals. J Physiol 2022; 600:4347-4359. [PMID: 36040213 PMCID: PMC9825848 DOI: 10.1113/jp283328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/26/2022] [Indexed: 01/11/2023] Open
Abstract
Noradrenaline is a neurotransmitter released in response to homeostatic challenge and activates the hypothalamic-pituitary-adrenal axis via stimulation of corticotropin-releasing hormone (CRH) neurons. Here we investigated the mechanism through which noradrenaline regulates activity within the CRH neuronal network. Using a combination of in vitro GCaMP6f Ca2+ imaging and electrophysiology, we show that noradrenaline induces a robust increase in excitability in a proportion of CRH neurons with many neurons displaying a bursting mode of activity. Noradrenaline-induced activation required α1 -adrenoceptors and L-type voltage-gated Ca2+ channels, but not GABA/glutamate synaptic transmission or sodium action potentials. Exposure of mice to elevated corticosterone levels was able to suppress noradrenaline-induced activation. These results provide further insight into the mechanisms by which noradrenaline regulates CRH neural network activity and hence stress responses. KEY POINTS: GCaMP6f Ca2+ imaging and on-cell patch-clamp recordings reveal that corticotropin-releasing hormone neurons are activated by noradrenaline with many neurons displaying a bursting mode of activity. Noradrenaline-induced activation requires α1 -adrenoceptors. Noradrenaline-induced Ca2+ elevations persist after blocking GABAA , AMPA, NMDA receptors and voltage-gated Na+ channels. Noradrenaline-induced Ca2+ elevations require L-type voltage-gated Ca2+ channels. Corticosterone suppresses noradrenaline-induced excitation.
Collapse
Affiliation(s)
- Julia M. Gouws
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinOtagoNew Zealand
| | - Aidan Sherrington
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinOtagoNew Zealand
| | - Shaojie Zheng
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinOtagoNew Zealand
| | - Joon S. Kim
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinOtagoNew Zealand
| | - Karl J. Iremonger
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinOtagoNew Zealand
| |
Collapse
|
23
|
Rao MS, Mizuno H, Iwasato T, Mizuno H. Ras GTPase-activating proteins control neuronal circuit development in barrel cortex layer 4. Front Neurosci 2022; 16:901774. [PMID: 36188467 PMCID: PMC9523512 DOI: 10.3389/fnins.2022.901774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022] Open
Abstract
The cerebral cortex comprises a complex and exquisite network of neuronal circuits that is formed during development. To explore the molecular mechanisms involved in cortical circuit formation, the tactile somatosensory pathway that connects the whiskers and cortex of rodents is a useful model. Here, we analyzed the roles of Ras GTPase-activating proteins (RasGAPs) in the circuit formation in the somatosensory cortex layer 4 (L4). We suppressed the function of RasGAPs in L4 neurons using Supernova RNAi, a plasmid vector-based sparse cell gene knockdown (KD) system. The results showed disrupted dendritic pattern formation of L4 spiny stellate neurons on the barrel edge by RasGAP KD. Furthermore, the number of presynaptic boutons on L4 neurons was reduced by RasGAP KD. These results demonstrate the essential roles of RasGAPs in circuit formation in the cerebral cortex and imply that developmental changes in dendrites and synapses in RasGAP KD neurons may be related to cognitive disabilities in RasGAP-deficient individuals, such as patients with neurofibromatosis type 1.
Collapse
Affiliation(s)
- Madhura S. Rao
- Laboratory of Multi-Dimensional Imaging, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
- Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiromi Mizuno
- Laboratory of Multi-Dimensional Imaging, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
- Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takuji Iwasato
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima, Japan
- Department of Genetics, SOKENDAI The Graduate University for Advanced Studies, Mishima, Japan
| | - Hidenobu Mizuno
- Laboratory of Multi-Dimensional Imaging, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
- Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- *Correspondence: Hidenobu Mizuno,
| |
Collapse
|
24
|
Neurotensin Release from Dopamine Neurons Drives Long-Term Depression of Substantia Nigra Dopamine Signaling. J Neurosci 2022; 42:6186-6194. [PMID: 35794014 PMCID: PMC9374153 DOI: 10.1523/jneurosci.1395-20.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 05/11/2022] [Accepted: 06/12/2022] [Indexed: 11/21/2022] Open
Abstract
Midbrain dopamine neurons play central physiological roles in voluntary movement, reward learning, and motivated behavior. Inhibitory signaling at somatodendritic dopamine D2 receptor (D2R) synapses modulates excitability of dopamine neurons. The neuropeptide neurotensin is expressed by many inputs to the midbrain and induces LTD of D2R synaptic currents (LTDDA); however, the source of neurotensin that is responsible for LTDDA is not known. Here we show, in brain slices from male and female mice, that LTDDA is driven by neurotensin released by dopamine neurons themselves. Optogenetic stimulation of dopamine neurons was sufficient to induce LTDDA in the substantia nigra, but not the VTA, and was dependent on neurotensin receptor signaling, postsynaptic calcium, and vacuolar-type H+-ATPase activity in the postsynaptic cell. These findings reveal a novel form of signaling between dopamine neurons involving release of the peptide neurotensin, which may act as a feedforward mechanism to increase dopamine neuron excitability.SIGNIFICANCE STATEMENT Dopamine neurons in the midbrain play a critical role in reward learning and the initiation of movement. Aberrant dopamine neuron function is implicated in a range of diseases and disorders, including Parkinson's disease, schizophrenia, obesity, and substance use disorders. D2 receptor-mediated PSCs are produced by a rare form of dendrodendritic synaptic transmission between dopamine neurons. These D2 receptor-mediated PSCs undergo LTD following application of the neuropeptide neurotensin. Here we show that release of neurotensin by dopamine neurons themselves is sufficient to induce LTD of dopamine transmission in the substantia nigra. Neurotensin signaling therefore mediates a second form of interdopamine neuron communication and may provide a mechanism by which dopamine neurons maintain excitability when nigral dopamine is elevated.
Collapse
|
25
|
Wong NF, Xu-Friedman MA. Induction of Activity-Dependent Plasticity at Auditory Nerve Synapses. J Neurosci 2022; 42:6211-6220. [PMID: 35790402 PMCID: PMC9374128 DOI: 10.1523/jneurosci.0666-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/26/2022] [Accepted: 06/25/2022] [Indexed: 11/21/2022] Open
Abstract
Exposure to nontraumatic noise in vivo drives long-lasting changes in auditory nerve synapses, which may influence hearing, but the induction mechanisms are not known. We mimicked activity in acute slices of the cochlear nucleus from mice of both sexes by treating them with high potassium, after which voltage-clamp recordings from bushy cells indicated that auditory nerve synapses had reduced EPSC amplitude, quantal size, and vesicle release probability (P r). The effects of high potassium were prevented by blockers of nitric oxide (NO) synthase and protein kinase A. Treatment with the NO donor, PAPA-NONOate, also decreased P r, suggesting NO plays a central role in inducing synaptic changes. To identify the source of NO, we activated auditory nerve fibers specifically using optogenetics. Strobing for 2 h led to decreased EPSC amplitude and P r, which was prevented by antagonists against ionotropic glutamate receptors and NO synthase. This suggests that the activation of AMPA and NMDA receptors in postsynaptic targets of auditory nerve fibers drives release of NO, which acts retrogradely to cause long-term changes in synaptic function in auditory nerve synapses. This may provide insight into preventing or treating disorders caused by noise exposure.SIGNIFICANCE STATEMENT Auditory nerve fibers undergo long-lasting changes in synaptic properties in response to noise exposure in vivo, which may contribute to changes in hearing. Here, we investigated the cellular mechanisms underlying induction of synaptic changes using high potassium and optogenetic stimulation in vitro and identified important signaling pathways using pharmacology. Our results suggest that auditory nerve activity drives postsynaptic depolarization through AMPA and NMDA receptors, leading to the release of nitric oxide, which acts retrogradely to regulate presynaptic neurotransmitter release. These experiments revealed that auditory nerve synapses are unexpectedly sensitive to activity and can show dramatic, long-lasting changes in a few hours that could affect hearing.
Collapse
Affiliation(s)
- Nicole F Wong
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| | - Matthew A Xu-Friedman
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| |
Collapse
|
26
|
Moorjani S, Walvekar S, Fetz EE, Perlmutter SI. Movement-dependent electrical stimulation for volitional strengthening of cortical connections in behaving monkeys. Proc Natl Acad Sci U S A 2022; 119:e2116321119. [PMID: 35759657 PMCID: PMC9271159 DOI: 10.1073/pnas.2116321119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 04/29/2022] [Indexed: 12/30/2022] Open
Abstract
Correlated activity of neurons can lead to long-term strengthening or weakening of the connections between them. In addition, the behavioral context, imparted by execution of physical movements or the presence of a reward, can modulate the plasticity induced by Hebbian mechanisms. In the present study, we have combined behavior and induced neuronal correlations to strengthen connections in the motor cortex of adult behaving monkeys. Correlated activity was induced using an electrical-conditioning protocol in which stimuli gated by voluntary movements were used to produce coactivation of neurons at motor-cortical sites involved in those movements. Delivery of movement-dependent stimulation resulted in small increases in the strength of associated cortical connections immediately after conditioning. Remarkably, when paired with further repetition of the movements that gated the conditioning stimuli, there were substantially larger gains in the strength of cortical connections, which occurred in a use-dependent manner, without delivery of additional conditioning stimulation. In the absence of such movements, little change was observed in the strength of motor-cortical connections. Performance of the motor behavior in the absence of conditioning also did not produce any changes in connectivity. Our results show that combining movement-gated stimulation with further natural use of the "conditioned" pathways after stimulation ends can produce use-dependent strengthening of connections in adult primates, highlighting an important role for behavior in cortical plasticity. Our data also provide strong support for combining movement-gated stimulation with use-dependent physical rehabilitation for strengthening connections weakened by a stroke or spinal cord injury.
Collapse
Affiliation(s)
- Samira Moorjani
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195
- Center for Neurotechnology, University of Washington, Seattle, WA 98195
| | - Sarita Walvekar
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195
| | - Eberhard E Fetz
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195
- Center for Neurotechnology, University of Washington, Seattle, WA 98195
| | - Steve I Perlmutter
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195
- Center for Neurotechnology, University of Washington, Seattle, WA 98195
| |
Collapse
|
27
|
Varfolomeev SD, Panin AA, Semenova NA, Ublinskiy MV, Akhadov TA, Bykov VI, Tsybenova SB. Molecular mechanism and regulation of neurovascular coupling in the human brain: The influence of thermoheliox. Chem Phys Lett 2022. [DOI: 10.1016/j.cplett.2022.139485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Fukumitsu K, Kaneko M, Maruyama T, Yoshihara C, Huang AJ, McHugh TJ, Itohara S, Tanaka M, Kuroda KO. Amylin-Calcitonin receptor signaling in the medial preoptic area mediates affiliative social behaviors in female mice. Nat Commun 2022; 13:709. [PMID: 35136064 PMCID: PMC8825811 DOI: 10.1038/s41467-022-28131-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 01/10/2022] [Indexed: 01/27/2023] Open
Abstract
Social animals actively engage in contact with conspecifics and experience stress upon isolation. However, the neural mechanisms coordinating the sensing and seeking of social contacts are unclear. Here we report that amylin-calcitonin receptor (Calcr) signaling in the medial preoptic area (MPOA) mediates affiliative social contacts among adult female mice. Isolation of females from free social interactions first induces active contact-seeking, then depressive-like behavior, concurrent with a loss of Amylin mRNA expression in the MPOA. Reunion with peers induces physical contacts, activates both amylin- and Calcr-expressing neurons, and leads to a recovery of Amylin mRNA expression. Chemogenetic activation of amylin neurons increases and molecular knockdown of either amylin or Calcr attenuates contact-seeking behavior, respectively. Our data provide evidence in support of a previously postulated origin of social affiliation in mammals.
Collapse
Affiliation(s)
- Kansai Fukumitsu
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, 351-0198, Japan.
| | - Misato Kaneko
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, 351-0198, Japan.,Department of Animal Science, Faculty of Applied Life Science, Nippon Veterinary and Life Science University, Musashino, Tokyo, 180-8602, Japan
| | - Teppo Maruyama
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, 351-0198, Japan.,Department of Animal Science, Faculty of Applied Life Science, Nippon Veterinary and Life Science University, Musashino, Tokyo, 180-8602, Japan
| | - Chihiro Yoshihara
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, 351-0198, Japan
| | - Arthur J Huang
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Saitama, 351-0198, Japan
| | - Thomas J McHugh
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Saitama, 351-0198, Japan
| | - Shigeyoshi Itohara
- Laboratory for Behavioral Genetics, RIKEN Center for Brain Science, Saitama, 351-0198, Japan
| | - Minoru Tanaka
- Department of Animal Science, Faculty of Applied Life Science, Nippon Veterinary and Life Science University, Musashino, Tokyo, 180-8602, Japan
| | - Kumi O Kuroda
- Laboratory for Affiliative Social Behavior, RIKEN Center for Brain Science, Saitama, 351-0198, Japan.
| |
Collapse
|
29
|
Ren P, Xiao B, Wang LP, Li YS, Jin H, Jin QH. Nitric oxide impairs spatial learning and memory in a rat model of Alzheimer's disease via disturbance of glutamate response in the hippocampal dentate gyrus during spatial learning. Behav Brain Res 2022; 422:113750. [PMID: 35033612 DOI: 10.1016/j.bbr.2022.113750] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/19/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023]
Abstract
Nitric oxide (NO)-dependent pathways may play a significant role in the decline of synaptic and cognitive functions in Alzheimer's disease (AD). However, whether NO in the hippocampal dentate gyrus (DG) is involved in the spatial learning and memory impairments of AD by affecting the glutamate (Glu) response during these processes is not well-understood. Here, we prepared an AD rat model by long-term i.p. of D-galactose into ovariectomized rats, and then the effects of L-NMMA (a NO synthase inhibitor) on Glu concentration and amplitude of field excitatory postsynaptic potential (fEPSP) were measured in the DG region during the Morris water maze (MWM) test in freely-moving rats. During the MWM test, compared with the sham group, the escape latency was increased in the place navigation trial, and the percentage of time spent in target quadrant and the number of platform crossings were decreased in the spatial probe trial, in addition, the increase of fEPSP amplitude in the DG was significantly attenuated in AD group rats. L-NMMA significantly attenuated the spatial learning and memory impairment in AD rats, and reversed the inhibitory effect of AD on increase of fEPSP amplitude in the DG during the MWM test. In sham group rats, the Glu level in the DG increased significantly during the MWM test, and this response was markedly enhanced in AD rats. Furthermore, the response of Glu in the DG during spatial learning was recovered by microinjection of L-NMMA into the DG. Our results suggest that NO in the DG impairs spatial learning and memory and related synaptic plasticity in AD rats, by disturbing the Glu response during spatial learning.
Collapse
Affiliation(s)
- Peng Ren
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Bin Xiao
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Lin-Ping Wang
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Ying-Shun Li
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| | - Hua Jin
- Department of Internal Medicine, Yanbian University Hospital, Yanji, China.
| | - Qing-Hua Jin
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University, Yanji, China
| |
Collapse
|
30
|
Zheng Z, Chen J, Chopp M. Mechanisms of Plasticity Remodeling and Recovery. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
31
|
de Melo Reis RA, Isaac AR, Freitas HR, de Almeida MM, Schuck PF, Ferreira GC, Andrade-da-Costa BLDS, Trevenzoli IH. Quality of Life and a Surveillant Endocannabinoid System. Front Neurosci 2021; 15:747229. [PMID: 34776851 PMCID: PMC8581450 DOI: 10.3389/fnins.2021.747229] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/01/2021] [Indexed: 12/11/2022] Open
Abstract
The endocannabinoid system (ECS) is an important brain modulatory network. ECS regulates brain homeostasis throughout development, from progenitor fate decision to neuro- and gliogenesis, synaptogenesis, brain plasticity and circuit repair, up to learning, memory, fear, protection, and death. It is a major player in the hypothalamic-peripheral system-adipose tissue in the regulation of food intake, energy storage, nutritional status, and adipose tissue mass, consequently affecting obesity. Loss of ECS control might affect mood disorders (anxiety, hyperactivity, psychosis, and depression), lead to drug abuse, and impact neurodegenerative (Alzheimer's, Parkinson, Huntington, Multiple, and Amyotrophic Lateral Sclerosis) and neurodevelopmental (autism spectrum) disorders. Practice of regular physical and/or mind-body mindfulness and meditative activities have been shown to modulate endocannabinoid (eCB) levels, in addition to other players as brain-derived neurotrophic factor (BDNF). ECS is involved in pain, inflammation, metabolic and cardiovascular dysfunctions, general immune responses (asthma, allergy, and arthritis) and tumor expansion, both/either in the brain and/or in the periphery. The reason for such a vast impact is the fact that arachidonic acid, a precursor of eCBs, is present in every membrane cell of the body and on demand eCBs synthesis is regulated by electrical activity and calcium shifts. Novel lipid (lipoxins and resolvins) or peptide (hemopressin) players of the ECS also operate as regulators of physiological allostasis. Indeed, the presence of cannabinoid receptors in intracellular organelles as mitochondria or lysosomes, or in nuclear targets as PPARγ might impact energy consumption, metabolism and cell death. To live a better life implies in a vigilant ECS, through healthy diet selection (based on a balanced omega-3 and -6 polyunsaturated fatty acids), weekly exercises and meditation therapy, all of which regulating eCBs levels, surrounded by a constructive social network. Cannabidiol, a diet supplement has been a major player with anti-inflammatory, anxiolytic, antidepressant, and antioxidant activities. Cognitive challenges and emotional intelligence might strengthen the ECS, which is built on a variety of synapses that modify human behavior. As therapeutically concerned, the ECS is essential for maintaining homeostasis and cannabinoids are promising tools to control innumerous targets.
Collapse
Affiliation(s)
- Ricardo Augusto de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alinny Rosendo Isaac
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hércules Rezende Freitas
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana Macedo de Almeida
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Fernanda Schuck
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Costa Ferreira
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Isis Hara Trevenzoli
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Kourosh-Arami M, Hosseini N, Mohsenzadegan M, Komaki A, Joghataei MT. Neurophysiologic implications of neuronal nitric oxide synthase. Rev Neurosci 2021; 31:617-636. [PMID: 32739909 DOI: 10.1515/revneuro-2019-0111] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/21/2020] [Indexed: 12/12/2022]
Abstract
The molecular and chemical properties of neuronal nitric oxide synthase (nNOS) have made it a key mediator in many physiological functions and signaling transduction. The NOS monomer is inactive, but the dimer form is active. There are three forms of NOS, which are neuronal (nNOS), inducible (iNOS), and endothelial (eNOS) nitric oxide synthase. nNOS regulates nitric oxide (NO) synthesis which is the mechanism used mostly by neurons to produce NO. nNOS expression and activation is regulated by some important signaling proteins, such as cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB), calmodulin (CaM), heat shock protein 90 (HSP90)/HSP70. nNOS-derived NO has been implicated in modulating many physiological functions, such as synaptic plasticity, learning, memory, neurogenesis, etc. In this review, we have summarized recent studies that have characterized structural features, subcellular localization, and factors that regulate nNOS function. Finally, we have discussed the role of nNOS in the developing brain under a wide range of physiological conditions, especially long-term potentiation and depression.
Collapse
Affiliation(s)
- Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Nasrin Hosseini
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Monireh Mohsenzadegan
- Department of Laboratory Sciences, Allied Medical College, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Alireza Komaki
- Department of Physiology, Medical College, Hamedan University of Medical Sciences, Hamedan, Islamic Republic of Iran
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| |
Collapse
|
33
|
Sinha M, Narayanan R. Active Dendrites and Local Field Potentials: Biophysical Mechanisms and Computational Explorations. Neuroscience 2021; 489:111-142. [PMID: 34506834 PMCID: PMC7612676 DOI: 10.1016/j.neuroscience.2021.08.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 10/27/2022]
Abstract
Neurons and glial cells are endowed with membranes that express a rich repertoire of ion channels, transporters, and receptors. The constant flux of ions across the neuronal and glial membranes results in voltage fluctuations that can be recorded from the extracellular matrix. The high frequency components of this voltage signal contain information about the spiking activity, reflecting the output from the neurons surrounding the recording location. The low frequency components of the signal, referred to as the local field potential (LFP), have been traditionally thought to provide information about the synaptic inputs that impinge on the large dendritic trees of various neurons. In this review, we discuss recent computational and experimental studies pointing to a critical role of several active dendritic mechanisms that can influence the genesis and the location-dependent spectro-temporal dynamics of LFPs, spanning different brain regions. We strongly emphasize the need to account for the several fast and slow dendritic events and associated active mechanisms - including gradients in their expression profiles, inter- and intra-cellular spatio-temporal interactions spanning neurons and glia, heterogeneities and degeneracy across scales, neuromodulatory influences, and activitydependent plasticity - towards gaining important insights about the origins of LFP under different behavioral states in health and disease. We provide simple but essential guidelines on how to model LFPs taking into account these dendritic mechanisms, with detailed methodology on how to account for various heterogeneities and electrophysiological properties of neurons and synapses while studying LFPs.
Collapse
Affiliation(s)
- Manisha Sinha
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka 560012, India.
| |
Collapse
|
34
|
Chatterjee R, Paluh JL, Chowdhury S, Mondal S, Raha A, Mukherjee A. SyNC, a Computationally Extensive and Realistic Neural Net to Identify Relative Impacts of Synaptopathy Mechanisms on Glutamatergic Neurons and Their Networks in Autism and Complex Neurological Disorders. Front Cell Neurosci 2021; 15:674030. [PMID: 34354570 PMCID: PMC8330424 DOI: 10.3389/fncel.2021.674030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/25/2021] [Indexed: 11/24/2022] Open
Abstract
Synaptic function and experience-dependent plasticity across multiple synapses are dependent on the types of neurons interacting as well as the intricate mechanisms that operate at the molecular level of the synapse. To understand the complexity of information processing at synaptic networks will rely in part on effective computational models. Such models should also evaluate disruptions to synaptic function by multiple mechanisms. By co-development of algorithms alongside hardware, real time analysis metrics can be co-prioritized along with biological complexity. The hippocampus is implicated in autism spectrum disorders (ASD) and within this region glutamatergic neurons constitute 90% of the neurons integral to the functioning of neuronal networks. Here we generate a computational model referred to as ASD interrogator (ASDint) and corresponding hardware to enable in silicon analysis of multiple ASD mechanisms affecting glutamatergic neuron synapses. The hardware architecture Synaptic Neuronal Circuit, SyNC, is a novel GPU accelerator or neural net, that extends discovery by acting as a biologically relevant realistic neuron synapse in real time. Co-developed ASDint and SyNC expand spiking neural network models of plasticity to comparative analysis of retrograde messengers. The SyNC model is realized in an ASIC architecture, which enables the ability to compute increasingly complex scenarios without sacrificing area efficiency of the model. Here we apply the ASDint model to analyse neuronal circuitry dysfunctions associated with autism spectral disorder (ASD) synaptopathies and their effects on the synaptic learning parameter and demonstrate SyNC on an ideal ASDint scenario. Our work highlights the value of secondary pathways in regard to evaluating complex ASD synaptopathy mechanisms. By comparing the degree of variation in the synaptic learning parameter to the response obtained from simulations of the ideal scenario we determine the potency and time of the effect of a particular evaluated mechanism. Hence simulations of such scenarios in even a small neuronal network now allows us to identify relative impacts of changed parameters and their effect on synaptic function. Based on this, we can estimate the minimum fraction of a neuron exhibiting a particular dysfunction scenario required to lead to complete failure of a neural network to coordinate pre-synaptic and post-synaptic outputs.
Collapse
Affiliation(s)
- Rounak Chatterjee
- Department of Electronics and Telecommunication Engineering, Jadavpur University, Kolkata, India
| | - Janet L Paluh
- SUNY Polytechnic Institute, College of Nanoscale Science and Engineering, Nanobioscience, Albany, NY, United States
| | - Souradeep Chowdhury
- Department of Electronics and Telecommunication Engineering, Jadavpur University, Kolkata, India
| | - Soham Mondal
- Flash Controller Team, Memory Solutions, Samsung Semiconductor India Research, Samsung Electronics Co., Ltd., Bangalore, India
| | - Arnab Raha
- Advanced Architecture Research, Intel Intelligent Systems Group, Intel Edge AI, Intel Corporation, Santa Clara, CA, United States
| | | |
Collapse
|
35
|
Nordengen K, Morland C, Slusher BS, Gundersen V. Dendritic Localization and Exocytosis of NAAG in the Rat Hippocampus. Cereb Cortex 2021; 30:1422-1435. [PMID: 31504271 PMCID: PMC7132944 DOI: 10.1093/cercor/bhz176] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 07/04/2019] [Accepted: 07/04/2019] [Indexed: 12/16/2022] Open
Abstract
While a lot is known about classical, anterograde neurotransmission, less is known about the mechanisms and molecules involved in retrograde neurotransmission. Our hypothesis is that N-acetylaspartylglutamate (NAAG), the most abundant dipeptide in the brain, may act as a retrograde transmitter in the brain. NAAG was predominantly localized in dendritic compartments of glutamatergic synapses in the intact hippocampus, where it was present in close proximity to synaptic-like vesicles. In acute hippocampal slices, NAAG was depleted from postsynaptic dendritic elements during neuronal stimulation induced by depolarizing concentrations of potassium or by exposure to glutamate receptor (GluR) agonists. The depletion was completely blocked by botulinum toxin B and strictly dependent on extracellular calcium, indicating exocytotic release. In contrast, there were low levels of NAAG and no effect by depolarization or GluR agonists in presynaptic glutamatergic terminals or GABAergic pre- and postsynaptic elements. Together these data suggest a possible role for NAAG as a retrograde signaling molecule at glutamatergic synapses via exocytotic release.
Collapse
Affiliation(s)
- K Nordengen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo NO-0317, Norway.,Department of Neurology, Akershus University Hospital, Lørenskog N-1478, Norway
| | - C Morland
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo NO-0317, Norway.,Department of Pharmaceutical Biosciences, Institute of Pharmacy, University of Oslo, Oslo NO-0317, Norway
| | - B S Slusher
- Department of Neurology and Johns Hopkins Drug Discovery, John Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - V Gundersen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo NO-0317, Norway.,Department of Neurology, Oslo University Hospital, Rikshospitalet, Oslo N-0424, Norway.,Department of Neurology, Institute of Clinical Medicine, University of Oslo, Oslo NO-0317, Norway
| |
Collapse
|
36
|
Kitcher SR, Pederson AM, Weisz CJC. Diverse identities and sites of action of cochlear neurotransmitters. Hear Res 2021; 419:108278. [PMID: 34108087 DOI: 10.1016/j.heares.2021.108278] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/30/2021] [Accepted: 05/18/2021] [Indexed: 11/18/2022]
Abstract
Accurate encoding of acoustic stimuli requires temporally precise responses to sound integrated with cellular mechanisms that encode the complexity of stimuli over varying timescales and orders of magnitude of intensity. Sound in mammals is initially encoded in the cochlea, the peripheral hearing organ, which contains functionally specialized cells (including hair cells, afferent and efferent neurons, and a multitude of supporting cells) to allow faithful acoustic perception. To accomplish the demanding physiological requirements of hearing, the cochlea has developed synaptic arrangements that operate over different timescales, with varied strengths, and with the ability to adjust function in dynamic hearing conditions. Multiple neurotransmitters interact to support the precision and complexity of hearing. Here, we review the location of release, action, and function of neurotransmitters in the mammalian cochlea with an emphasis on recent work describing the complexity of signaling.
Collapse
Affiliation(s)
- Siân R Kitcher
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, United States
| | - Alia M Pederson
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, United States
| | - Catherine J C Weisz
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, United States.
| |
Collapse
|
37
|
Vandael D, Okamoto Y, Jonas P. Transsynaptic modulation of presynaptic short-term plasticity in hippocampal mossy fiber synapses. Nat Commun 2021; 12:2912. [PMID: 34006874 PMCID: PMC8131630 DOI: 10.1038/s41467-021-23153-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 04/06/2021] [Indexed: 11/21/2022] Open
Abstract
The hippocampal mossy fiber synapse is a key synapse of the trisynaptic circuit. Post-tetanic potentiation (PTP) is the most powerful form of plasticity at this synaptic connection. It is widely believed that mossy fiber PTP is an entirely presynaptic phenomenon, implying that PTP induction is input-specific, and requires neither activity of multiple inputs nor stimulation of postsynaptic neurons. To directly test cooperativity and associativity, we made paired recordings between single mossy fiber terminals and postsynaptic CA3 pyramidal neurons in rat brain slices. By stimulating non-overlapping mossy fiber inputs converging onto single CA3 neurons, we confirm that PTP is input-specific and non-cooperative. Unexpectedly, mossy fiber PTP exhibits anti-associative induction properties. EPSCs show only minimal PTP after combined pre- and postsynaptic high-frequency stimulation with intact postsynaptic Ca2+ signaling, but marked PTP in the absence of postsynaptic spiking and after suppression of postsynaptic Ca2+ signaling (10 mM EGTA). PTP is largely recovered by inhibitors of voltage-gated R- and L-type Ca2+ channels, group II mGluRs, and vacuolar-type H+-ATPase, suggesting the involvement of retrograde vesicular glutamate signaling. Transsynaptic regulation of PTP extends the repertoire of synaptic computations, implementing a brake on mossy fiber detonation and a "smart teacher" function of hippocampal mossy fiber synapses.
Collapse
Affiliation(s)
- David Vandael
- Cellular Neuroscience, IST Austria (Institute of Science and Technology Austria), Klosterneuburg, Austria.
- Netherlands Institute for Neuroscience, Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, The Netherlands.
| | - Yuji Okamoto
- Cellular Neuroscience, IST Austria (Institute of Science and Technology Austria), Klosterneuburg, Austria
| | - Peter Jonas
- Cellular Neuroscience, IST Austria (Institute of Science and Technology Austria), Klosterneuburg, Austria.
| |
Collapse
|
38
|
Jankowska M, Klimek A, Valsecchi C, Stankiewicz M, Wyszkowska J, Rogalska J. Electromagnetic field and TGF-β enhance the compensatory plasticity after sensory nerve injury in cockroach Periplaneta americana. Sci Rep 2021; 11:6582. [PMID: 33753758 PMCID: PMC7985317 DOI: 10.1038/s41598-021-85341-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/01/2021] [Indexed: 11/17/2022] Open
Abstract
Recovery of function after sensory nerves injury involves compensatory plasticity, which can be observed in invertebrates. The aim of the study was the evaluation of compensatory plasticity in the cockroach (Periplaneta americana) nervous system after the sensory nerve injury and assessment of the effect of electromagnetic field exposure (EMF, 50 Hz, 7 mT) and TGF-β on this process. The bioelectrical activities of nerves (pre-and post-synaptic parts of the sensory path) were recorded under wind stimulation of the cerci before and after right cercus ablation and in insects exposed to EMF and treated with TGF-β. Ablation of the right cercus caused an increase of activity of the left presynaptic part of the sensory path. Exposure to EMF and TGF-β induced an increase of activity in both parts of the sensory path. This suggests strengthening effects of EMF and TGF-β on the insect ability to recognize stimuli after one cercus ablation. Data from locomotor tests proved electrophysiological results. The takeover of the function of one cercus by the second one proves the existence of compensatory plasticity in the cockroach escape system, which makes it a good model for studying compensatory plasticity. We recommend further research on EMF as a useful factor in neurorehabilitation.
Collapse
Affiliation(s)
- Milena Jankowska
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100, Toruń, Poland
| | - Angelika Klimek
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100, Toruń, Poland
| | - Chiara Valsecchi
- Federal University of Pampa, Campus Alegrete, Alegrete, RS, Brazil
| | - Maria Stankiewicz
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100, Toruń, Poland
| | - Joanna Wyszkowska
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100, Toruń, Poland.
| | - Justyna Rogalska
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Lwowska 1, 87-100, Toruń, Poland
| |
Collapse
|
39
|
Cao LL, Marshall JM, Fabritz L, Brain KL. Resting cardiac sympathetic firing frequencies suppress terminal norepinephrine transporter uptake. Auton Neurosci 2021; 232:102794. [PMID: 33714751 DOI: 10.1016/j.autneu.2021.102794] [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: 01/19/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 10/22/2022]
Abstract
The prejunctional norepinephrine transporter (NET) is responsible for the clearance of released norepinephrine (NE) back into the sympathetic nerve terminal. NET regulation must be tightly controlled as variations could have important implications for neurotransmission. Thus far, the effects of sympathetic neuronal activity on NET function have been unclear. Here, we optically monitor single-terminal cardiac NET activity ex vivo in response to a broad range of sympathetic postganglionic action potential (AP) firing frequencies. Isolated murine left atrial appendages were loaded with a fluorescent NET substrate [Neurotransmitter Transporter Uptake Assay (NTUA)] and imaged with confocal microscopy. Sympathetic APs were induced with electrical field stimulation at 0.2-10 Hz (0.1-0.2 ms pulse width). Exogenous NE was applied during the NTUA uptake- and washout phases to investigate substrate competition and displacement, respectively, on transport. Single-terminal NET reuptake rate was rapidly suppressed in a frequency-dependent manner with an inhibitory EF50 of 0.9 Hz. At 2 Hz, the effect was reversed by the α2-adrenoceptor antagonist yohimbine (1 μM) (p < 0.01) with no further effect imposed by the muscarinic receptor antagonist atropine (1 μM). Additionally, high exogenous NE concentrations abolished NET reuptake (1 μM NE; p < 0.0001) and displaced terminal specific NTUA during washout (1-100 μM NE; p < 0.0001). We have also identified α2-adrenoceptor-induced suppression of NET reuptake rate during resting stimulation frequencies, which could oppose the effect of autoinhibition-mediated suppression of exocytosis and thus amplify the effects of sympathetic drive on cardiac function.
Collapse
Affiliation(s)
- Lily L Cao
- School of Biomedical Science, Institute of Clinical Science, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK.
| | - Janice M Marshall
- School of Biomedical Science, Institute of Clinical Science, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK.
| | - Larissa Fabritz
- Institute of Cardiovascular Science, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK; Department of Cardiology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| | - Keith L Brain
- School of Biomedical Science, Institute of Clinical Science, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK.
| |
Collapse
|
40
|
Retrograde Suppression of Post-Tetanic Potentiation at the Mossy Fiber-CA3 Pyramidal Cell Synapse. eNeuro 2021; 8:ENEURO.0450-20.2021. [PMID: 33593734 PMCID: PMC7986537 DOI: 10.1523/eneuro.0450-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/17/2020] [Accepted: 01/16/2021] [Indexed: 11/21/2022] Open
Abstract
In the hippocampus, the excitatory synapse between dentate granule cell (GC) axons, or mossy fibers (MFs), and CA3 pyramidal cells (MF-CA3) expresses robust forms of short-term plasticity, such as frequency facilitation and post-tetanic potentiation (PTP). These forms of plasticity are due to increases in presynaptic neurotransmitter release, and can be engaged when dentate GCs fire in bursts (e.g., during exploratory behaviors) and bring CA3 pyramidal neurons above threshold. While frequency facilitation at this synapse is limited by endogenous activation of presynaptic metabotropic glutamate receptors (mGluRs), whether MF-PTP can be regulated in an activity-dependent manner is unknown. Here, using physiologically relevant patterns of MF stimulation in acute mouse hippocampal slices, we found that disrupting postsynaptic Ca2+ dynamics increases MF-PTP, strongly suggesting a form of Ca2+-dependent retrograde suppression of this form of plasticity. PTP suppression requires a few seconds of MF bursting activity and Ca2+ release from internal stores. Our findings raise the possibility that the powerful MF-CA3 synapse can negatively regulate its own strength not only during PTP-inducing activity typical of normal exploratory behaviors, but also during epileptic activity.
Collapse
|
41
|
Schmidt SD, Zinn CG, Behling JAK, Furian AF, Furini CRG, de Carvalho Myskiw J, Izquierdo I. Inhibition of PACAP/PAC1/VPAC2 signaling impairs the consolidation of social recognition memory and nitric oxide prevents this deficit. Neurobiol Learn Mem 2021; 180:107423. [PMID: 33705861 DOI: 10.1016/j.nlm.2021.107423] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/18/2021] [Accepted: 03/03/2021] [Indexed: 11/17/2022]
Abstract
Social recognition memory (SRM) forms the basis of social relationships of animals. It is essential for social interaction and adaptive behavior, reproduction and species survival. Evidence demonstrates that social deficits of psychiatric disorders such as autism and schizophrenia are caused by alterations in SRM processing by the hippocampus and amygdala. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) and its receptors PAC1, VPAC1 and VPAC2 are highly expressed in these regions. PACAP is a pleiotropic neuropeptide that modulates synaptic function and plasticity and is thought to be involved in social behavior. PACAP signaling also stimulates the nitric oxide (NO) production and targets outcomes to synapses. In the present work, we investigate the effect of the infusion of PACAP-38 (endogenous neuropeptide and potent stimulator of adenylyl cyclase), PACAP 6-38 (PAC1/VPAC2 receptors antagonist) and S-Nitroso-N-acetyl-DL-penicillamine (SNAP, NO donor) in the CA1 region of the hippocampus and in the basolateral amygdala (BLA) on the consolidation of SRM. For this, male Wistar rats with cannulae implanted in CA1 or in BLA were subjected to a social discrimination paradigm, which is based on the natural ability of rodents to investigate unfamiliar conspecifics more than familiar one. In the sample phase (acquisition), animals were exposed to a juvenile conspecific for 1 h. Immediately, 60 or 150 min after, animals received one of different pharmacological treatments. Twenty-four hours later, they were submitted to a 5 min retention test in the presence of the previously presented juvenile (familiar) and a novel juvenile. Animals that received infusions of PACAP 6-38 (40 pg/side) into CA1 immediately after the sample phase or into BLA immediately or 60 min after the sample phase were unable to recognize the familiar juvenile during the retention test. This impairment was abolished by the coinfusion of PACAP 6-38 plus SNAP (5 μg/side). These results show that the blockade of PACAP/PAC1/VPAC2 signaling in the CA1 and BLA during a restricted post-acquisition time window impairs the consolidation of SRM and that the SNAP is able to abolish this deficit. Findings like this could potentially be used in the future to influence studies of psychiatric disorders involving social behavior.
Collapse
Affiliation(s)
- Scheila Daiane Schmidt
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil.
| | - Carolina Garrido Zinn
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil
| | - Jonny Anderson Kielbovicz Behling
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil
| | - Ana Flávia Furian
- Laboratory of Neurotoxicity, Federal University of Santa Maria (UFSM), Av. Roraima, 1000, 97105-900 Santa Maria, RS, Brazil
| | - Cristiane Regina Guerino Furini
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil; National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Jociane de Carvalho Myskiw
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil; National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.
| | - Ivan Izquierdo
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690-2nd Floor, 90610-000 Porto Alegre, RS, Brazil; National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.
| |
Collapse
|
42
|
Madhurantakam S, Karnam JB, Brabazon D, Takai M, Ahad IU, Balaguru Rayappan JB, Krishnan UM. "Nano": An Emerging Avenue in Electrochemical Detection of Neurotransmitters. ACS Chem Neurosci 2020; 11:4024-4047. [PMID: 33285063 DOI: 10.1021/acschemneuro.0c00355] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The growing importance of nanomaterials toward the detection of neurotransmitter molecules has been chronicled in this review. Neurotransmitters (NTs) are chemicals that serve as messengers in synaptic transmission and are key players in brain functions. Abnormal levels of NTs are associated with numerous psychotic and neurodegenerative diseases. Therefore, their sensitive and robust detection is of great significance in clinical diagnostics. For more than three decades, electrochemical sensors have made a mark toward clinical detection of NTs. The superiority of these electrochemical sensors lies in their ability to enable sensitive, simple, rapid, and selective determination of analyte molecules while remaining relatively inexpensive. Additionally, these sensors are capable of being integrated in robust, portable, and miniaturized devices to establish point-of-care diagnostic platforms. Nanomaterials have emerged as promising materials with significant implications for electrochemical sensing due to their inherent capability to achieve high surface coverage, superior sensitivity, and rapid response in addition to simple device architecture and miniaturization. Considering the enormous significance of the levels of NTs in biological systems and the advances in sensing ushered in with the integration of nanotechnology in electrochemistry, the analysis of NTs by employing nanomaterials as interface materials in various matrices has emerged as an active area of research. This review explores the advancements made in the field of electrochemical sensors for the sensitive and selective determination of NTs which have been described in the past two decades with a distinctive focus on extremely innovative attributes introduced by nanotechnology.
Collapse
Affiliation(s)
- Sasya Madhurantakam
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata 951-8510, Japan
| | - Jayanth Babu Karnam
- School of Electrical and Electronics Engineering, SASTRA Deemed University, Thanjavur 613401, India
- Centre for Nanotechnology and Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur 613401, India
| | - Dermot Brabazon
- I-Form, Advanced Manufacturing Research Centre, Advanced Processing Technology Research Centre, Dublin City University, Dublin, Ireland
| | - Madoka Takai
- Department of Bioengineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Inam Ul Ahad
- I-Form, Advanced Manufacturing Research Centre, Advanced Processing Technology Research Centre, Dublin City University, Dublin, Ireland
| | | | - Uma Maheswari Krishnan
- Centre for Nanotechnology and Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur 613401, India
- School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613401, India
| |
Collapse
|
43
|
Voorn RA, Vogl C. Molecular Assembly and Structural Plasticity of Sensory Ribbon Synapses-A Presynaptic Perspective. Int J Mol Sci 2020; 21:E8758. [PMID: 33228215 PMCID: PMC7699581 DOI: 10.3390/ijms21228758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
In the mammalian cochlea, specialized ribbon-type synapses between sensory inner hair cells (IHCs) and postsynaptic spiral ganglion neurons ensure the temporal precision and indefatigability of synaptic sound encoding. These high-through-put synapses are presynaptically characterized by an electron-dense projection-the synaptic ribbon-which provides structural scaffolding and tethers a large pool of synaptic vesicles. While advances have been made in recent years in deciphering the molecular anatomy and function of these specialized active zones, the developmental assembly of this presynaptic interaction hub remains largely elusive. In this review, we discuss the dynamic nature of IHC (pre-) synaptogenesis and highlight molecular key players as well as the transport pathways underlying this process. Since developmental assembly appears to be a highly dynamic process, we further ask if this structural plasticity might be maintained into adulthood, how this may influence the functional properties of a given IHC synapse and how such plasticity could be regulated on the molecular level. To do so, we take a closer look at other ribbon-bearing systems, such as retinal photoreceptors and pinealocytes and aim to infer conserved mechanisms that may mediate these phenomena.
Collapse
MESH Headings
- Alcohol Oxidoreductases/genetics
- Alcohol Oxidoreductases/metabolism
- Animals
- Co-Repressor Proteins/genetics
- Co-Repressor Proteins/metabolism
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Cytoskeleton/metabolism
- Cytoskeleton/ultrastructure
- Gene Expression Regulation, Developmental
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/ultrastructure
- Hair Cells, Auditory, Outer/metabolism
- Hair Cells, Auditory, Outer/ultrastructure
- Hair Cells, Vestibular/metabolism
- Hair Cells, Vestibular/ultrastructure
- Mechanotransduction, Cellular
- Mice
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neuronal Plasticity/genetics
- Neuropeptides/genetics
- Neuropeptides/metabolism
- Rats
- Synapses/metabolism
- Synapses/ultrastructure
- Synaptic Transmission/genetics
- Synaptic Vesicles/metabolism
- Synaptic Vesicles/ultrastructure
Collapse
Affiliation(s)
- Roos Anouk Voorn
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Goettingen, 37075 Goettingen, Germany;
- Göttingen Graduate Center for Neurosciences, Biophysics and Molecular Biosciences, 37075 Goettingen, Germany
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”, 37075 Goettingen, Germany
| | - Christian Vogl
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Goettingen, 37075 Goettingen, Germany;
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”, 37075 Goettingen, Germany
| |
Collapse
|
44
|
Albergaria C, Silva NT, Darmohray DM, Carey MR. Cannabinoids modulate associative cerebellar learning via alterations in behavioral state. eLife 2020; 9:61821. [PMID: 33077026 PMCID: PMC7575324 DOI: 10.7554/elife.61821] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
Cannabinoids are notorious and profound modulators of behavioral state. In the brain, endocannabinoids act via Type 1-cannabinoid receptors (CB1) to modulate synaptic transmission and mediate multiple forms of synaptic plasticity. CB1 knockout (CB1KO) mice display a range of behavioral phenotypes, in particular hypoactivity and various deficits in learning and memory, including cerebellum-dependent delay eyeblink conditioning. Here we find that the apparent effects of CB1 deletion on cerebellar learning are not due to direct effects on CB1-dependent plasticity, but rather, arise as a secondary consequence of altered behavioral state. Hypoactivity of CB1KO mice accounts for their impaired eyeblink conditioning across both animals and trials. Moreover, learning in these mutants is rescued by walking on a motorized treadmill during training. Finally, cerebellar granule-cell-specific CB1KOs exhibit normal eyeblink conditioning, and both global and granule-cell-specific CB1KOs display normal cerebellum-dependent locomotor coordination and learning. These findings highlight the modulation of behavioral state as a powerful independent means through which individual genes contribute to complex behaviors.
Collapse
Affiliation(s)
- Catarina Albergaria
- Champalimaud Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - N Tatiana Silva
- Champalimaud Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Dana M Darmohray
- Champalimaud Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Megan R Carey
- Champalimaud Neuroscience Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| |
Collapse
|
45
|
Askow AT, McKenna CF, Box AG, Khan NA, Petruzzello SJ, De Lisio M, Phillips SM, Burd NA. Of Sound Mind and Body: Exploring the Diet-Strength Interaction in Healthy Aging. Front Nutr 2020; 7:145. [PMID: 32984401 PMCID: PMC7485317 DOI: 10.3389/fnut.2020.00145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/23/2020] [Indexed: 12/26/2022] Open
Abstract
Strength is a vital component of healthy aging. However, “strength” comes in different forms (includes both physical and mental aspects) and can look different at various phases of adult life. Healthy eating and regular exercise are clearly important pillars for strength. This paper proposes a framework that underlines the value of protein foods and resistance exercise for aging strong.
Collapse
Affiliation(s)
- Andrew T Askow
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Colleen F McKenna
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Allyson G Box
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Naiman A Khan
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Steven J Petruzzello
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Michael De Lisio
- Department of Cellular and Molecular Medicine, School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Nicholas A Burd
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
46
|
Neurotrophic factors and target-specific retrograde signaling interactions define the specificity of classical and neuropeptide cotransmitter release at identified Lymnaea synapses. Sci Rep 2020; 10:13526. [PMID: 32782285 PMCID: PMC7419297 DOI: 10.1038/s41598-020-70322-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/22/2020] [Indexed: 12/26/2022] Open
Abstract
Many neurons concurrently and/or differentially release multiple neurotransmitter substances to selectively modulate the activity of distinct postsynaptic targets within a network. However, the molecular mechanisms that produce synaptic heterogeneity by regulating the cotransmitter release characteristics of individual presynaptic terminals remain poorly defined. In particular, we know little about the regulation of neuropeptide corelease, despite the fact that they mediate synaptic transmission, plasticity and neuromodulation. Here, we report that an identified Lymnaea neuron selectively releases its classical small molecule and peptide neurotransmitters, acetylcholine and FMRFamide-derived neuropeptides, to differentially influence the activity of distinct postsynaptic targets that coordinate cardiorespiratory behaviour. Using a combination of electrophysiological, molecular, and pharmacological approaches, we found that neuropeptide cotransmitter release was regulated by cross-talk between extrinsic neurotrophic factor signaling and target-specific retrograde arachidonic acid signaling, which converged on modulation of glycogen synthase kinase 3. In this context, we identified a novel role for the Lymnaea synaptophysin homologue as a specific and synapse-delimited inhibitory regulator of peptide neurotransmitter release. This study is among the first to define the cellular and molecular mechanisms underlying the differential release of cotransmitter substances from individual presynaptic terminals, which allow for context-dependent tuning and plasticity of the synaptic networks underlying patterned motor behaviour.
Collapse
|
47
|
Rushton E, Kopke DL, Broadie K. Extracellular heparan sulfate proteoglycans and glycan-binding lectins orchestrate trans-synaptic signaling. J Cell Sci 2020; 133:133/15/jcs244186. [PMID: 32788209 DOI: 10.1242/jcs.244186] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The exceedingly narrow synaptic cleft (<20 nm) and adjacent perisynaptic extracellular space contain an astonishing array of secreted and membrane-anchored glycoproteins. A number of these extracellular molecules regulate intercellular trans-synaptic signaling by binding to ligands, acting as co-receptors or modulating ligand-receptor interactions. Recent work has greatly expanded our understanding of extracellular proteoglycan and glycan-binding lectin families as key regulators of intercellular signaling at the synapse. These secreted proteins act to regulate the compartmentalization of glycoprotein ligands and receptors, crosslink dynamic extracellular and cell surface lattices, modulate both exocytosis and endocytosis vesicle cycling, and control postsynaptic receptor trafficking. Here, we focus closely on the Drosophila glutamatergic neuromuscular junction (NMJ) as a model synapse for understanding extracellular roles of the many heparan sulfate proteoglycan (HSPG) and lectin proteins that help determine synaptic architecture and neurotransmission strength. We particularly concentrate on the roles of extracellular HSPGs and lectins in controlling trans-synaptic signaling, especially that mediated by the Wnt and BMP pathways. These signaling mechanisms are causally linked to a wide spectrum of neurological disease states that impair coordinated movement and cognitive functions.
Collapse
Affiliation(s)
- Emma Rushton
- Department of Biological Sciences, Brain Institute, and Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Danielle L Kopke
- Department of Biological Sciences, Brain Institute, and Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Brain Institute, and Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
48
|
|
49
|
Capogna M, Castillo PE, Maffei A. The ins and outs of inhibitory synaptic plasticity: Neuron types, molecular mechanisms and functional roles. Eur J Neurosci 2020; 54:6882-6901. [PMID: 32663353 DOI: 10.1111/ejn.14907] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/30/2020] [Accepted: 07/08/2020] [Indexed: 01/05/2023]
Abstract
GABAergic interneurons are highly diverse, and their synaptic outputs express various forms of plasticity. Compelling evidence indicates that activity-dependent changes of inhibitory synaptic transmission play a significant role in regulating neural circuits critically involved in learning and memory and circuit refinement. Here, we provide an updated overview of inhibitory synaptic plasticity with a focus on the hippocampus and neocortex. To illustrate the diversity of inhibitory interneurons, we discuss the case of two highly divergent interneuron types, parvalbumin-expressing basket cells and neurogliaform cells, which support unique roles on circuit dynamics. We also present recent progress on the molecular mechanisms underlying long-term, activity-dependent plasticity of fast inhibitory transmission. Lastly, we discuss the role of inhibitory synaptic plasticity in neuronal circuits' function. The emerging picture is that inhibitory synaptic transmission in the CNS is extremely diverse, undergoes various mechanistically distinct forms of plasticity and contributes to a much more refined computational role than initially thought. Both the remarkable diversity of inhibitory interneurons and the various forms of plasticity expressed by GABAergic synapses provide an amazingly rich inhibitory repertoire that is central to a variety of complex neural circuit functions, including memory.
Collapse
Affiliation(s)
- Marco Capogna
- Department of Biomedicine, Danish National Research Foundation Center of Excellence PROMEMO, Aarhus University, Aarhus, Denmark
| | - Pablo E Castillo
- Dominck P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Arianna Maffei
- Center for Neural Circuit Dynamics and Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
50
|
Molla HM, Tseng KY. Neural substrates underlying the negative impact of cannabinoid exposure during adolescence. Pharmacol Biochem Behav 2020; 195:172965. [PMID: 32526217 DOI: 10.1016/j.pbb.2020.172965] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/24/2020] [Accepted: 06/07/2020] [Indexed: 11/15/2022]
Abstract
As cannabinoid use among the adolescent population becomes widespread with recent legalizations, understanding more about its effects on the developing brain becomes increasingly important. Adolescent cannabinoid use has been shown to elicit both short and long lasting effects on cortical function, in part due to its impact on maturing brain regions including the prefrontal cortex and associated inputs. This paper provides an overview of current state of knowledge on the lasting impact of repeated cannabinoid exposure on behavior and associated neural circuits in adolescents compared to other age groups. Data obtained from human and rodent literature are integrated to discuss potential neural mechanisms underpinning the enduring negative impact of cannabinoid exposure during this sensitive period of brain development.
Collapse
Affiliation(s)
- Hanna M Molla
- Department of Anatomy and Cell Biology, University of Illinois at Chicago - College of Medicine, Chicago, IL, USA; Department of Cellular and Molecular Pharmacology, Rosalind Franklin University, North Chicago, IL, USA
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago - College of Medicine, Chicago, IL, USA.
| |
Collapse
|