1
|
Maal-Bared G, Yee M, Harding EK, Ghebreselassie M, Bergamini M, Choy R, Kim E, Di Vito S, Patel M, Amirzadeh M, Grieder TE, Coles BL, Nagy JI, Bonin RP, Steenland HW, van der Kooy D. Connexin-36-positive gap junctions in ventral tegmental area GABA neurons sustain opiate dependence. Eur J Neurosci 2024; 59:3422-3444. [PMID: 38679044 DOI: 10.1111/ejn.16366] [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] [Academic Contribution Register] [Received: 05/17/2023] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
Drug dependence is characterized by a switch in motivation wherein a positively reinforcing substance can become negatively reinforcing. Put differently, drug use can transform from a form of pleasure-seeking to a form of relief-seeking. Ventral tegmental area (VTA) GABA neurons form an anatomical point of divergence between two double dissociable pathways that have been shown to be functionally implicated and necessary for these respective motivations to seek drugs. The tegmental pedunculopontine nucleus (TPP) is necessary for opiate conditioned place preferences (CPP) in previously drug-naïve rats and mice, whereas dopaminergic (DA) transmission in the nucleus accumbens (NAc) is necessary for opiate CPP in opiate-dependent and withdrawn (ODW) rats and mice. Here, we show that this switch in functional anatomy is contingent upon the gap junction-forming protein, connexin-36 (Cx36), in VTA GABA neurons. Intra-VTA infusions of the Cx36 blocker, mefloquine, in ODW rats resulted in a reversion to a drug-naïve-like state wherein the TPP was necessary for opiate CPP and where opiate withdrawal aversions were lost. Consistent with these data, conditional knockout mice lacking Cx36 in GABA neurons (GAD65-Cre;Cx36 fl(CFP)/fl(CFP)) exhibited a perpetual drug-naïve-like state wherein opiate CPP was always DA independent, and opiate withdrawal aversions were absent even in mice subjected to an opiate dependence and withdrawal induction protocol. Further, viral-mediated rescue of Cx36 in VTA GABA neurons was sufficient to restore their susceptibility to an ODW state wherein opiate CPP was DA dependent. Our findings reveal a functional role for VTA gap junctions that has eluded prevailing circuit models of addiction.
Collapse
Affiliation(s)
- Geith Maal-Bared
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Mandy Yee
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Erika K Harding
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Martha Ghebreselassie
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Michael Bergamini
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Roxanne Choy
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Ethan Kim
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Stephanie Di Vito
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Maryam Patel
- Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Mohammadreza Amirzadeh
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Taryn E Grieder
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Brenda L Coles
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - James I Nagy
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robert P Bonin
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | | | - Derek van der Kooy
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Datta D, Perone I, Morozov YM, Arellano J, Duque A, Rakic P, van Dyck CH, Arnsten AFT. Localization of PDE4D, HCN1 channels, and mGluR3 in rhesus macaque entorhinal cortex may confer vulnerability in Alzheimer's disease. Cereb Cortex 2023; 33:11501-11516. [PMID: 37874022 PMCID: PMC10724870 DOI: 10.1093/cercor/bhad382] [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] [Academic Contribution Register] [Received: 05/01/2023] [Revised: 08/28/2023] [Accepted: 09/27/2023] [Indexed: 10/25/2023] Open
Abstract
Alzheimer's disease cortical tau pathology initiates in the layer II cell clusters of entorhinal cortex, but it is not known why these specific neurons are so vulnerable. Aging macaques exhibit the same qualitative pattern of tau pathology as humans, including initial pathology in layer II entorhinal cortex clusters, and thus can inform etiological factors driving selective vulnerability. Macaque data have already shown that susceptible neurons in dorsolateral prefrontal cortex express a "signature of flexibility" near glutamate synapses on spines, where cAMP-PKA magnification of calcium signaling opens nearby potassium and hyperpolarization-activated cyclic nucleotide-gated channels to dynamically alter synapse strength. This process is regulated by PDE4A/D, mGluR3, and calbindin, to prevent toxic calcium actions; regulatory actions that are lost with age/inflammation, leading to tau phosphorylation. The current study examined whether a similar "signature of flexibility" expresses in layer II entorhinal cortex, investigating the localization of PDE4D, mGluR3, and HCN1 channels. Results showed a similar pattern to dorsolateral prefrontal cortex, with PDE4D and mGluR3 positioned to regulate internal calcium release near glutamate synapses, and HCN1 channels concentrated on spines. As layer II entorhinal cortex stellate cells do not express calbindin, even when young, they may be particularly vulnerable to magnified calcium actions and ensuing tau pathology.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Isabella Perone
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yury M Morozov
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Jon Arellano
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Alvaro Duque
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Pasko Rakic
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | - Amy F T Arnsten
- Departments of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
3
|
Abstract
Gap junctions between neurons of the brain are thought to be present in only certain cell types, and they mostly connect dendrites, somata, and axons. Synapses with gap junctions serve bidirectional metabolic and electrical coupling between connected neuronal compartments. Although plasticity of electrical synapses has been described, recent evidence of the presence of silent, but activatable, gap junctions suggests that electrical nodes in a neuronal circuit can be added or suppressed by changes in the synaptic microenvironment. This opens the possibility of reconfiguration of neuronal ensembles in response to activity. Moreover, the coexistence of gap junctions in a glutamatergic synapse may add electric and metabolic coupling to a neuronal aggregate and may serve to constitute primed ensembles within a higher-order neural network. The interaction of chemical with electrical synapses should be further explored to find, especially, emerging properties of neuronal ensembles. It will be worth to reexamine in a new light the "functional" implications of the "anatomic" concepts: "continuity" and "contiguity," which were championed by Golgi and Ramón y Cajal, respectively. In any case, exploring the versatility of the gap junctions will likely enrich the heuristic aspects of the neural and network postulates.
Collapse
Affiliation(s)
- Rafael Gutiérrez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
4
|
Woo E, Datta D, Arnsten AFT. Glutamate Metabotropic Receptor Type 3 (mGlu3) Localization in the Rat Prelimbic Medial Prefrontal Cortex. Front Neuroanat 2022; 16:849937. [PMID: 35444520 PMCID: PMC9013768 DOI: 10.3389/fnana.2022.849937] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/06/2022] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Metabotropic glutamate receptors type 3 (mGlu3, encoded by GRM3) are increasingly related to cognitive functioning, including the working memory operations of the prefrontal cortex (PFC). In rhesus monkeys, mGlu3 are most commonly expressed on glia (36%), but are also very prominent on layer III dendritic spines (23%) in the dorsolateral PFC (dlPFC) where they enhance working memory-related neuronal firing. In contrast, mGlu2 are predominately presynaptic in layer III of macaque dlPFC, indicating a pre- vs. post-synaptic dissociation by receptor subtype. The current study examined the cellular and subcellular localizations of mGlu3 in the rat prelimbic medial PFC (PL mPFC), a region needed for spatial working memory performance in rodents. Multiple label immunofluorescence demonstrated mGlu3 expression in neurons and astrocytes, with rare labeling in microglia. Immunoelectron microscopy of layers III and V found that the predominant location for mGlu3 was on axons (layer III: 35.9%; layer V: 44.1%), with labeling especially prominent within the intervaricose segments distant from axon terminals. mGlu3 were also found on glia (likely astrocytes), throughout the glial membrane (layer III: 28.2%; layer V: 29.5%). Importantly, mGlu3 could be seen on dendritic spines, especially in layer III (layer III: 15.6%; layer V: 8.2%), with minor labeling on dendrites. These data show that there are some similarities between mGlu3 expression in rat PL mPFC and macaque dlPFC, but the spine expression enriches and differentiates in the more recently evolved primate dlPFC.
Collapse
|
5
|
Function and Plasticity of Electrical Synapses in the Mammalian Brain: Role of Non-Junctional Mechanisms. BIOLOGY 2022; 11:biology11010081. [PMID: 35053079 PMCID: PMC8773336 DOI: 10.3390/biology11010081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 12/13/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 01/27/2023]
Abstract
Simple Summary Relevant brain functions, such as perception, organization of behavior, and cognitive processes, are the outcome of information processing by neural circuits. Within these circuits, communication between neurons mainly relies on two modalities of synaptic transmission: chemical and electrical. Moreover, changes in the strength of these connections, aka synaptic plasticity, are believed to underlie processes of learning and memory, and its dysfunction has been suggested to underlie a variety of neurological disorders. While the relevance of chemical transmission and its plastic changes are known in great detail, analogous mechanisms and functional impact of their electrical counterparts were only recently acknowledged. In this article, we review the basic physical principles behind electrical transmission between neurons, the plethora of functional operations supported by this modality of neuron-to-neuron communication, as well as the basic principles of plasticity at these synapses. Abstract Electrical transmission between neurons is largely mediated by gap junctions. These junctions allow the direct flow of electric current between neurons, and in mammals, they are mostly composed of the protein connexin36. Circuits of electrically coupled neurons are widespread in these animals. Plus, experimental and theoretical evidence supports the notion that, beyond synchronicity, these circuits are able to perform sophisticated operations such as lateral excitation and inhibition, noise reduction, as well as the ability to selectively respond upon coincident excitatory inputs. Although once considered stereotyped and unmodifiable, we now know that electrical synapses are subject to modulation and, by reconfiguring neural circuits, these modulations can alter relevant operations. The strength of electrical synapses depends on the gap junction resistance, as well as on its functional interaction with the electrophysiological properties of coupled neurons. In particular, voltage and ligand gated channels of the non-synaptic membrane critically determine the efficacy of transmission at these contacts. Consistently, modulatory actions on these channels have been shown to represent relevant mechanisms of plasticity of electrical synaptic transmission. Here, we review recent evidence on the regulation of electrical synapses of mammals, the underlying molecular mechanisms, and the possible ways in which they affect circuit function.
Collapse
|
6
|
Datta D, Leslie SN, Woo E, Amancharla N, Elmansy A, Lepe M, Mecca AP, Slusher BS, Nairn AC, Arnsten AFT. Glutamate Carboxypeptidase II in Aging Rat Prefrontal Cortex Impairs Working Memory Performance. Front Aging Neurosci 2021; 13:760270. [PMID: 34867287 PMCID: PMC8634091 DOI: 10.3389/fnagi.2021.760270] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/17/2021] [Accepted: 09/28/2021] [Indexed: 11/29/2022] Open
Abstract
Glutamate carboxypeptidase II (GCPII) expression in brain is increased by inflammation, and reduces NAAG (N-acetyl aspartyl glutamate) stimulation of mGluR3 signaling. Genetic insults in this signaling cascade are increasingly linked to cognitive disorders in humans, where increased GCPII and or decreased NAAG-mGluR3 are associated with impaired prefrontal cortical (PFC) activation and cognitive impairment. As aging is associated with increased inflammation and PFC cognitive deficits, the current study examined GCPII and mGluR3 expression in the aging rat medial PFC, and tested whether GCPII inhibition with 2-(3-mercaptopropyl) pentanedioic acid (2-MPPA) would improve working memory performance. We found that GCPII protein was expressed on astrocytes and some microglia as expected from previous studies, but was also prominently expressed on neurons, and showed increased levels with advancing age. Systemic administration of the GCPII inhibitor, 2-MPPA, improved working memory performance in young and aged rats, and also improved performance after local infusion into the medial PFC. As GCPII inhibitors are well-tolerated, they may provide an important new direction for treatment of cognitive disorders associated with aging and/or inflammation.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Shannon N Leslie
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Elizabeth Woo
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Nishita Amancharla
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Ayah Elmansy
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Miguel Lepe
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Adam P Mecca
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Barbara S Slusher
- Department of Neurology and Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
7
|
Zeng X, Komanome Y, Kawasaki T, Inada K, Jonaitis J, Pulver SR, Kazama H, Nose A. An electrically coupled pioneer circuit enables motor development via proprioceptive feedback in Drosophila embryos. Curr Biol 2021; 31:5327-5340.e5. [PMID: 34666002 DOI: 10.1016/j.cub.2021.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2021] [Revised: 09/03/2021] [Accepted: 10/04/2021] [Indexed: 02/02/2023]
Abstract
Precocious movements are widely seen in embryos of various animal species. Whether such movements via proprioceptive feedback play instructive roles in motor development or are a mere reflection of activities in immature motor circuits is a long-standing question. Here we image the emerging motor activities in Drosophila embryos that lack proprioceptive feedback and show that proprioceptive experience is essential for the development of locomotor central pattern generators (CPGs). Downstream of proprioceptive inputs, we identify a pioneer premotor circuit composed of two pairs of segmental interneurons, whose gap-junctional transmission requires proprioceptive experience and plays a crucial role in CPG formation. The circuit autonomously generates rhythmic plateau potentials via IP3-mediated Ca2+ release from internal stores, which contribute to muscle contractions and hence produce proprioceptive feedback. Our findings demonstrate the importance of self-generated movements in instructing motor development and identify the cells, circuit, and physiology at the core of this proprioceptive feedback.
Collapse
Affiliation(s)
- Xiangsunze Zeng
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8561, Japan
| | - Yuko Komanome
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8561, Japan
| | - Tappei Kawasaki
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8561, Japan
| | - Kengo Inada
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Julius Jonaitis
- School of Psychology and Neuroscience, University of St Andrews, St Mary's Quad, South Street, St Andrews KY16 9JP, UK
| | - Stefan R Pulver
- School of Psychology and Neuroscience, University of St Andrews, St Mary's Quad, South Street, St Andrews KY16 9JP, UK
| | - Hokto Kazama
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan.
| | - Akinao Nose
- Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8561, Japan; Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
8
|
Bragg-Gonzalo L, De León Reyes NS, Nieto M. Genetic and activity dependent-mechanisms wiring the cortex: Two sides of the same coin. Semin Cell Dev Biol 2021; 118:24-34. [PMID: 34030948 DOI: 10.1016/j.semcdb.2021.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/26/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 01/17/2023]
Abstract
The cerebral cortex is responsible for the higher-order functions of the brain such as planning, cognition, or social behaviour. It provides us with the capacity to interact with and transform our world. The substrates of cortical functions are complex neural circuits that arise during development from the dynamic remodelling and progressive specialization of immature undefined networks. Here, we review the genetic and activity-dependent mechanisms of cortical wiring focussing on the importance of their interaction. Cortical circuits emerge from an initial set of neuronal types that engage in sequential forms of embryonic and postnatal activity. Such activities further complement the cells' genetic programs, increasing neuronal diversity and modifying the electrical properties while promoting selective connectivity. After a temporal window of enhanced plasticity, the main features of mature circuits are established. Failures in these processes can lead to neurodevelopmental disorders whose treatment remains elusive. However, a deeper dissection of cortical wiring will pave the way for innovative therapies.
Collapse
Affiliation(s)
- L Bragg-Gonzalo
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - N S De León Reyes
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain; Instituto de Neurociencias de Alicante, CSIC-UMH, 03550 San Juan de Alicante, Spain
| | - M Nieto
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain.
| |
Collapse
|
9
|
Abstract
Synaptic transmission is a fundamental neurobiological process by which neurons interact with each other and non-neuronal cells. It involves release of active substances from the presynaptic neuron onto receptive elements of postsynaptic cells, inducing waves of spreading electrochemical response. While much has been learned about the cellular and molecular mechanisms driving and governing transmitter release and sensing, the evolutionary origin of synaptic connections remains obscure. Herein, we review emerging evidence and concepts suggesting that key components of chemical synapse arose independently from neurons, in different functional and biological contexts, before the rise of multicellular living forms. We argue that throughout evolution, distinct synaptic constituents have been co-opted from ancestral forms for a new role in early metazoan, leading to the rise of chemical synapses and neurotransmission. Such a mosaic model of the origin of chemical synapses agrees with and supports the pluralistic hypothesis of evolutionary change.
Collapse
Affiliation(s)
- Saak V Ovsepian
- Institute for Biological and Medical Imaging, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg, Germany; International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland; Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic; Department of Psychiatry and Medical Psychology, 3rd Faculty of Medicine of Charles University, Prague, Czech Republic.
| | - Valerie B O'Leary
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská, Czech Republic
| | - Nikolai P Vesselkin
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia; Faculty of Medicine, The State University of Saint Petersburg, St. Petersburg, Russia
| |
Collapse
|
10
|
Cachope R, Pereda AE. Regulatory Roles of Metabotropic Glutamate Receptors on Synaptic Communication Mediated by Gap Junctions. Neuroscience 2020; 456:85-94. [PMID: 32619474 DOI: 10.1016/j.neuroscience.2020.06.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/11/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/18/2022]
Abstract
Variations of synaptic strength are thought to underlie forms of learning and can functionally reshape neural circuits. Metabotropic glutamate receptors play key roles in regulating the strength of chemical synapses. However, information within neural circuits is also conveyed via a second modality of transmission: gap junction-mediated synapses. We review here evidence indicating that metabotropic glutamate receptors also play important roles in the regulation of synaptic communication mediated by neuronal gap junctions, also known as 'electrical synapses'. Activity-driven interactions between metabotropic glutamate receptors and neuronal gap junctions can lead to long-term changes in the strength of electrical synapses. Further, the regulatory action of metabotropic glutamate receptors on neuronal gap junctions is not restricted to adulthood but is also of critical relevance during brain development and contributes to the pathological mechanisms that follow brain injury.
Collapse
Affiliation(s)
- Roger Cachope
- CHDI Foundation, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
11
|
Abstract
Electrical synaptic transmission via gap junctions underlies direct and rapid neuronal communication in the central nervous system. The diversity of functional roles played by electrical synapses is perhaps best exemplified in the vertebrate retina, in which gap junctions are expressed by each of the five major neuronal types. These junctions are highly plastic; they are dynamically regulated by ambient illumination and circadian rhythms acting through light-activated neuromodulators. The networks formed by electrically coupled neurons provide plastic, reconfigurable circuits positioned to play key and diverse roles in the transmission and processing of visual information at every retinal level. Recent work indicates gap junctions also play a role in the progressive cell death and aberrant activity seen in various pathological conditions of the retina. Gap junctions thus form potential targets for novel neuroprotective therapies in the treatment of neurodegenerative retinal diseases such as glaucoma and ischemic retinopathies.
Collapse
Affiliation(s)
- John O'Brien
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Stewart A. Bloomfield
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY 10036, USA
| |
Collapse
|
12
|
Abstract
Neurons communicate with each other via electrical or chemical synaptic connections. The pattern and strength of connections between neurons are critical for generating appropriate output. What mechanisms govern the formation of electrical and/or chemical synapses between two neurons? Recent studies indicate that common molecular players could regulate the formation of both of these classes of synapses. In addition, electrical and chemical synapses can mutually coregulate each other’s formation. Electrical activity, generated spontaneously by the nervous system or initiated from sensory experience, plays an important role in this process, leading to the selection of appropriate connections and the elimination of inappropriate ones. In this review, we discuss recent studies that shed light on the formation and developmental interactions of chemical and electrical synapses.
Collapse
Affiliation(s)
- Shaista Jabeen
- National Centre for Biological Sciences, Tata Institute for Fundamental Research , Bangalore , India.,Manipal Academy of Higher Education, Madhav Nagar, Manipal , India
| | - Vatsala Thirumalai
- National Centre for Biological Sciences, Tata Institute for Fundamental Research , Bangalore , India
| |
Collapse
|
13
|
Li J, Zhang S, Liu X, Han D, Xu J, Ma Y. Neuroprotective effects of leonurine against oxygen-glucose deprivation by targeting Cx36/CaMKII in PC12 cells. PLoS One 2018; 13:e0200705. [PMID: 30016355 PMCID: PMC6049927 DOI: 10.1371/journal.pone.0200705] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/12/2017] [Accepted: 07/02/2018] [Indexed: 01/31/2023] Open
Abstract
Leonurine has been reported to play an important role in ameliorating cognitive dysfunction, inhibiting ischemic stroke, and attenuating perihematomal edema and neuroinflammation in intracerebral hemorrhage. However, the exact mechanism and potential molecular targets of this effect remain unclear. Thus, in this study we investigated the neuroprotective effects of leonurine on hypoxia ischemia injury and explored the underlying mechanisms. An in vitro model of oxygen-glucose deprivation (OGD)-induced PC12 cells was established to mimic ischemic-like conditions. Cell viability, apoptosis, Cx36 and pCaMKII/CaMKII expression levels were evaluated after treatment with leonurine. The Cx36-selective antagonist mefloquine and CaMKII Inhibitor KN-93 were used to investigate the neuroprotective effect of leonurine on and the involvement of Cx36/CaMKII in this process. The results revealed that cell viability decreased and cell apoptosis and the protein expression of Cx36 and pCaMKII/CaMKII increased in the OGD-induced PC12 cells. Leonurine significantly increased cell viability and decreased cell apoptosis and the protein expression of Cx36 and pCaMKII/CaMKII in the OGD-induced PC12 cells. The specific inhibitor of Cx36 and CaMKII displayed similar protective effects. Moreover, the inhibition of Cx36 reduced pCaMKII levels and the ratio of pCaMKII/CaMKII in the OGD-induced PC12 cells, and vice versa. Taken together, these results suggest that leonurine might have a protective effect on OGD-induced PC12 cells through targeting the Cx36/CaMKII pathway. Thus, leonurine appears to have potential as a preventive or therapeutic drug against ischemic-induced neuronal injury.
Collapse
Affiliation(s)
- Jiao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shuang Zhang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoxi Liu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Deping Han
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jianqin Xu
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yunfei Ma
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
14
|
Belousov AB, Nishimune H, Denisova JV, Fontes JD. A potential role for neuronal connexin 36 in the pathogenesis of amyotrophic lateral sclerosis. Neurosci Lett 2018; 666:1-4. [PMID: 29246791 PMCID: PMC5805564 DOI: 10.1016/j.neulet.2017.12.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2017] [Revised: 12/04/2017] [Accepted: 12/11/2017] [Indexed: 11/26/2022]
Abstract
Neuronal gap junctional protein connexin 36 (Cx36) contributes to neuronal death following a range of acute brain insults such as ischemia, traumatic brain injury and epilepsy. Whether Cx36 contributes to neuronal death and pathological outcomes in chronic neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), is not known. We show here that the expression of Cx36 is significantly decreased in lumbar segments of the spinal cord of both human ALS subjects and SOD1G93A mice as compared to healthy human and wild-type mouse controls, respectively. In purified neuronal cultures prepared from the spinal cord of wild-type mice, knockdown of Cx36 reduces neuronal death caused by overexpression of the mutant human SOD1-G93A protein. Taken together, these data suggest a possible contribution of Cx36 to ALS pathogenesis. A perspective for the use of blockers of Cx36 gap junction channels for ALS therapy is discussed.
Collapse
Affiliation(s)
- Andrei B Belousov
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Hiroshi Nishimune
- Departments of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Janna V Denisova
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Joseph D Fontes
- Departments of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
15
|
Protein Kinase C Enhances Electrical Synaptic Transmission by Acting on Junctional and Postsynaptic Ca 2+ Currents. J Neurosci 2018; 38:2796-2808. [PMID: 29440551 DOI: 10.1523/jneurosci.2619-17.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/12/2017] [Revised: 01/15/2018] [Accepted: 02/02/2018] [Indexed: 11/21/2022] Open
Abstract
By synchronizing neuronal activity, electrical transmission influences the coordination, pattern, and/or frequency of firing. In the hemaphroditic marine-snail, Aplysia calfornica, the neuroendocrine bag cell neurons use electrical synapses to synchronize a 30 min afterdischarge of action potentials for the release of reproductive hormone. During the afterdischarge, protein kinase C (PKC) is activated, although its impact on bag cell neuron electrical transmission is unknown. This was investigated here by monitoring electrical synapses between paired cultured bag cell neurons using dual whole-cell recording. Voltage clamp revealed a largely voltage-independent junctional current, which was enhanced by treating with a PKC activator, PMA, before recording. We also examined the transfer of presynaptic action potential-like waveforms (generated in voltage clamp) to the postsynaptic cell (measured in current clamp). For control pairs, the presynaptic spike-like waveforms mainly evoked electrotonic potentials; however, when PKC was triggered, these stimuli consistently produced postsynaptic action potentials. To assess whether this involved changes to postsynaptic responsiveness, single bag cell neurons were injected with junctional-like current mimicking that evoked by a presynaptic action potential. Unlike control neurons, which were less likely to spike, cells in PMA always fired action potentials to the junctional-like current. Furthermore, PKC activation increased a postsynaptic voltage-gated Ca2+ current, which was recruited even by modest depolarization associated with an electrotonic potential. Whereas PKC inhibits gap junctions in most systems, bag cell neurons are rather unique, as the kinase potentiates the electrical synapse; in turn, this synergizes with augmented postsynaptic Ca2+ current to promote synchronous firing.SIGNIFICANCE STATEMENT Electrical coupling is a fundamental form of communication. For the bag cell neurons of Aplysia, electrical synapses coordinate a prolonged burst of action potentials known as the afterdischarge. We looked at how protein kinase C, which is upregulated with the afterdischarge, influences information transfer across the synapse. The kinase activation increased junctional current, a remarkable finding given that this enzyme is largely considered inhibitory for gap junctions. There was also an augmentation in the ability of a presynaptic neuron to provoke postsynaptic action potentials. This increased excitability was, in part, due to enhanced postsynaptic voltage-dependent Ca2+ current. Thus, protein kinase C improves the fidelity of electrotonic transmission and promotes synchronous firing by modulating both junctional and membrane conductances.
Collapse
|
16
|
Belousov AB, Fontes JD, Freitas-Andrade M, Naus CC. Gap junctions and hemichannels: communicating cell death in neurodevelopment and disease. BMC Cell Biol 2017; 18:4. [PMID: 28124625 PMCID: PMC5267333 DOI: 10.1186/s12860-016-0120-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/05/2023] Open
Abstract
Gap junctions are unique membrane channels that play a significant role in intercellular communication in the developing and mature central nervous system (CNS). These channels are composed of connexin proteins that oligomerize into hexamers to form connexons or hemichannels. Many different connexins are expressed in the CNS, with some specificity with regard to the cell types in which distinct connexins are found, as well as the timepoints when they are expressed in the developing and mature CNS. Both the main neuronal Cx36 and glial Cx43 play critical roles in neurodevelopment. These connexins also mediate distinct aspects of the CNS response to pathological conditions. An imbalance in the expression, translation, trafficking and turnover of connexins, as well as mutations of connexins, can impact their function in the context of cell death in neurodevelopment and disease. With the ever-increasing understanding of connexins in the brain, therapeutic strategies could be developed to target these membrane channels in various neurological disorders.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
| | - Joseph D Fontes
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
| | - Moises Freitas-Andrade
- Department of Cellular & Physiological Sciences, Faculty of Medicine, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Christian C Naus
- Department of Cellular & Physiological Sciences, Faculty of Medicine, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
17
|
Tu HY, Chiao CC. Cx36 expression in the AII-mediated rod pathway is activity dependent in the developing rabbit retina. Dev Neurobiol 2016; 76:473-86. [PMID: 26084632 DOI: 10.1002/dneu.22320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/21/2015] [Revised: 06/09/2015] [Accepted: 06/11/2015] [Indexed: 12/20/2022]
Abstract
Gap junctions are composed of connexin 36 (Cx36) and play a critical role in the rod photoreceptor signaling pathways of the vertebrate retina. Despite the fact that their connection and modulation in various rod pathways have been extensively studied in adult animals, little is known about the contribution and regulation of gap junctions to the development of the AII amacrine cell (AC)-mediated rod pathway. Using immunohistochemistry and microinjection, this study demonstrates a steady increase in relative Cx36 protein expression in both plexiform layers of the rabbit retina at around the time of eye opening. However, immediately after eye opening, most Cx36 immunoreactive AII ACs show no gap junction coupling pattern to neighboring cells and it is not until the third postnatal week that AII cells begin to exhibit an adult-like tracer-coupling pattern. Moreover, studies using dark-rearing and AMPA receptor blockade during postnatal development both revealed that relative levels of Cx36 immunoreactivity in AII ACs were increased when neural activity was inhibited. Our findings suggest that Cx36 expression in the AII-mediated rod pathway is activity dependent in the developing rabbit retina.
Collapse
Affiliation(s)
- Hung-Ya Tu
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chuan-Chin Chiao
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, 30013, Taiwan
| |
Collapse
|
18
|
Moore KB, O'Brien J. Connexins in neurons and glia: targets for intervention in disease and injury. Neural Regen Res 2015; 10:1013-7. [PMID: 26330808 PMCID: PMC4541216 DOI: 10.4103/1673-5374.160092] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 05/14/2015] [Indexed: 01/13/2023] Open
Abstract
Both neurons and glia throughout the central nervous system are organized into networks by gap junctions. Among glia, gap junctions facilitate metabolic homeostasis and intercellular communication. Among neurons, gap junctions form electrical synapses that function primarily for communication. However, in neurodegenerative states due to disease or injury gap junctions may be detrimental to survival. Electrical synapses may facilitate hyperactivity and bystander killing among neurons, while gap junction hemichannels in glia may facilitate inflammatory signaling and scar formation. Advances in understanding mechanisms of plasticity of electrical synapses and development of molecular therapeutics to target glial gap junctions and hemichannels offer new hope to pharmacologically limit neuronal degeneration and enhance recovery.
Collapse
Affiliation(s)
- Keith B Moore
- Richard S. Ruiz, M.D. Department of Ophthalmology & Visual Science, The University of Texas Health Science Center at Houston, TX, USA
| | - John O'Brien
- Richard S. Ruiz, M.D. Department of Ophthalmology & Visual Science, The University of Texas Health Science Center at Houston, TX, USA ; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
19
|
Voytenko LP, Lushnikova IV, Savotchenko AV, Isaeva EV, Skok MV, Lykhmus OY, Patseva MA, Skibo GG. Hippocampal GABAergic interneurons coexpressing alpha7-nicotinic receptors and connexin-36 are able to improve neuronal viability under oxygen-glucose deprivation. Brain Res 2015; 1616:134-45. [PMID: 25966616 DOI: 10.1016/j.brainres.2015.04.061] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/11/2014] [Revised: 02/10/2015] [Accepted: 04/30/2015] [Indexed: 01/09/2023]
Abstract
The hippocampal interneurons are very diverse by chemical profiles and rather inconsistent by sensitivity to CI. Some hippocampal GABAergic interneurons survive certain time after ischemia while ischemia-sensitive interneurons and pyramidal neurons are damaged. GABAergic signaling, nicotinic receptors expressing α7-subunit (α7nAChRs(+)) and connexin-36 (Cx36(+), electrotonic gapjunctions protein) contradictory modulate post-ischemic environment. We hypothesized that hippocampal ischemia-resistant GABAergic interneurons coexpressing glutamate decarboxylase-67 isoform (GAD67(+)), α7nAChRs(+), Cx36(+) are able to enhance neuronal viability. To check this hypothesis the histochemical and electrophysiological investigations have been performed using rat hippocampal organotypic culture in the condition of 30-min oxygen-glucose deprivation (OGD). Post-OGD reoxygenation (4h) revealed in CA1 pyramidal layer numerous damaged cells, decreased population spike amplitude and increased pair-pulse depression. In these conditions GAD67(+) interneurons displayed the OGD-resistance and significant increase of GABA synthesis/metabolism (GAD67-immunofluorescence, mitochondrial activity). The α7nAChRs(+) and Cx36(+) co-localizations were revealed in resistant GAD67(+) interneurons. Under OGD: GABAA-receptors (GABAARs) blockade increased cell damage and exacerbated the pair-pulse depression in CA1 pyramidal layer; α7nAChRs and Cx36-channels separate blockades sufficiently decreased cell damage while interneuronal GAD67-immunofluorescence and mitochondrial activity were similar to the control. Thus, hippocampal GABAergic interneurons co-expressing α7nAChRs and Cx36 remained resistant certain time after OGD and were able to modulate CA1 neuron survival through GABAARs, α7nAChRs and Cx36-channels activity. The enhancements of the neuronal viability together with GABA synthesis/metabolism normalization suggest cooperative neuroprotective mechanism that could be used for increase in efficiency of therapeutic strategies against post-ischemic pathology.
Collapse
Affiliation(s)
- L P Voytenko
- Department of Cytology, Bogomoletz Institute of Physiology, Kiev, Ukraine.
| | - I V Lushnikova
- Department of Cytology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - A V Savotchenko
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Ukraine
| | - E V Isaeva
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Ukraine
| | - M V Skok
- Palladin Institute of Biochemistry, Kiev, Ukraine
| | - O Yu Lykhmus
- Palladin Institute of Biochemistry, Kiev, Ukraine
| | - M A Patseva
- Department of Cytology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - G G Skibo
- Department of Cytology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| |
Collapse
|
20
|
Decrock E, De Bock M, Wang N, Bultynck G, Giaume C, Naus CC, Green CR, Leybaert L. Connexin and pannexin signaling pathways, an architectural blueprint for CNS physiology and pathology? Cell Mol Life Sci 2015; 72:2823-51. [PMID: 26118660 PMCID: PMC11113968 DOI: 10.1007/s00018-015-1962-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 02/06/2023]
Abstract
The central nervous system (CNS) is composed of a highly heterogeneous population of cells. Dynamic interactions between different compartments (neuronal, glial, and vascular systems) drive CNS function and allow to integrate and process information as well as to respond accordingly. Communication within this functional unit, coined the neuro-glio-vascular unit (NGVU), typically relies on two main mechanisms: direct cell-cell coupling via gap junction channels (GJCs) and paracrine communication via the extracellular compartment, two routes to which channels composed of transmembrane connexin (Cx) or pannexin (Panx) proteins can contribute. Multiple isoforms of both protein families are present in the CNS and each CNS cell type is characterized by a unique Cx/Panx portfolio. Over the last two decades, research has uncovered a multilevel platform via which Cxs and Panxs can influence different cellular functions within a tissue: (1) Cx GJCs enable a direct cell-cell communication of small molecules, (2) Cx hemichannels and Panx channels can contribute to autocrine/paracrine signaling pathways, and (3) different structural domains of these proteins allow for channel-independent functions, such as cell-cell adhesion, interactions with the cytoskeleton, and the activation of intracellular signaling pathways. In this paper, we discuss current knowledge on their multifaceted contribution to brain development and to specific processes in the NGVU, including synaptic transmission and plasticity, glial signaling, vasomotor control, and blood-brain barrier integrity in the mature CNS. By highlighting both physiological and pathological conditions, it becomes evident that Cxs and Panxs can play a dual role in the CNS and that an accurate fine-tuning of each signaling mechanism is crucial for normal CNS physiology.
Collapse
Affiliation(s)
- Elke Decrock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Marijke De Bock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Nan Wang
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, 75231 Paris Cedex 05, France
- University Pierre et Marie
Curie, ED, N°158, 75005 Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Colin R. Green
- Department of Ophthalmology, The University of Auckland, Auckland, New Zealand
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| |
Collapse
|
21
|
Fontes JD, Ramsey J, Polk JM, Koop A, Denisova JV, Belousov AB. Death of Neurons following Injury Requires Conductive Neuronal Gap Junction Channels but Not a Specific Connexin. PLoS One 2015; 10:e0125395. [PMID: 26017008 PMCID: PMC4446213 DOI: 10.1371/journal.pone.0125395] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/12/2014] [Accepted: 03/24/2015] [Indexed: 01/01/2023] Open
Abstract
Pharmacological blockade or genetic knockout of neuronal connexin 36 (Cx36)-containing gap junctions reduces neuronal death caused by ischemia, traumatic brain injury and NMDA receptor (NMDAR)-mediated excitotoxicity. However, whether Cx36 gap junctions contribute to neuronal death via channel-dependent or channel-independent mechanism remains an open question. To address this, we manipulated connexin protein expression via lentiviral transduction of mouse neuronal cortical cultures and analyzed neuronal death twenty-four hours following administration of NMDA (a model of NMDAR excitotoxicity) or oxygen-glucose deprivation (a model of ischemic injury). In cultures prepared from wild-type mice, over-expression and knockdown of Cx36-containing gap junctions augmented and prevented, respectively, neuronal death from NMDAR-mediated excitotoxicity and ischemia. In cultures obtained form from Cx36 knockout mice, re-expression of functional gap junction channels, containing either neuronal Cx36 or non-neuronal Cx43 or Cx31, resulted in increased neuronal death following insult. In contrast, the expression of communication-deficient gap junctions (containing mutated connexins) did not have this effect. Finally, the absence of ethidium bromide uptake in non-transduced wild-type neurons two hours following NMDAR excitotoxicity or ischemia suggested the absence of active endogenous hemichannels in those neurons. Taken together, these results suggest a role for neuronal gap junctions in cell death via a connexin type-independent mechanism that likely relies on channel activities of gap junctional complexes among neurons. A possible contribution of gap junction channel-permeable death signals in neuronal death is discussed.
Collapse
Affiliation(s)
- Joseph D. Fontes
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Jon Ramsey
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Jeremy M Polk
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Andre Koop
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Janna V. Denisova
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Andrei B. Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| |
Collapse
|
22
|
Ovsepian SV, Vesselkin NP. Wiring prior to firing: the evolutionary rise of electrical and chemical modes of synaptic transmission. Rev Neurosci 2015; 25:821-32. [PMID: 25051277 DOI: 10.1515/revneuro-2014-0037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/22/2014] [Accepted: 06/16/2014] [Indexed: 01/08/2023]
Abstract
Paracrine signaling and coupling via intercellular conduits are widely utilized for cell-cell interactions from primitive eukaryotes to advanced metazoa. Here, we review the functional and molecular data suggestive of a phylogenic continuum between these primeval forms of communication with the chemical and electrical synaptic transmission of neurons. We discuss selective evidence for the essential role played by the shift of function in early cellular morphologies and protosynaptic scaffolds, with their co-optation for new functionality, which ultimately lead to the rise of the chemical synapse. It is proposed that, rather than representing a transitional element, mixed electrochemical synapses exemplify an exaptive effect. The nonadaptive model of the synaptic origin described herein supports the pluralistic hypothesis of evolutionary change.
Collapse
|
23
|
|
24
|
Serrano-Velez JL, Rodriguez-Alvarado M, Torres-Vazquez II, Fraser SE, Yasumura T, Vanderpool KG, Rash JE, Rosa-Molinar E. Abundance of gap junctions at glutamatergic mixed synapses in adult Mosquitofish spinal cord neurons. Front Neural Circuits 2014; 8:66. [PMID: 25018700 PMCID: PMC4072101 DOI: 10.3389/fncir.2014.00066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2014] [Accepted: 05/28/2014] [Indexed: 11/13/2022] Open
Abstract
"Dye-coupling", whole-mount immunohistochemistry for gap junction channel protein connexin 35 (Cx35), and freeze-fracture replica immunogold labeling (FRIL) reveal an abundance of electrical synapses/gap junctions at glutamatergic mixed synapses in the 14th spinal segment that innervates the adult male gonopodium of Western Mosquitofish, Gambusia affinis (Mosquitofish). To study gap junctions' role in fast motor behavior, we used a minimally-invasive neural-tract-tracing technique to introduce gap junction-permeant or -impermeant dyes into deep muscles controlling the gonopodium of the adult male Mosquitofish, a teleost fish that rapidly transfers (complete in <20 mS) spermatozeugmata into the female reproductive tract. Dye-coupling in the 14th spinal segment controlling the gonopodium reveals coupling between motor neurons and a commissural primary ascending interneuron (CoPA IN) and shows that the 14th segment has an extensive and elaborate dendritic arbor and more gap junctions than do other segments. Whole-mount immunohistochemistry for Cx35 results confirm dye-coupling and show it occurs via gap junctions. Finally, FRIL shows that gap junctions are at mixed synapses and reveals that >50 of the 62 gap junctions at mixed synapses are in the 14th spinal segment. Our results support and extend studies showing gap junctions at mixed synapses in spinal cord segments involved in control of genital reflexes in rodents, and they suggest a link between mixed synapses and fast motor behavior. The findings provide a basis for studies of specific roles of spinal neurons in the generation/regulation of sex-specific behavior and for studies of gap junctions' role in regulating fast motor behavior. Finally, the CoPA IN provides a novel candidate neuron for future studies of gap junctions and neural control of fast motor behaviors.
Collapse
Affiliation(s)
| | | | | | - Scott E Fraser
- Molecular and Computational Biology Section, University of Southern California Los Angeles, CA, USA
| | - Thomas Yasumura
- Department of Biomedical Sciences, Colorado State University Fort Collins, CO, USA
| | | | - John E Rash
- Department of Biomedical Sciences, Colorado State University Fort Collins, CO, USA ; Program in Molecular, Cellular and Integrative Neurosciences, Colorado State University Fort Collins, CO, USA
| | - Eduardo Rosa-Molinar
- Biological Imaging Group, University of Puerto Rico San Juan, PR, USA ; Institute of Neurobiology, School of Medicine, University of Puerto Rico San Juan, PR, USA
| |
Collapse
|
25
|
Abstract
A wealth of research has revealed that electrical synapses in the central nervous system exhibit a high degree of plasticity. Several recent studies, particularly in the retina and inferior olive, highlight this plasticity. Three classes of mechanisms can alter electrical coupling over time courses ranging from milliseconds to days. Changes of membrane conductance through synaptic input or spiking activity shunt current and decouple neurons on the millisecond time scale. Such activity can also alter coupling symmetry, rectifying electrical synapses. More stable rectification can be accomplished through molecular asymmetry of the synapse itself. On the minutes time scale, changes in connexin phosphorylation can change coupling quasi-stably with an order of magnitude dynamic range. On the hours to days time scale, changes in expression level of connexins alter coupling through the course of circadian time, over developmental time, or in response to tissue injury. Combined, all of these mechanisms allow electrical coupling to be highly dynamic, changing in response to demands at the whole network level, in small portions of a network, or at the level of an individual synapse.
Collapse
|
26
|
Yu GX, Mueller M, Hawkins BE, Mathew BP, Parsley MA, Vergara LA, Hellmich HL, Prough DS, Dewitt DS. Traumatic brain injury in vivo and in vitro contributes to cerebral vascular dysfunction through impaired gap junction communication between vascular smooth muscle cells. J Neurotrauma 2014; 31:739-48. [PMID: 24341563 PMCID: PMC4047850 DOI: 10.1089/neu.2013.3187] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/04/2023] Open
Abstract
Gap junctions (GJs) contribute to cerebral vasodilation, vasoconstriction, and, perhaps, to vascular compensatory mechanisms, such as autoregulation. To explore the effects of traumatic brain injury (TBI) on vascular GJ communication, we assessed GJ coupling in A7r5 vascular smooth muscle (VSM) cells subjected to rapid stretch injury (RSI) in vitro and VSM in middle cerebral arteries (MCAs) harvested from rats subjected to fluid percussion TBI in vivo. Intercellular communication was evaluated by measuring fluorescence recovery after photobleaching (FRAP). In VSM cells in vitro, FRAP increased significantly (p<0.05 vs. sham RSI) after mild RSI, but decreased significantly (p<0.05 vs. sham RSI) after moderate or severe RSI. FRAP decreased significantly (p<0.05 vs. sham RSI) 30 min and 2 h, but increased significantly (p<0.05 vs. sham RSI) 24 h after RSI. In MCAs harvested from rats 30 min after moderate TBI in vivo, FRAP was reduced significantly (p<0.05), compared to MCAs from rats after sham TBI. In VSM cells in vitro, pretreatment with the peroxynitrite (ONOO(-)) scavenger, 5,10,15,20-tetrakis(4-sulfonatophenyl)prophyrinato iron[III], prevented RSI-induced reductions in FRAP. In isolated MCAs from rats treated with the ONOO(-) scavenger, penicillamine, GJ coupling was not impaired by fluid percussion TBI. In addition, penicillamine treatment improved vasodilatory responses to reduced intravascular pressure in MCAs harvested from rats subjected to moderate fluid percussion TBI. These results indicate that TBI reduced GJ coupling in VSM cells in vitro and in vivo through mechanisms related to generation of the potent oxidant, ONOO(-).
Collapse
Affiliation(s)
- Guang-Xiang Yu
- Charles R. Allen Research Laboratories, Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Brain function relies on the ability of neurons to communicate with each other. Interneuronal communication primarily takes place at synapses, where information from one neuron is rapidly conveyed to a second neuron. There are two main modalities of synaptic transmission: chemical and electrical. Far from functioning independently and serving unrelated functions, mounting evidence indicates that these two modalities of synaptic transmission closely interact, both during development and in the adult brain. Rather than conceiving synaptic transmission as either chemical or electrical, this article emphasizes the notion that synaptic transmission is both chemical and electrical, and that interactions between these two forms of interneuronal communication might be required for normal brain development and function.
Collapse
|
28
|
Prochnow N. Relevance of gap junctions and large pore channels in traumatic brain injury. Front Physiol 2014; 5:31. [PMID: 24575046 PMCID: PMC3920098 DOI: 10.3389/fphys.2014.00031] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/15/2013] [Accepted: 01/15/2014] [Indexed: 11/13/2022] Open
Abstract
In case of traumatic brain injury (TBI), occurrence of central nervous tissue damage is frequently aligned with local modulations of neuronal and glial gap junction channel expression levels. The degree of gap junctional protein expression and intercellular coupling efficiency, as well as hemichannel function has substantially impact on the course of trauma recovery and outcome. During TBI, gap junctions are especially involved in the intercellular molecule trafficking on repair of blood vessels and the regulation of vasomotor tone. Furthermore, gliosis and astrocytic swelling due to mechanical strain injury point out the consequences of derailed gap junction communication. This review addresses the outstanding role of gap junction channels in TBI pathophysiology and links the current state of results to applied clinical procedures as well as perspectives in acute and long-term treatment options.
Collapse
Affiliation(s)
- Nora Prochnow
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum Bochum, Germany
| |
Collapse
|
29
|
Neuronal gap junction coupling as the primary determinant of the extent of glutamate-mediated excitotoxicity. J Neural Transm (Vienna) 2013; 121:837-46. [PMID: 24178243 DOI: 10.1007/s00702-013-1109-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/11/2013] [Accepted: 10/16/2013] [Indexed: 01/12/2023]
Abstract
In the mammalian central nervous system (CNS), coupling of neurons by gap junctions (electrical synapses) increases during early postnatal development, then decreases, but increases in the mature CNS following neuronal injury, such as ischemia, traumatic brain injury and epilepsy. Glutamate-dependent neuronal death also occurs in the CNS during development and neuronal injury, i.e., at the time when neuronal gap junction coupling is increased. Here, we review our recent studies on regulation of neuronal gap junction coupling by glutamate in developing and injured neurons and on the role of gap junctions in neuronal cell death. A modified model of the mechanisms of glutamate-dependent neuronal death is discussed, which includes neuronal gap junction coupling as a critical part of these mechanisms.
Collapse
|
30
|
Anava S, Saad Y, Ayali A. The role of gap junction proteins in the development of neural network functional topology. INSECT MOLECULAR BIOLOGY 2013; 22:457-472. [PMID: 23782271 DOI: 10.1111/imb.12036] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/02/2023]
Abstract
Gap junctions (GJs) provide a common form of intercellular communication in most animal cells and tissues, from Hydra to human, including electrical synaptic signalling. Cell coupling via GJs has an important role in development in general, and in neural network development in particular. However, quantitative studies monitoring GJ proteins throughout nervous system development are few. Direct investigations demonstrating a role for GJ proteins by way of experimental manipulation of their expression are also rare. In the current work we focused on the role of invertebrate GJ proteins (innexins) in the in vitro development of neural network functional topology, using two-dimensional neural culture preparations derived from the frontal ganglion of the desert locust, Schistocerca gregaria. Immunocytochemistry and quantitative real-time PCR revealed a dynamic expression pattern of the innexins during development of the cultured networks. Changes were observed both in the levels and in the localization of expression. Down-regulating the expression of innexins, by using double-strand RNA for the first time in locust neural cultures, induced clear changes in network morphology, as well as inhibition of synaptogenesis, thus suggesting a role for GJs during the development of the functional topology of neuronal networks.
Collapse
Affiliation(s)
- S Anava
- Department of Zoology, Tel-Aviv University, Tel Aviv, Israel
| | | | | |
Collapse
|
31
|
Abstract
The presence of direct, cytoplasmatic, communication between neurons in the brain of vertebrates has been demonstrated a long time ago. These gap junctions have been characterized in many brain areas in terms of subunit composition, biophysical properties, neuronal connectivity patterns, and developmental regulation. Although interesting findings emerged, showing that different subunits are specifically regulated during development, or that excitatory and inhibitory neuronal networks exhibit various electrical connectivity patterns, gap junctions did not receive much further interest. Originally, it was believed that gap junctions represent simple passageways for electrical and biochemical coordination early in development. Today, we know that gap junction connectivity is tightly regulated, following independent developmental patterns for excitatory and inhibitory networks. Electrical connections are important for many specific functions of neurons, and are, for example, required for the development of neuronal stimulus tuning in the visual system. Here, we integrate the available data on neuronal connectivity and gap junction properties, as well as the most recent findings concerning the functional implications of electrical connections in the developing thalamus and neocortex.
Collapse
Affiliation(s)
- Dragos Niculescu
- Department of Synapse and Network Development, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Christian Lohmann
- Department of Synapse and Network Development, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
32
|
Belousov AB, Fontes JD. Neuronal gap junctions: making and breaking connections during development and injury. Trends Neurosci 2013; 36:227-36. [PMID: 23237660 PMCID: PMC3609876 DOI: 10.1016/j.tins.2012.11.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/22/2012] [Revised: 11/02/2012] [Accepted: 11/02/2012] [Indexed: 01/08/2023]
Abstract
In the mammalian central nervous system (CNS), coupling of neurons by gap junctions (i.e., electrical synapses) and the expression of the neuronal gap junction protein, connexin 36 (Cx36), transiently increase during early postnatal development. The levels of both subsequently decline and remain low in the adult, confined to specific subsets of neurons. However, following neuronal injury [such as ischemia, traumatic brain injury (TBI), and epilepsy], the coupling and expression of Cx36 rise. Here we summarize new findings on the mechanisms of regulation of Cx36-containing gap junctions in the developing and mature CNS and following injury. We also review recent studies suggesting various roles for neuronal gap junctions and in particular their role in glutamate-mediated neuronal death.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | |
Collapse
|
33
|
Gap junction channels and hemichannels in the CNS: regulation by signaling molecules. Neuropharmacology 2013; 75:567-82. [PMID: 23499663 DOI: 10.1016/j.neuropharm.2013.02.020] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/18/2012] [Revised: 02/08/2013] [Accepted: 02/26/2013] [Indexed: 11/20/2022]
Abstract
Coordinated interaction among cells is critical to develop the extremely complex and dynamic tasks performed by the central nervous system (CNS). Cell synchronization is in part mediated by connexins and pannexins; two different protein families that form gap junction channels and hemichannels. Whereas gap junction channels connect the cytoplasm of contacting cells and coordinate electric and metabolic activities, hemichannels communicate intra- and extra-cellular compartments and serve as diffusional pathways for ions and small molecules. Cells in the CNS depend on paracrine/autocrine communication via several extracellular signaling molecules, such as, cytokines, growth factors, transmitters and free radical species to sense changes in microenvironment as well as to adapt to them. These signaling molecules modulate crucial processes of the CNS, including, cellular migration and differentiation, synaptic transmission and plasticity, glial activation, cell viability and microvascular blood flow. Gap junction channels and hemichannels are affected by different signaling transduction pathways triggered by these paracrine/autocrine signaling molecules. Most of the modulatory effects induced by these signaling molecules are specific to the cell type and the connexin and pannexin subtype expressed in different brain areas. In this review, we summarized and discussed most of the relevant and recently published information on the effects of signaling molecules on connexin or pannexin based channels and their possible relevance in CNS physiology and pathology. This article is part of the Special Issue Section entitled 'Current Pharmacology of Gap Junction Channels and Hemichannels'.
Collapse
|
34
|
Kotak VC, Péndola LM, Rodríguez-Contreras A. Spontaneous activity in the developing gerbil auditory cortex in vivo involves GABAergic transmission. Neuroscience 2012; 226:130-44. [PMID: 22986170 DOI: 10.1016/j.neuroscience.2012.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/25/2011] [Revised: 09/04/2012] [Accepted: 09/04/2012] [Indexed: 10/27/2022]
Abstract
A salient feature of the developing brain is that spontaneous oscillations (SOs) and waves may influence the emergence of synaptic connections. While GABA produces depolarization and may support SOs in the neurons of developing rodents, it elicits hyperpolarization and diminishes SOs in developing gerbil auditory cortex (ACx). Therefore, we asked whether SOs exist in developing gerbil ACx in vivo and if GABAergic involvement can be manipulated. In vivo extracellular recordings in P3-5 ACx revealed SOs with longer burst durations and shorter inter-event intervals compared to ACx SOs in slices. ACx was then validated by gross anatomical features and lesions created at the in vivo recording site that corresponded with the electrophysiological coordinates of thalamorecipient ACx in slices. Further, NeuroVue Red, a lipophilic dye loaded at the in vivo recording sites, stained anatomically identifiable fiber tracks between the ACx and the auditory thalamus, medial geniculate body (MG). Separately, to chronically perturb GABAergic role in SOs, P2-5 pups were administered daily with GABA(A) receptor blocker, bicuculline (BIC). We then recorded from P14-17 ACx neurons in slices generated after hearing onset. ACx neurons from BIC-administered pups exhibited spontaneous action potentials in contrast to subthreshold synaptic potentials in neurons from sham-injected animals. Finally, to elucidate whether the gap junction blocker mefloquine (MFQ) previously shown to dampen ACx SOs in slices affected GABAergic transmission, MFQ was acutely applied in P3-5 slices while spontaneous inhibitory postsynaptic currents (sIPSCs) were recorded. Whereas MFQ increased the amplitude and frequency of sIPSCs in ACx neurons, the broad-spectrum gap junction blocker carbenoxolone decreased sIPSC amplitudes only. Together, we show that P2-5 gerbil ACx can endogenously generate SOs in vivo. Persistence of activity in ACx in P14-17 slices from pups administered with BIC at P2-5 implies that inhibitory GABAergic activity linked with gap-junction participates in the maturation of ACx.
Collapse
Affiliation(s)
- V C Kotak
- Center for Neural Science, 4 Washington Place, New York, NY 10003, USA.
| | | | | |
Collapse
|
35
|
Abstract
In the mammalian CNS, excessive release of glutamate and overactivation of glutamate receptors are responsible for the secondary (delayed) neuronal death following neuronal injury, including ischemia, traumatic brain injury (TBI) and epilepsy. The coupling of neurons by gap junctions (electrical synapses) increases during neuronal injury. In a recent study with the use of in vivo and in vitro models of cortical ischemia in mice, we have demonstrated that the ischemic increase in neuronal gap junction coupling is regulated by glutamate via group II metabotropic glutamate receptors (mGluR). Specifically, we found that activation of group II mGluRs increases background levels of neuronal gap junction coupling and expression of connexin 36 (Cx36; neuronal gap junction protein), whereas inactivation of group II mGluRs prevents the ischemia-mediated increases in the coupling and Cx36 expression. Using the analysis of neuronal death, we also established that inactivation of group II mGluRs or genetic elimination of Cx36 both dramatically reduce ischemic neuronal death in vitro and in vivo. Similar results were obtained using in vitro models of TBI and epilepsy. Our study demonstrated that mechanisms for the injury-mediated increase in neuronal gap junction coupling are part of the mechanisms for glutamate-dependent neuronal death.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
36
|
Belousov AB, Wang Y, Song JH, Denisova JV, Berman NE, Fontes JD. Neuronal gap junctions play a role in the secondary neuronal death following controlled cortical impact. Neurosci Lett 2012; 524:16-9. [PMID: 22781494 PMCID: PMC3414632 DOI: 10.1016/j.neulet.2012.06.065] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/12/2012] [Accepted: 06/25/2012] [Indexed: 11/24/2022]
Abstract
In the mammalian CNS, excessive release of glutamate and overactivation of glutamate receptors are responsible for the secondary (delayed) neuronal death following neuronal injury, including ischemia, traumatic brain injury (TBI) and epilepsy. Recent studies in mice showed a critical role for neuronal gap junctions in NMDA receptor-mediated excitotoxicity and ischemia-mediated neuronal death. Here, using controlled cortical impact (CCI) in adult mice, as a model of TBI, and Fluoro-Jade B staining for analysis of neuronal death, we set to determine whether neuronal gap junctions play a role in the CCI-mediated secondary neuronal death. We report that 24h post-CCI, substantial neuronal death is detected in a number of brain regions outside the injury core, including the striatum. The striatal neuronal death is reduced both in wild-type mice by systemic administration of mefloquine (a relatively selective blocker of neuronal gap junctions) and in knockout mice lacking connexin 36 (neuronal gap junction protein). It is also reduced by inactivation of group II metabotropic glutamate receptors (with LY341495) which, as reported previously, control the rapid increase in neuronal gap junction coupling following different types of neuronal injury. The results suggest that neuronal gap junctions play a critical role in the CCI-induced secondary neuronal death.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Belousov AB. Novel model for the mechanisms of glutamate-dependent excitotoxicity: role of neuronal gap junctions. Brain Res 2012; 1487:123-30. [PMID: 22771704 DOI: 10.1016/j.brainres.2012.05.063] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/23/2012] [Revised: 05/23/2012] [Accepted: 05/31/2012] [Indexed: 11/16/2022]
Abstract
In the mammalian central nervous system (CNS), coupling of neurons by gap junctions (electrical synapses) increases during early post-natal development, then decreases, but increases in the mature CNS following neuronal injury, such as ischemia, traumatic brain injury and epilepsy. Glutamate-dependent neuronal death also occurs in the CNS during development and neuronal injury, i.e., at the time when neuronal gap junction coupling is increased. Here, we review our recent studies on the regulation of neuronal gap junction coupling by glutamate during development and injury and on the role of gap junctions in neuronal cell death. A novel model of the mechanisms of glutamate-dependent neuronal death is discussed, which includes neuronal gap junction coupling as a critical part of these mechanisms.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
38
|
Pereda AE, Curti S, Hoge G, Cachope R, Flores CE, Rash JE. Gap junction-mediated electrical transmission: regulatory mechanisms and plasticity. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:134-46. [PMID: 22659675 DOI: 10.1016/j.bbamem.2012.05.026] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Academic Contribution Register] [Received: 02/24/2012] [Revised: 05/16/2012] [Accepted: 05/23/2012] [Indexed: 02/08/2023]
Abstract
The term synapse applies to cellular specializations that articulate the processing of information within neural circuits by providing a mechanism for the transfer of information between two different neurons. There are two main modalities of synaptic transmission: chemical and electrical. While most efforts have been dedicated to the understanding of the properties and modifiability of chemical transmission, less is still known regarding the plastic properties of electrical synapses, whose structural correlate is the gap junction. A wealth of data indicates that, rather than passive intercellular channels, electrical synapses are more dynamic and modifiable than was generally perceived. This article will discuss the factors determining the strength of electrical transmission and review current evidence demonstrating its dynamic properties. Like their chemical counterparts, electrical synapses can also be plastic and modifiable. This article is part of a Special Issue entitled: The Communicating junctions, roles and dysfunctions.
Collapse
Affiliation(s)
- Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Song JH, Wang Y, Fontes JD, Belousov AB. Regulation of connexin 36 expression during development. Neurosci Lett 2012; 513:17-9. [PMID: 22342304 PMCID: PMC3302950 DOI: 10.1016/j.neulet.2012.01.075] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/29/2011] [Accepted: 01/30/2012] [Indexed: 01/10/2023]
Abstract
In the mammalian CNS, the expression of neuronal gap junction protein, connexin 36 (Cx36), increases during the first 2 weeks of postnatal development and then decreases during the following 2 weeks. Recently we showed that the developmental increase in Cx36 expression is augmented by chronic (2 weeks) activation of group II metabotropic glutamate receptors (mGluR), prevented by chronic receptor inactivation, and the receptor-dependent increase in Cx36 expression is regulated via transcriptional control of the Cx36 gene activity. We demonstrate here that acute (60 min) activation of group II mGluRs in developing cortical neuronal cultures causes transient increase in Cx36 protein expression with decrease during the following 24h. However, there is no change in Cx36 mRNA expression. In addition, the data indicate that transient increase in Cx36 expression is due to new protein synthesis. The results suggest that, during development, acute activation of group II mGluRs causes up-regulation of Cx36 via post-transcriptional mechanisms. However, if the receptor activation is sustained, transcriptional activation of the Cx36 gene occurs.
Collapse
Affiliation(s)
- Ji-Hoon Song
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yongfu Wang
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Joseph D. Fontes
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Andrei B. Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
40
|
Wang Y, Song JH, Denisova JV, Park WM, Fontes JD, Belousov AB. Neuronal gap junction coupling is regulated by glutamate and plays critical role in cell death during neuronal injury. J Neurosci 2012; 32:713-25. [PMID: 22238107 PMCID: PMC3567463 DOI: 10.1523/jneurosci.3872-11.2012] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/28/2011] [Revised: 10/10/2011] [Accepted: 10/31/2011] [Indexed: 11/21/2022] Open
Abstract
In the mammalian CNS, excessive release of glutamate and overactivation of glutamate receptors are responsible for the secondary (delayed) neuronal death following neuronal injury, including ischemia, traumatic brain injury (TBI), and epilepsy. The coupling of neurons by gap junctions (electrical synapses) increases during neuronal injury. We report here that the ischemic increase in neuronal gap junction coupling is regulated by glutamate via group II metabotropic glutamate receptors (mGluRs). Specifically, using electrotonic coupling, Western blots, and siRNA in the mouse somatosensory cortex in vivo and in vitro, we demonstrate that activation of group II mGluRs increases background levels of neuronal gap junction coupling and expression of connexin 36 (Cx36) (neuronal gap junction protein), and inactivation of group II mGluRs prevents the ischemia-mediated increases in the coupling and Cx36 expression. We also show that the regulation is via cAMP/PKA (cAMP-dependent protein kinase)-dependent signaling and posttranscriptional control of Cx36 expression and that other glutamate receptors are not involved in these regulatory mechanisms. Furthermore, using the analysis of neuronal death, we show that inactivation of group II mGluRs or genetic elimination of Cx36 both dramatically reduce ischemia-mediated neuronal death in vitro and in vivo. Similar results are obtained using in vitro models of TBI and epilepsy. Our results indicate that neuronal gap junction coupling is a critical component of glutamate-dependent neuronal death. They also suggest that causal link among group II mGluR function, neuronal gap junction coupling, and neuronal death has a universal character and operates in different types of neuronal injuries.
Collapse
Affiliation(s)
- Yongfu Wang
- Departments of Molecular and Integrative Physiology and
| | - Ji-Hoon Song
- Departments of Molecular and Integrative Physiology and
| | | | - Won-Mee Park
- Departments of Molecular and Integrative Physiology and
| | - Joseph D. Fontes
- Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | | |
Collapse
|
41
|
Wang Y, Belousov AB. Deletion of neuronal gap junction protein connexin 36 impairs hippocampal LTP. Neurosci Lett 2011; 502:30-2. [PMID: 21798314 PMCID: PMC3159746 DOI: 10.1016/j.neulet.2011.07.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/09/2011] [Revised: 07/07/2011] [Accepted: 07/08/2011] [Indexed: 11/15/2022]
Abstract
In the mammalian CNS, deletion of neuronal gap junction protein, connexin 36 (Cx36), causes deficiencies in learning and memory. Here we tested whether Cx36 deletion affects the hippocampal long-term potentiation (LTP), which is considered as a cellular model of learning and memory mechanisms. We report that in acute slices of the hippocampal CA1 area, LTP is reduced in Cx36 knockout mice as compared to wild-type mice. Western blot analysis of NMDA receptor subunits indicates a higher NR2A/NR2B ratio in Cx36 knockout mice, indicating that there is shift in the threshold for LTP induction in knockout animals. Data suggest a possibility that learning and memory deficiencies in Cx36 knockout mice are due to deficiencies in LTP mechanisms.
Collapse
Affiliation(s)
- Yongfu Wang
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 2146 W. 39th Avenue, Kansas City, KS 66160, USA
| | | |
Collapse
|
42
|
Belousov AB. The regulation and role of neuronal gap junctions during development. Commun Integr Biol 2011; 4:579-81. [PMID: 22046468 DOI: 10.4161/cib.4.5.16380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/08/2011] [Accepted: 05/08/2011] [Indexed: 11/19/2022] Open
Abstract
Coupling of neurons by electrical synapses (gap junctions) transiently increases in the mammalian CNS during development and plays a role in a number of developmental events, including neuronal death. The coupling subsequently decreases and remains low in the adult, confined to specific subsets of neurons. In a recent study we have demonstrated that the developmental increase in neuronal gap junction coupling is regulated by the balance between the activity of two neurotransmitter receptors, group II metabotropic glutamate receptors (mGluR) and GABA(A) receptors. Specifically, we found that activation of group II mGluRs induces the developmental increases in neuronal gap junction coupling and expression of connexin 36 (Cx36; neuronal gap junction protein) and activation of GABA(A) receptors counteracts to these increases. We also established that the regulation by both neurotransmitter receptors is via a neuron-restrictive silencer element in the Cx36 gene promoter and the 3'-untranslated region of the Cx36 mRNA. Importantly, we demonstrated that mechanisms for the developmental increase in neuronal gap junction coupling directly control the death/survival mechanisms in developing neurons.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular and Integrative Physiology; University of Kansas Medical Center; Kansas City, KS USA
| |
Collapse
|