1
|
Zhang J, Li Y. Propofol-Induced Developmental Neurotoxicity: From Mechanisms to Therapeutic Strategies. ACS Chem Neurosci 2023; 14:1017-1032. [PMID: 36854650 DOI: 10.1021/acschemneuro.2c00755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
Propofol is the most commonly used intravenous general anesthetic in clinical anesthesia, and it is also widely used in general anesthesia for pregnant women and infants. Some clinical and preclinical studies have found that propofol causes damage to the immature nervous system, which may lead to neurodevelopmental disorders and cognitive dysfunction in infants and children. However, its potential molecular mechanism has not been fully elucidated. Recent in vivo and in vitro studies have found that some exogenous drugs and interventions can effectively alleviate propofol-induced neurotoxicity. In this review, we focus on the relevant preclinical studies and summarize the latest findings on the potential mechanisms and therapeutic strategies of propofol-induced developmental neurotoxicity.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao 266000, China.,Department of Medicine, Qingdao University, Qingdao 266000, China
| | - Yu Li
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
2
|
Kreis A, Issa F, Yerna X, Jabbour C, Schakman O, de Clippele M, Tajeddine N, Pierrot N, Octave JN, Gualdani R, Gailly P. Conditional deletion of KCC2 impairs synaptic plasticity and both spatial and nonspatial memory. Front Mol Neurosci 2023; 16:1081657. [PMID: 37168681 PMCID: PMC10164999 DOI: 10.3389/fnmol.2023.1081657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/31/2023] [Indexed: 05/13/2023] Open
Abstract
The postsynaptic inhibition through GABAA receptors (GABAAR) relies on two mechanisms, a shunting effect due to an increase in the postsynaptic membrane conductance and, in mature neurons, a hyperpolarization effect due to an entry of chloride into postsynaptic neurons. The second effect requires the action of the K+-Cl- cotransporter KCC2 which extrudes Cl- from the cell and maintains its cytosolic concentration very low. Neuronal chloride equilibrium seems to be dysregulated in several neurological and psychiatric conditions such as epilepsy, anxiety, schizophrenia, Down syndrome, or Alzheimer's disease. In the present study, we used the KCC2 Cre-lox knockdown system to investigate the role of KCC2 in synaptic plasticity and memory formation in adult mice. Tamoxifen-induced conditional deletion of KCC2 in glutamatergic neurons of the forebrain was performed at 3 months of age and resulted in spatial and nonspatial learning impairment. On brain slices, the stimulation of Schaffer collaterals by a theta burst induced long-term potentiation (LTP). The lack of KCC2 did not affect potentiation of field excitatory postsynaptic potentials (fEPSP) measured in the stratum radiatum (dendrites) but increased population spike (PS) amplitudes measured in the CA1 somatic layer, suggesting a reinforcement of the EPSP-PS potentiation, i.e., an increased ability of EPSPs to generate action potentials. At the cellular level, KCC2 deletion induced a positive shift in the reversal potential of GABAAR-driven Cl- currents (EGABA), suggesting an intracellular accumulation of chloride subsequent to the downregulation of KCC2. After treatment with bumetanide, an antagonist of the Na+-K+-Cl- cotransporter NKCC1, spatial memory impairment, chloride accumulation, and EPSP-PS potentiation were rescued in mice lacking KCC2. The presented results emphasize the importance of chloride equilibrium and GABA-inhibiting ability in synaptic plasticity and memory formation.
Collapse
|
3
|
Miles KD, Doll CA. Chloride imbalance in Fragile X syndrome. Front Neurosci 2022; 16:1008393. [PMID: 36312023 PMCID: PMC9596984 DOI: 10.3389/fnins.2022.1008393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022] Open
Abstract
Developmental changes in ionic balance are associated with crucial hallmarks in neural circuit formation, including changes in excitation and inhibition, neurogenesis, and synaptogenesis. Neuronal excitability is largely mediated by ionic concentrations inside and outside of the cell, and chloride (Cl-) ions are highly influential in early neurodevelopmental events. For example, γ-aminobutyric acid (GABA) is the main inhibitory neurotransmitter of the mature central nervous system (CNS). However, during early development GABA can depolarize target neurons, and GABAergic depolarization is implicated in crucial neurodevelopmental processes. This developmental shift of GABAergic neurotransmission from depolarizing to hyperpolarizing output is induced by changes in Cl- gradients, which are generated by the relative expression of Cl- transporters Nkcc1 and Kcc2. Interestingly, the GABA polarity shift is delayed in Fragile X syndrome (FXS) models; FXS is one of the most common heritable neurodevelopmental disorders. The RNA binding protein FMRP, encoded by the gene Fragile X Messenger Ribonucleoprotein-1 (Fmr1) and absent in FXS, appears to regulate chloride transporter expression. This could dramatically influence FXS phenotypes, as the syndrome is hypothesized to be rooted in defects in neural circuit development and imbalanced excitatory/inhibitory (E/I) neurotransmission. In this perspective, we summarize canonical Cl- transporter expression and investigate altered gene and protein expression of Nkcc1 and Kcc2 in FXS models. We then discuss interactions between Cl- transporters and neurotransmission complexes, and how these links could cause imbalances in inhibitory neurotransmission that may alter mature circuits. Finally, we highlight current therapeutic strategies and promising new directions in targeting Cl- transporter expression in FXS patients.
Collapse
Affiliation(s)
| | - Caleb Andrew Doll
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Children’s Hospital Colorado, Aurora, CO, United States
| |
Collapse
|
4
|
GABAergic and Glutamatergic Phenotypes of Neurons Expressing Calcium-Binding Proteins in the Preoptic Area of the Guinea Pig. Int J Mol Sci 2022; 23:ijms23147963. [PMID: 35887305 PMCID: PMC9320123 DOI: 10.3390/ijms23147963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
The mammalian preoptic area (POA) has large populations of calbindin (CB), calretinin (CR) and parvalbumin (PV) neurons, but phenotypes of these cells are unknown. Therefore, the question is whether neurons expressing CB, CR, and/or PV are GABAergic or glutamatergic. Double-immunofluorescence staining followed by epifluorescence and confocal microscopy was used to determine the coexpression patterns of CB, CR and PV expressing neurons with vesicular GABA transporters (VGAT) as specific markers of GABAergic neurons and vesicular glutamate transporters (VGLUT 2) as specific markers of glutamatergic neurons. The guinea pig was adopted as, like humans, it has a reproductive cycle with a true luteal phase and a long gestation period. The results demonstrated that in the guinea pig POA of both sexes, ~80% of CB+ and ~90% of CR+ neurons coexpress VGAT; however, one-fifth of CB+ neurons and one-third of CR+ cells coexpress VGLUT. About two-thirds of PV+ neurons express VGAT, and similar proportion of them coexpress VGLUT. Thus, many CB+, CR+ and PV+ neurons may be exclusively GABAergic (VGAT-expressing cells) or glutamatergic (VGLUT-expressing cells); however, at least a small fraction of CR+ cells and at least one-third of PV+ cells are likely neurons with a dual GABA/glutamate phenotype that may coexpress both transporters.
Collapse
|
5
|
Lee R, McGee A, Fernandez FX. Systematic review of drugs that modify the circadian system's phase-shifting responses to light exposure. Neuropsychopharmacology 2022; 47:866-879. [PMID: 34961774 PMCID: PMC8882192 DOI: 10.1038/s41386-021-01251-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/08/2021] [Accepted: 11/30/2021] [Indexed: 11/09/2022]
Abstract
We searched PubMed for primary research quantifying drug modification of light-induced circadian phase-shifting in rodents. This search, conducted for work published between 1960 and 2018, yielded a total of 146 papers reporting results from 901 studies. Relevant articles were those with any extractable data on phase resetting in wildtype (non-trait selected) rodents administered a drug, alongside a vehicle/control group, near or at the time of exposure. Most circadian pharmacology experiments were done using drugs thought to act directly on either the brain's central pacemaker, the suprachiasmatic nucleus (SCN), the SCN's primary relay, the retinohypothalamic tract, secondary pathways originating from the medial/dorsal raphe nuclei and intergeniculate leaflet, or the brain's sleep-arousal centers. While the neurotransmitter systems underlying these circuits were of particular interest, including those involving glutamate, gamma-aminobutyric acid, serotonin, and acetylcholine, other signaling modalities have also been assessed, including agonists and antagonists of receptors linked to dopamine, histamine, endocannabinoids, adenosine, opioids, and second-messenger pathways downstream of glutamate receptor activation. In an effort to identify drugs that unduly influence circadian responses to light, we quantified the net effects of each drug class by ratioing the size of the phase-shift observed after administration to that observed with vehicle in a given experiment. This allowed us to organize data across the literature, compare the relative efficacy of one mechanism versus another, and clarify which drugs might best suppress or potentiate phase resetting. Aggregation of the available data in this manner suggested that several candidates might be clinically relevant as auxiliary treatments to suppress ectopic light responses during shiftwork or amplify the circadian effects of timed bright light therapy. Future empirical research will be necessary to validate these possibilities.
Collapse
Affiliation(s)
- Robert Lee
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Austin McGee
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Fabian-Xosé Fernandez
- Department of Psychology, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- BIO5 and McKnight Brain Research Institutes, Tucson, AZ, USA.
| |
Collapse
|
6
|
Lodovichi C, Ratto GM, Trevelyan AJ, Arosio D. Genetically encoded sensors for Chloride concentration. J Neurosci Methods 2022; 368:109455. [PMID: 34952088 DOI: 10.1016/j.jneumeth.2021.109455] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 12/11/2021] [Accepted: 12/18/2021] [Indexed: 12/12/2022]
Abstract
Insights into chloride regulation in neurons have come slowly, but they are likely to be critical for our understanding of how the brain works. The reason is that the intracellular Cl- level ([Cl-]i) is the key determinant of synaptic inhibitory function, and this in turn dictates all manner of neuronal network function. The true impact on the network will only be apparent, however, if Cl- is measured at many locations at once (multiple neurons, and also across the subcellular compartments of single neurons), which realistically, can only be achieved using imaging. The development of genetically-encoded anion biosensors (GABs) brings the additional benefit that Cl- imaging may be done in identified cell-classes and hopefully in subcellular compartments. Here, we describe the historical background and motivation behind the development of these sensors and how they have been used so far. There are, however, still major limitations for their use, the most important being the fact that all GABs are sensitive to both pH and Cl-. Disambiguating the two signals has proved a major challenge, but there are potential solutions; notable among these is ClopHensor, which has now been developed for in vivo measurements of both ion species. We also speculate on how these biosensors may yet be improved, and how this could advance our understanding of Cl- regulation and its impact on brain function.
Collapse
Affiliation(s)
- Claudia Lodovichi
- Neuroscience Institute-CNR, Depart. Biomedical Sciences, Unipd, Padova, Veneto Institute of Molecular Medicine, Padova Neuroscience Center, Padova, Italy.
| | - Gian Michele Ratto
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore Pisa, 56127 Pisa, Italy
| | - Andrew J Trevelyan
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Daniele Arosio
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Biofisica, 38123 Trento, Italy.
| |
Collapse
|
7
|
Bohler MW, Chowdhury VS, Cline MA, Gilbert ER. Heat Stress Responses in Birds: A Review of the Neural Components. BIOLOGY 2021; 10:biology10111095. [PMID: 34827087 PMCID: PMC8614992 DOI: 10.3390/biology10111095] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 12/26/2022]
Abstract
Heat stress is one of the major environmental conditions causing significant losses in the poultry industry and having negative impacts on the world's food economy. Heat exposure causes several physiological impairments in birds, including oxidative stress, weight loss, immunosuppression, and dysregulated metabolism. Collectively, these lead not only to decreased production in the meat industry, but also decreases in the number of eggs laid by 20%, and overall loss due to mortality during housing and transit. Mitigation techniques have been discussed in depth, and include changes in air flow and dietary composition, improved building insulation, use of air cooling in livestock buildings (fogging systems, evaporation panels), and genetic alterations. Most commonly observed during heat exposure are reduced food intake and an increase in the stress response. However, very little has been explored regarding heat exposure, food intake and stress, and how the neural circuitry responsible for sensing temperatures mediate these responses. That thermoregulation, food intake, and the stress response are primarily mediated by the hypothalamus make it reasonable to assume that it is the central hub at which these systems interact and coordinately regulate downstream changes in metabolism. Thus, this review discusses the neural circuitry in birds associated with thermoregulation, food intake, and stress response at the level of the hypothalamus, with a focus on how these systems might interact in the presence of heat exposure.
Collapse
Affiliation(s)
- Mark W. Bohler
- Department of Animal and Poultry Sciences, 2160 Litton-Reaves Hall, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (M.W.B.); (M.A.C.)
| | - Vishwajit S. Chowdhury
- Laboratory of Stress Physiology and Metabolism, Faculty of Arts and Science, Kyushu University, Fukuoka 819-0395, Japan;
| | - Mark A. Cline
- Department of Animal and Poultry Sciences, 2160 Litton-Reaves Hall, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (M.W.B.); (M.A.C.)
| | - Elizabeth R. Gilbert
- Department of Animal and Poultry Sciences, 2160 Litton-Reaves Hall, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (M.W.B.); (M.A.C.)
- Correspondence: ; Tel.: +1-(540)-231-4750
| |
Collapse
|
8
|
Fueta Y, Hinoue M, Noguchi H, Matsuno M, Ishidao T, Garner CE, Hori H. Cerebral excitability in pup rats prenatally exposed to 1-bromopropane is suppressed by bromide accumulated in the brain. INDUSTRIAL HEALTH 2021; 59:239-248. [PMID: 34261823 PMCID: PMC8426544 DOI: 10.2486/indhealth.2020-0098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 04/13/2021] [Indexed: 06/13/2023]
Abstract
Previously, we reported that prenatal exposure to 1-bromopropane (1-BP) causes the accumulation of bromide (Br-) in the brain of rat pups. Here, we aimed to investigate the effects of Br- accumulation in rat pups prenatally exposed to 1-BP vapor. Dam rats were exposed to 1-BP (400 or 700 ppm; 1-BP group) by inhalation, or to NaBr (20 mM; Br- group) in drinking water during gestation days 1-20. We also analyzed pentylenetetrazole (PTZ, 60 mg/kg, ip)-induced behavioral changes in pups prenatally exposed to 1-BP or Br- on postnatal day (PND) 14. PTZ-induced epileptic convulsions were inhibited in both 1-BP (700 ppm) and Br- groups. The inhibition of neuronal excitability induced by Br- was evaluated electrophysiologically using the hippocampal slices obtained from PND14-16 pups. PTZ (2 mM) failed to induce epileptiform discharge in the presence of 1.2 mM Br- in the slices obtained from the control group. However, it induced epileptiform discharge following the removal of Br-, by perfusing artificial cerebrospinal fluid into the slices obtained from the Br- group. Our results indicate that Br- accumulates in the brain of neonatal rat pups prenatally exposed to 1-BP vapor suppressed neuronal excitability.
Collapse
Affiliation(s)
- Yukiko Fueta
- Department of Environmental Management and Control, School of Health Sciences, University of Occupational and Environmental Health, Japan
| | - Mitsuo Hinoue
- Department of Environmental Management and Control, School of Health Sciences, University of Occupational and Environmental Health, Japan
| | - Hazuki Noguchi
- Department of Environmental Management and Control, School of Health Sciences, University of Occupational and Environmental Health, Japan
| | - Mizuki Matsuno
- Department of Environmental Management and Control, School of Health Sciences, University of Occupational and Environmental Health, Japan
| | - Toru Ishidao
- Department of Environmental Management and Control, School of Health Sciences, University of Occupational and Environmental Health, Japan
| | - C Edwin Garner
- Pharmaceutical Development, Radikal Therapeutics, Inc., USA
| | - Hajime Hori
- Department of Environmental Management and Control, School of Health Sciences, University of Occupational and Environmental Health, Japan
| |
Collapse
|
9
|
Electrophysiology of ionotropic GABA receptors. Cell Mol Life Sci 2021; 78:5341-5370. [PMID: 34061215 PMCID: PMC8257536 DOI: 10.1007/s00018-021-03846-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/02/2021] [Accepted: 04/23/2021] [Indexed: 10/30/2022]
Abstract
GABAA receptors are ligand-gated chloride channels and ionotropic receptors of GABA, the main inhibitory neurotransmitter in vertebrates. In this review, we discuss the major and diverse roles GABAA receptors play in the regulation of neuronal communication and the functioning of the brain. GABAA receptors have complex electrophysiological properties that enable them to mediate different types of currents such as phasic and tonic inhibitory currents. Their activity is finely regulated by membrane voltage, phosphorylation and several ions. GABAA receptors are pentameric and are assembled from a diverse set of subunits. They are subdivided into numerous subtypes, which differ widely in expression patterns, distribution and electrical activity. Substantial variations in macroscopic neural behavior can emerge from minor differences in structure and molecular activity between subtypes. Therefore, the diversity of GABAA receptors widens the neuronal repertoire of responses to external signals and contributes to shaping the electrical activity of neurons and other cell types.
Collapse
|
10
|
Uchino K, Kawano H, Tanaka Y, Adaniya Y, Asahara A, Deshimaru M, Kubota K, Watanabe T, Katsurabayashi S, Iwasaki K, Hirose S. Inhibitory synaptic transmission is impaired at higher extracellular Ca 2+ concentrations in Scn1a +/- mouse model of Dravet syndrome. Sci Rep 2021; 11:10634. [PMID: 34017040 PMCID: PMC8137694 DOI: 10.1038/s41598-021-90224-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Dravet syndrome (DS) is an intractable form of childhood epilepsy that occurs in infancy. More than 80% of all patients have a heterozygous abnormality in the SCN1A gene, which encodes a subunit of Na+ channels in the brain. However, the detailed pathogenesis of DS remains unclear. This study investigated the synaptic pathogenesis of this disease in terms of excitatory/inhibitory balance using a mouse model of DS. We show that excitatory postsynaptic currents were similar between Scn1a knock-in neurons (Scn1a+/- neurons) and wild-type neurons, but inhibitory postsynaptic currents were significantly lower in Scn1a+/- neurons. Moreover, both the vesicular release probability and the number of inhibitory synapses were significantly lower in Scn1a+/- neurons compared with wild-type neurons. There was no proportional increase in inhibitory postsynaptic current amplitude in response to increased extracellular Ca2+ concentrations. Our study revealed that the number of inhibitory synapses is significantly reduced in Scn1a+/- neurons, while the sensitivity of inhibitory synapses to extracellular Ca2+ concentrations is markedly increased. These data suggest that Ca2+ tethering in inhibitory nerve terminals may be disturbed following the synaptic burst, likely leading to epileptic symptoms.
Collapse
Affiliation(s)
- Kouya Uchino
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Hiroyuki Kawano
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
| | - Yasuyoshi Tanaka
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
| | - Yuna Adaniya
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Ai Asahara
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Masanobu Deshimaru
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Takuya Watanabe
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shutaro Katsurabayashi
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan.
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shinichi Hirose
- Research Institute for the Molecular Pathogeneses of Epilepsy, Fukuoka University, Fukuoka, Japan
- Department of Pediatrics, School of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
11
|
The role of GABAergic signalling in neurodevelopmental disorders. Nat Rev Neurosci 2021; 22:290-307. [PMID: 33772226 PMCID: PMC9001156 DOI: 10.1038/s41583-021-00443-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
GABAergic inhibition shapes the connectivity, activity and plasticity of the brain. A series of exciting new discoveries provides compelling evidence that disruptions in a number of key facets of GABAergic inhibition have critical roles in the aetiology of neurodevelopmental disorders (NDDs). These facets include the generation, migration and survival of GABAergic neurons, the formation of GABAergic synapses and circuit connectivity, and the dynamic regulation of the efficacy of GABAergic signalling through neuronal chloride transporters. In this Review, we discuss recent work that elucidates the functions and dysfunctions of GABAergic signalling in health and disease, that uncovers the contribution of GABAergic neural circuit dysfunction to NDD aetiology and that leverages such mechanistic insights to advance precision medicine for the treatment of NDDs.
Collapse
|
12
|
Romagnoni A, Colonnese MT, Touboul JD, Gutkin BS. Progressive alignment of inhibitory and excitatory delay may drive a rapid developmental switch in cortical network dynamics. J Neurophysiol 2020; 123:1583-1599. [PMID: 32049596 DOI: 10.1152/jn.00402.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nervous system maturation occurs on multiple levels-synaptic, circuit, and network-at divergent timescales. For example, many synaptic properties mature gradually, whereas emergent network dynamics can change abruptly. Here we combine experimental and theoretical approaches to investigate a sudden transition in spontaneous and sensory evoked thalamocortical activity necessary for the development of vision. Inspired by in vivo measurements of timescales and amplitudes of synaptic currents, we extend the Wilson and Cowan model to take into account the relative onset timing and amplitudes of inhibitory and excitatory neural population responses. We study this system as these parameters are varied within amplitudes and timescales consistent with developmental observations to identify the bifurcations of the dynamics that might explain the network behaviors in vivo. Our findings indicate that the inhibitory timing is a critical determinant of thalamocortical activity maturation; a gradual decay of the ratio of inhibitory to excitatory onset time drives the system through a bifurcation that leads to a sudden switch of the network spontaneous activity from high-amplitude oscillations to a nonoscillatory active state. This switch also drives a change from a threshold bursting to linear response to transient stimuli, also consistent with in vivo observation. Thus we show that inhibitory timing is likely critical to the development of network dynamics and may underlie rapid changes in activity without similarly rapid changes in the underlying synaptic and cellular parameters.NEW & NOTEWORTHY Relying on a generalization of the Wilson-Cowan model, which allows a solid analytic foundation for the understanding of the link between maturation of inhibition and network dynamics, we propose a potential explanation for the role of developing excitatory/inhibitory synaptic delays in mediating a sudden switch in thalamocortical visual activity preceding vision onset.
Collapse
Affiliation(s)
- Alberto Romagnoni
- Group for Neural Theory, LNC INSERM Unité 960, Département d'Études Cognitives, École Normale Supérieure, PSL Research University, Paris, France.,Centre de recherche sur l'inflammation UMR 1149, INSERM-Université Paris Diderot, Paris, France.,Data Team, Département d'informatique de l'ENS, École Normale Supérieure, CNRS, PSL Research University, Paris, France
| | - Matthew T Colonnese
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia
| | - Jonathan D Touboul
- Department of Mathematics and Volen National Center for Complex Systems, Brandeis University, Waltham, Massachusetts
| | - Boris S Gutkin
- Group for Neural Theory, LNC INSERM Unité 960, Département d'Études Cognitives, École Normale Supérieure, PSL Research University, Paris, France.,Center for Cognition and Decision Making, Department of Psychology, NRU Higher School of Economics, Moscow, Russia
| |
Collapse
|
13
|
Role of adult-born granule cells in the hippocampal functions: Focus on the GluN2B-containing NMDA receptors. Eur Neuropsychopharmacol 2019; 29:1065-1082. [PMID: 31371103 DOI: 10.1016/j.euroneuro.2019.07.135] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/19/2019] [Accepted: 07/15/2019] [Indexed: 02/06/2023]
Abstract
Adult-born granule cells constitute a small subpopulation of the dentate gyrus (DG) in the hippocampus. However, they greatly influence several hippocampus-dependent behaviors, suggesting that adult-born granule cells have specific roles that influence behavior. In order to understand how exactly these adult-born granule cells contribute to behavior, it is critical to understand the underlying electrophysiology and neurochemistry of these cells. Here, this review simultaneously focuses on the specific electrophysiological properties of adult-born granule cells, relying on the GluN2B subunit of NMDA glutamate receptors, and how it influences neurochemistry throughout the brain. Especially in a critical age from 4 to 6 weeks post-division during which they modulate hippocampal functions, adult-born granule cells exhibit a higher intrinsic excitability and an enhanced long-term potentiation. Their stimulation decreases the overall excitation/inhibition balance of the DG via recruitment of local interneurons, and in the CA3 region of the hippocampus. However, the link between neurochemical effects of adult-born granule cells and behavior remain to be further examined.
Collapse
|
14
|
Ghali MGZ, Beshay S. Role of fast inhibitory synaptic transmission in neonatal respiratory rhythmogenesis and pattern formation. Mol Cell Neurosci 2019; 100:103400. [PMID: 31472222 DOI: 10.1016/j.mcn.2019.103400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/31/2019] [Accepted: 08/25/2019] [Indexed: 10/26/2022] Open
Abstract
Several studies have investigated the general role of chloride-based neurotransmission (GABAA and glycinergic signaling) in respiratory rhythmogenesis and pattern formation. In several brain regions, developmental alterations in these signaling pathways have been shown to be mediated by changes in cation-chloride cotransporter (CC) expression. For instance, CC expression changes during the course of neonatal development in medullary respiratory nuclei and other brain/spinal cord regions in a manner which decreases the cellular import, and increases the export, of chloride ions, shifting reversal potentials for chloride to progressively more negative values with maturation. In slice preparations of the same, this is related to an excitatory-to-inhibitory shift of GABAA- and glycinergic signaling. In medullary slices, GABAA-/glycinergic signaling in the early neonatal period is excitatory, becoming inhibitory over time. Additionally, blockade of the Na+/K+/2Cl- cotransporter, which imports these ions via secondary active transport, converts excitatory response to inhibitory ones. These effects have not yet been demonstrated at the individual respiratory-related neuron level to occur in intact (in vivo or in situ) animal preparations, which in contrast to slices, possess normal network connectivity and natural sources of tonic drive. Developmental changes in respiratory rhythm generating and pattern forming pontomedullary respiratory circuitry may contribute to critical periods, during which there exist increased risk for perinatal respiratory disturbances of central, obstructive, or hypoxia/hypercapnia-induced origin, including the sudden infant death syndrome. Thus, better characterizing the neurochemical maturation of the central respiratory network will enhance our understanding of these conditions, which will facilitate development of targeted therapies for respiratory disturbances in neonates and infants.
Collapse
Affiliation(s)
- Michael George Zaki Ghali
- Department of Neurological Surgery, Houston Methodist Hospital, Houston, TX 77030, United States of America.
| | - Sarah Beshay
- Department of Pulmonology and Critical Care Medicine, Houston Methodist Hospital, Houston, TX 77030, United States of America
| |
Collapse
|
15
|
Tang X, Drotar J, Li K, Clairmont CD, Brumm AS, Sullins AJ, Wu H, Liu XS, Wang J, Gray NS, Sur M, Jaenisch R. Pharmacological enhancement of KCC2 gene expression exerts therapeutic effects on human Rett syndrome neurons and Mecp2 mutant mice. Sci Transl Med 2019; 11:eaau0164. [PMID: 31366578 PMCID: PMC8140401 DOI: 10.1126/scitranslmed.aau0164] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 04/14/2019] [Accepted: 07/12/2019] [Indexed: 12/14/2022]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the methyl CpG binding protein 2 (MECP2) gene. There are currently no approved treatments for RTT. The expression of K+/Cl- cotransporter 2 (KCC2), a neuron-specific protein, has been found to be reduced in human RTT neurons and in RTT mouse models, suggesting that KCC2 might play a role in the pathophysiology of RTT. To develop neuron-based high-throughput screening (HTS) assays to identify chemical compounds that enhance the expression of the KCC2 gene, we report the generation of a robust high-throughput drug screening platform that allows for the rapid assessment of KCC2 gene expression in genome-edited human reporter neurons. From an unbiased screen of more than 900 small-molecule chemicals, we have identified a group of compounds that enhance KCC2 expression termed KCC2 expression-enhancing compounds (KEECs). The identified KEECs include U.S. Food and Drug Administration-approved drugs that are inhibitors of the fms-like tyrosine kinase 3 (FLT3) or glycogen synthase kinase 3β (GSK3β) pathways and activators of the sirtuin 1 (SIRT1) and transient receptor potential cation channel subfamily V member 1 (TRPV1) pathways. Treatment with hit compounds increased KCC2 expression in human wild-type (WT) and isogenic MECP2 mutant RTT neurons, and rescued electrophysiological and morphological abnormalities of RTT neurons. Injection of KEEC KW-2449 or piperine in Mecp2 mutant mice ameliorated disease-associated respiratory and locomotion phenotypes. The small-molecule compounds described in our study may have therapeutic effects not only in RTT but also in other neurological disorders involving dysregulation of KCC2.
Collapse
Affiliation(s)
- Xin Tang
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Jesse Drotar
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Keji Li
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | - Austin J Sullins
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hao Wu
- Fulcrum Therapeutics, Cambridge, MA 02139, USA
| | | | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Mriganka Sur
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
16
|
Wu Z, Huo Q, Ren L, Dong F, Feng M, Wang Y, Bai Y, Lüscher B, Li ST, Wang GL, Long C, Wang Y, Wu G, Chen G. Gluconate suppresses seizure activity in developing brains by inhibiting CLC-3 chloride channels. Mol Brain 2019; 12:50. [PMID: 31088565 PMCID: PMC6518791 DOI: 10.1186/s13041-019-0465-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/17/2019] [Indexed: 12/03/2022] Open
Abstract
Neonatal seizures are different from adult seizures, and many antiepileptic drugs that are effective in adults often fail to treat neonates. Here, we report that gluconate inhibits neonatal seizure by inhibiting CLC-3 chloride channels. We detect a voltage-dependent outward rectifying Cl− current mediated by CLC-3 Cl− channels in early developing brains but not adult mouse brains. Blocking CLC-3 Cl− channels by gluconate inhibits seizure activity both in neonatal brain slices and in neonatal animals with in vivo EEG recordings. Consistently, neonatal neurons of CLC-3 knockout mice lack the outward rectifying Cl− current and show reduced epileptiform activity upon stimulation. Mechanistically, we demonstrate that activation of CLC-3 Cl− channels alters intracellular Cl− homeostasis and enhances GABA excitatory activity. Our studies suggest that gluconate can suppress neonatal seizure activities through inhibiting CLC-3 Cl− channels in developing brains.
Collapse
Affiliation(s)
- Zheng Wu
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Qingwei Huo
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China.,South China Research Center for Acupuncture-Moxibustion, Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou Univ Chinese Med, Guangzhou, 510006, China
| | - Liang Ren
- Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Fengping Dong
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Mengyang Feng
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yue Wang
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yuting Bai
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Bernhard Lüscher
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Sheng-Tian Li
- Bio-X Institutes, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Guan-Lei Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Yun Wang
- Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Gangyi Wu
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.,School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Gong Chen
- Department of Biology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
17
|
Kubová H, Bendová Z, Moravcová S, Pačesová D, Rocha LL, Mareš P. Neonatal Clonazepam Administration Induces Long-Lasting Changes in Glutamate Receptors. Front Mol Neurosci 2018; 11:382. [PMID: 30364265 PMCID: PMC6193113 DOI: 10.3389/fnmol.2018.00382] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 09/24/2018] [Indexed: 11/13/2022] Open
Abstract
γ-aminobutyric acid (GABA) pathways play an important role in neuronal circuitry formation during early postnatal development. Our previous studies revealed an increased risk for adverse neurodevelopmental consequences in animals exposed to benzodiazepines, which enhance GABA inhibition via GABAA receptors. We reported that administration of the benzodiazepine clonazepam (CZP) during postnatal days 7-11 resulted in permanent behavioral alterations. However, the mechanisms underlying these changes are unknown. We hypothesized that early CZP exposure modifies development of glutamatergic receptors and their composition due to the tight developmental link between GABAergic functions and maturation of glutamatergic signaling. These changes may alter excitatory synapses, as well as neuronal connectivity and function of the neural network. We used quantitative real-time PCR and quantitative autoradiography to examine changes in NMDA and AMPA receptor composition and binding in response to CZP (1 mg/kg/day) administration for five consecutive days, beginning on P7. Brains were collected 48 h, 1 week, or 60 days after treatment cessation, and mRNA subunit expression was assessed in the hippocampus and sensorimotor cortex. A separate group of animals was used to determine binding to NMDA in different brain regions. Patterns of CZP-induced alterations in subunit mRNA expression were dependent on brain structure, interval after CZP cessation, and receptor subunit type. In the hippocampus, upregulation of GluN1, GluN3, and GluR2 subunit mRNA was observed at the 48-h interval, and GluN2A and GluR1 mRNA expression levels were higher 1 week after CZP cessation compared to controls, while GluN2B was downregulated. CZP exposure increased GluN3 and GluR2 subunit mRNA expression levels in the sensorimotor cortex 48 h after treatment cessation. GluA3 was higher 1 week after the CZP exposure, and GluN2A and GluA4 mRNA were significantly upregulated 2 months later. Expression of other subunits was not significantly different from that of the controls. NMDA receptor binding increased 1 week after the end of exposure in most hippocampal and cortical areas, including the sensorimotor cortex at the 48-h interval. CZP exposure decreased NMDA receptor binding in most evaluated hippocampal and cortical areas 2 months after the end of administration. Overall, early CZP exposure likely results in long-term glutamatergic receptor modulation that may affect synaptic development and function, potentially causing behavioral impairment.
Collapse
Affiliation(s)
- Hana Kubová
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Zdenka Bendová
- Faculty of Science, Charles University, Prague, Czechia.,National Institute of Mental Health, Klecany, Czechia
| | - Simona Moravcová
- Faculty of Science, Charles University, Prague, Czechia.,National Institute of Mental Health, Klecany, Czechia
| | - Dominika Pačesová
- Faculty of Science, Charles University, Prague, Czechia.,National Institute of Mental Health, Klecany, Czechia
| | - Luisa Lilia Rocha
- Pharmacobiology Department, Center of Research and Advanced Studies, Mexico City, Mexico
| | - Pavel Mareš
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czechia
| |
Collapse
|
18
|
Klett NJ, Allen CN. Intracellular Chloride Regulation in AVP+ and VIP+ Neurons of the Suprachiasmatic Nucleus. Sci Rep 2017; 7:10226. [PMID: 28860458 PMCID: PMC5579040 DOI: 10.1038/s41598-017-09778-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/28/2017] [Indexed: 12/15/2022] Open
Abstract
Several reports have described excitatory GABA transmission in the suprachiasmatic nucleus (SCN), the master pacemaker of circadian physiology. However, there is disagreement regarding the prevalence, timing, and neuronal location of excitatory GABA transmission in the SCN. Whether GABA is inhibitory or excitatory depends, in part, on the intracellular concentration of chloride ([Cl-]i). Here, using ratiometric Cl- imaging, we have investigated intracellular chloride regulation in AVP and VIP-expressing SCN neurons and found evidence suggesting that [Cl-]i is higher during the day than during the night in both AVP+ and VIP+ neurons. We then investigated the contribution of the cation chloride cotransporters to setting [Cl-]i in these SCN neurons and found that the chloride uptake transporter NKCC1 contributes to [Cl-]i regulation in SCN neurons, but that the KCCs are the primary regulators of [Cl-]i in SCN neurons. Interestingly, we observed that [Cl-]i is differentially regulated between AVP+ and VIP+ neurons-a low concentration of the loop diuretic bumetanide had differential effects on AVP+ and VIP+ neurons, while blocking the KCCs with VU0240551 had a larger effect on VIP+ neurons compared to AVP+ neurons.
Collapse
Affiliation(s)
- Nathan J Klett
- Neuroscience Graduate Program, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
- Oregon Institute for Occupational Health Sciences, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Charles N Allen
- Oregon Institute for Occupational Health Sciences, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
19
|
Malhotra A, Yosh E, Xiong M. Propofol's Effects on the Fetal Brain for Non-Obstetric Surgery. Brain Sci 2017; 7:brainsci7080107. [PMID: 28820429 PMCID: PMC5575627 DOI: 10.3390/brainsci7080107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/13/2017] [Accepted: 08/15/2017] [Indexed: 11/16/2022] Open
Abstract
While the use of Propofol has been increasing in usage for general surgical procedures since its release to market, there has been little work done on its potential link to neurotoxicity in humans. Only recently, following the release of a warning label from the United States Food and Drug Administration (USFDA) regarding a potential link to "neurotoxicity" in the neonate, did the surgical and anesthesiology communities become more aware of its potential for harm. Given the widespread use of this drug in clinical practice, the warning label naturally raised controversy regarding intrapartum Propofol usage. While intended to generate further studies, the lack of a viable anesthetic alternative raises issues regarding its current usage for surgical procedures in pregnant women. To answer the question whether current evidence is supportive of Propofol usage at its current levels in pregnant women, this review summarizes available evidence of fetal Propofol exposure in animal studies.
Collapse
Affiliation(s)
- Ajay Malhotra
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| | - Emily Yosh
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| | - Ming Xiong
- Department of Anesthesiology, New Jersey Medical School, Rutgers University, Newark, NJ 07107, USA.
| |
Collapse
|
20
|
Ogawa Y, Kanda T, Vogt K, Yanagisawa M. Anatomical and electrophysiological development of the hypothalamic orexin neurons from embryos to neonates. J Comp Neurol 2017; 525:3809-3820. [PMID: 28608460 DOI: 10.1002/cne.24261] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 04/19/2017] [Accepted: 04/19/2017] [Indexed: 02/01/2023]
Abstract
The amount, quality, and diurnal pattern of sleep change greatly during development. Developmental changes of sleep/wake architecture are in a close relationship to brain development. The fragmentation of wake episodes is one of the salient features in the neonatal period, which is also observed in mature animals and human individuals lacking neuropeptide orexin/hypocretin signaling. This raises the possibility that developmental changes of lateral hypothalamic orexin neurons are relevant to the development of sleep/wake architecture. However, little information is available on morphological and physiological features of developing orexin neurons. To address the cellular basis for maturation of the sleep/wake regulatory system, we investigated the functional development of orexin neurons in the lateral hypothalamus. The anatomical development as well as the changes in the electrophysiological characteristics of orexin neurons was examined from embryonic to postnatal stages in orexin-EGFP mice. Prepro-orexin promoter activity was detectable at embryonic day (E) 12.0, followed by expression of orexin A after E14.0. The number of orexin neurons and their membrane capacitance reached similar levels to adults by postnatal day (P) 7, while their membrane potentials, firing rates, and action potential waveforms were developed by P21. The hyperpolarizing effect of serotonin, which is a major inhibitory signal for adult orexin neurons, was detected after E18.0 and matured at P1. These results suggest that the expression of orexin peptides precedes the maturation of electrophysiological activity of orexin neurons. The function of orexin neurons gradually matures by 3 weeks after birth, coinciding with maturation of sleep/wake architecture.
Collapse
Affiliation(s)
- Yukino Ogawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takeshi Kanda
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kaspar Vogt
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
21
|
Recent Insights Into Molecular Mechanisms of Propofol-Induced Developmental Neurotoxicity: Implications for the Protective Strategies. Anesth Analg 2017; 123:1286-1296. [PMID: 27551735 DOI: 10.1213/ane.0000000000001544] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mounting evidence has demonstrated that general anesthetics could induce developmental neurotoxicity, including acute widespread neuronal cell death, followed by long-term memory and learning abnormalities. Propofol is a commonly used intravenous anesthetic agent for the induction and maintenance of anesthesia and procedural and critical care sedation in children. Compared with other anesthetic drugs, little information is available on its potential contributions to neurotoxicity. Growing evidence from multiple experimental models showed a similar neurotoxic effect of propofol as observed in other anesthetic drugs, raising serious concerns regarding pediatric propofol anesthesia. The aim of this review is to summarize the current findings of propofol-induced developmental neurotoxicity. We first present the evidence of neurotoxicity from animal models, animal cell culture, and human stem cell-derived neuron culture studies. We then discuss the mechanism of propofol-induced developmental neurotoxicity, such as increased cell death in neurons and oligodendrocytes, dysregulation of neurogenesis, abnormal dendritic development, and decreases in neurotrophic factor expression. Recent findings of complex mechanisms of propofol action, including alterations in microRNAs and mitochondrial fission, are discussed as well. An understanding of the toxic effect of propofol and the underlying mechanisms may help to develop effective novel protective or therapeutic strategies for avoiding the neurotoxicity in the developing human brain.
Collapse
|
22
|
Cote-Vélez A, Martínez Báez A, Lezama L, Uribe RM, Joseph-Bravo P, Charli JL. A screen for modulators reveals that orexin-A rapidly stimulates thyrotropin releasing hormone expression and release in hypothalamic cell culture. Neuropeptides 2017; 62:11-20. [PMID: 28173961 DOI: 10.1016/j.npep.2017.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 01/25/2017] [Accepted: 01/30/2017] [Indexed: 12/21/2022]
Abstract
In the paraventricular nucleus of the mammalian hypothalamus, hypophysiotropic thyrotropin releasing hormone (TRH) neurons integrate metabolic information and control the activity of the thyroid axis. Additional populations of TRH neurons reside in various hypothalamic areas, with poorly defined connections and functions, albeit there is evidence that some may be related to energy balance. To establish extracellular modulators of TRH hypothalamic neurons activity, we performed a screen of neurotransmitters effects in hypothalamic cultures. Cell culture conditions were chosen to facilitate the full differentiation of the TRH neurons; these conditions had permitted the characterization of the effects of known modulators of hypophysiotropic TRH neurons. The major end-point of the screen was Trh mRNA levels, since they are generally rapidly (0.5-3h) modified by synaptic inputs onto TRH neurons; in some experiments, TRH cell content or release was also analyzed. Various modulators, including histamine, serotonin, β-endorphin, met-enkephalin, and melanin concentrating hormone, had no effect. Glutamate, as well as ionotropic agonists (kainate and N-Methyl-d-aspartic acid), increased Trh mRNA levels. Baclofen, a GABAB receptor agonist, and dopamine enhanced Trh mRNA levels. An endocannabinoid receptor 1 inverse agonist promoted TRH release. Somatostatin increased Trh mRNA levels and TRH cell content. Orexin-A rapidly increased Trh mRNA levels, TRH cell content and release, while orexin-B decreased Trh mRNA levels. These data reveal unaccounted regulators, which exert potent effects on hypothalamic TRH neurons in vitro.
Collapse
Affiliation(s)
- Antonieta Cote-Vélez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mor. 62271, Mexico
| | - Anabel Martínez Báez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mor. 62271, Mexico
| | - Leticia Lezama
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mor. 62271, Mexico
| | - Rosa María Uribe
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mor. 62271, Mexico
| | - Patricia Joseph-Bravo
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mor. 62271, Mexico
| | - Jean-Louis Charli
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mor. 62271, Mexico.
| |
Collapse
|
23
|
The ontogenetic development of neurons containing calcium-binding proteins in the septum of the guinea pig: Late onset of parvalbumin immunoreactivity versus calbindin and calretinin. J Chem Neuroanat 2017; 79:22-31. [DOI: 10.1016/j.jchemneu.2016.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/07/2016] [Accepted: 10/18/2016] [Indexed: 10/20/2022]
|
24
|
Nakahata Y, Eto K, Murakoshi H, Watanabe M, Kuriu T, Hirata H, Moorhouse AJ, Ishibashi H, Nabekura J. Activation-Dependent Rapid Postsynaptic Clustering of Glycine Receptors in Mature Spinal Cord Neurons. eNeuro 2017; 4:ENEURO.0194-16.2017. [PMID: 28197549 PMCID: PMC5292596 DOI: 10.1523/eneuro.0194-16.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 01/05/2017] [Accepted: 01/17/2017] [Indexed: 12/02/2022] Open
Abstract
Inhibitory synapses are established during development but continue to be generated and modulated in strength in the mature nervous system. In the spinal cord and brainstem, presynaptically released inhibitory neurotransmitter dominantly switches from GABA to glycine during normal development in vivo. While presynaptic mechanisms of the shift of inhibitory neurotransmission are well investigated, the contribution of postsynaptic neurotransmitter receptors to this shift is not fully elucidated. Synaptic clustering of glycine receptors (GlyRs) is regulated by activation-dependent depolarization in early development. However, GlyR activation induces hyperpolarization after the first postnatal week, and little is known whether and how presynaptically released glycine regulates postsynaptic receptors in a depolarization-independent manner in mature developmental stage. Here we developed spinal cord neuronal culture of rodents using chronic strychnine application to investigate whether initial activation of GlyRs in mature stage could change postsynaptic localization of GlyRs. Immunocytochemical analyses demonstrate that chronic blockade of GlyR activation until mature developmental stage resulted in smaller clusters of postsynaptic GlyRs that could be enlarged upon receptor activation for 1 h in the mature stage. Furthermore, live cell-imaging techniques show that GlyR activation decreases its lateral diffusion at synapses, and this phenomenon is dependent on PKC, but neither Ca2+ nor CaMKII activity. These results suggest that the GlyR activation can regulate receptor diffusion and cluster size at inhibitory synapses in mature stage, providing not only new insights into the postsynaptic mechanism of shifting inhibitory neurotransmission but also the inhibitory synaptic plasticity in mature nervous system.
Collapse
Affiliation(s)
- Yoshihisa Nakahata
- Division of Homeostatic Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8585, Japan
| | - Kei Eto
- Division of Homeostatic Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8585, Japan
| | - Hideji Murakoshi
- Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8585, Japan
- Supportive Center for Brain Research, National Institute for Physiological Science, Okazaki 444-8585, Japan
- PRESTO, Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan
| | - Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Toshihiko Kuriu
- Department of Neurophysiology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 769-2193, Japan
| | - Hiromi Hirata
- PRESTO, Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima 411-8540, Japan
- Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima 411-8540, Japan
- Department of Chemistry and Biological Science, Graduate School of Science and Engineering, Aoyama Gakuin University, Sagamihara 252-5258, Japan
| | - Andrew J. Moorhouse
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney 2052, Australia
| | - Hitoshi Ishibashi
- Division of Homeostatic Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8585, Japan
- Department of Physiology, Kitasato University School of Allied Health Sciences, Sagamihara 252-0373, Japan
| | - Junichi Nabekura
- Division of Homeostatic Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8585, Japan
- CREST, Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan
| |
Collapse
|
25
|
Pratt KG, Hiramoto M, Cline HT. An Evolutionarily Conserved Mechanism for Activity-Dependent Visual Circuit Development. Front Neural Circuits 2016; 10:79. [PMID: 27818623 PMCID: PMC5073143 DOI: 10.3389/fncir.2016.00079] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/26/2016] [Indexed: 12/01/2022] Open
Abstract
Neural circuit development is an activity-dependent process. This activity can be spontaneous, such as the retinal waves that course across the mammalian embryonic retina, or it can be sensory-driven, such as the activation of retinal ganglion cells (RGCs) by visual stimuli. Whichever the source, neural activity provides essential instruction to the developing circuit. Indeed, experimentally altering activity has been shown to impact circuit development and function in many different ways and in many different model systems. In this review, we contemplate the idea that retinal waves in amniotes, the animals that develop either in ovo or utero (namely reptiles, birds and mammals) could be an evolutionary adaptation to life on land, and that the anamniotes, animals whose development is entirely external (namely the aquatic amphibians and fish), do not display retinal waves, most likely because they simply don’t need them. We then review what is known about the function of both retinal waves and visual stimuli on their respective downstream targets, and predict that the experience-dependent development of the tadpole visual system is a blueprint of what will be found in future studies of the effects of spontaneous retinal waves on instructing development of retinorecipient targets such as the superior colliculus (SC) and the lateral geniculate nucleus.
Collapse
Affiliation(s)
- Kara G Pratt
- Program in Neuroscience, Department of Zoology and Physiology, University of Wyoming Laramie, WY, USA
| | - Masaki Hiramoto
- Department of Molecular and Cellular Neuroscience and The Dorris Neuroscience Center, The Scripps Research Institute La Jolla, CA, USA
| | - Hollis T Cline
- Department of Molecular and Cellular Neuroscience and The Dorris Neuroscience Center, The Scripps Research Institute La Jolla, CA, USA
| |
Collapse
|
26
|
Marlin BJ, Froemke RC. Oxytocin modulation of neural circuits for social behavior. Dev Neurobiol 2016; 77:169-189. [PMID: 27626613 DOI: 10.1002/dneu.22452] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/07/2016] [Accepted: 09/12/2016] [Indexed: 01/04/2023]
Abstract
Oxytocin is a hypothalamic neuropeptide that has gained attention for the effects on social behavior. Recent findings shed new light on the mechanisms of oxytocin in synaptic plasticity and adaptively modifying neural circuits for social interactions such as conspecific recognition, pair bonding, and maternal care. Here, we review several of these newer studies on oxytocin in the context of previous findings, with an emphasis on social behavior and circuit plasticity in various brain regions shown to be enriched for oxytocin receptors. We provide a framework that highlights current circuit-level mechanisms underlying the widespread action of oxytocin. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 169-189, 2017.
Collapse
Affiliation(s)
- Bianca J Marlin
- Department of Neuroscience, Columbia University, New York, New York, 10032.,Howard Hughes Medical Institute, College of Physicians and Surgeons, Columbia University, New York, New York, 10032
| | - Robert C Froemke
- Department of Otolaryngology, Skirball Institute for Biomolecular Medicine, Neuroscience Institute, New York University School of Medicine, New York, New York.,Department of Neuroscience and Physiology Skirball Institute for Biomolecular Medicine, Neuroscience Institute New York University School of Medicine, New York, New York.,Center for Neural Science, New York University, New York, New York
| |
Collapse
|
27
|
Zhu S, Henninger K, McGrath BC, Cavener DR. PERK Regulates Working Memory and Protein Synthesis-Dependent Memory Flexibility. PLoS One 2016; 11:e0162766. [PMID: 27627766 PMCID: PMC5023101 DOI: 10.1371/journal.pone.0162766] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 08/09/2016] [Indexed: 11/30/2022] Open
Abstract
PERK (EIF2AK3) is an ER-resident eIF2α kinase required for memory flexibility and metabotropic glutamate receptor-dependent long-term depression, processes known to be dependent on new protein synthesis. Here we investigated PERK’s role in working memory, a cognitive ability that is independent of new protein synthesis, but instead is dependent on cellular Ca2+ dynamics. We found that working memory is impaired in forebrain-specific Perk knockout and pharmacologically PERK-inhibited mice. Moreover, inhibition of PERK in wild-type mice mimics the fear extinction impairment observed in forebrain-specific Perk knockout mice. Our findings reveal a novel role of PERK in cognitive functions and suggest that PERK regulates both Ca2+ -dependent working memory and protein synthesis-dependent memory flexibility.
Collapse
Affiliation(s)
- Siying Zhu
- Department of Biology, Center of Cellular Dynamics, the Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Keely Henninger
- Department of Biology, Center of Cellular Dynamics, the Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Barbara C McGrath
- Department of Biology, Center of Cellular Dynamics, the Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Douglas R Cavener
- Department of Biology, Center of Cellular Dynamics, the Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
28
|
Jaggi AS, Kaur A, Bali A, Singh N. Expanding Spectrum of Sodium Potassium Chloride Co-transporters in the Pathophysiology of Diseases. Curr Neuropharmacol 2016; 13:369-88. [PMID: 26411965 PMCID: PMC4812803 DOI: 10.2174/1570159x13666150205130359] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Sodium potassium chloride co-transporter (NKCC) belongs to cation-dependent chloride co-transporter family, whose activation allows the entry of Na(+), K(+) and 2Cl(-) inside the cell. It acts in concert with K(+) Cl(-) co-transporter (KCC), which extrudes K(+) and Cl(-) ions from cell. NKCC1 is widely distributed throughout the body, while NKCC2 is exclusively present in kidney. Protein kinase A, protein kinase C, Ste20-related proline-alanine-rich kinase, oxidative stress responsive kinases, With No K=lysine kinase and protein phosphatase type 1 control the phosphorylation/dephosphorylation of key threonine residues of in regulatory domain of NKCC1. The selective inhibitors of NKCC1 including bumetanide and furosemide are conventionally employed as diuretics. However, recent studies have indicated that NKCC1 may be involved in the pathophysiology of anxiety, cerebral ischemia, epilepsy, neuropathic pain, fragile X syndrome, autism and schizophrenia. The inhibitors of NKCC1 are shown to produce anxiolytic effects; attenuate cerebral ischemia-induced neuronal injury; produce antiepileptic effects and attenuate neuropathic pain. In the early developing brain, GABAA activation primarily produces excitatory actions due to high NKCC1/KCC2 ratio. However, as the development progresses, the ratio of NKCC1/KCC2 ratio reverses and there is switch in the polarity of GABAA actions and latter acquires the inhibitory actions. The recapitulation of developmental-like state during pathological state may be associated with increase in the expression and functioning of NKCC1, which decreases the strength of inhibitory GABAergic neurotransmission. The present review describes the expanding role and mechanism of NKCC1 in the pathophysiology of different diseases.
Collapse
Affiliation(s)
- Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala- 147002.
| | | | | | | |
Collapse
|
29
|
Harris JP, Struzyna LA, Murphy PL, Adewole DO, Kuo E, Cullen DK. Advanced biomaterial strategies to transplant preformed micro-tissue engineered neural networks into the brain. J Neural Eng 2016; 13:016019. [PMID: 26760138 DOI: 10.1088/1741-2560/13/1/016019] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Connectome disruption is a hallmark of many neurological diseases and trauma with no current strategies to restore lost long-distance axonal pathways in the brain. We are creating transplantable micro-tissue engineered neural networks (micro-TENNs), which are preformed constructs consisting of embedded neurons and long axonal tracts to integrate with the nervous system to physically reconstitute lost axonal pathways. APPROACH We advanced micro-tissue engineering techniques to generate micro-TENNs consisting of discrete populations of mature primary cerebral cortical neurons spanned by long axonal fascicles encased in miniature hydrogel micro-columns. Further, we improved the biomaterial encasement scheme by adding a thin layer of low viscosity carboxymethylcellulose (CMC) to enable needle-less insertion and rapid softening for mechanical similarity with brain tissue. MAIN RESULTS The engineered architecture of cortical micro-TENNs facilitated robust neuronal viability and axonal cytoarchitecture to at least 22 days in vitro. Micro-TENNs displayed discrete neuronal populations spanned by long axonal fasciculation throughout the core, thus mimicking the general systems-level anatomy of gray matter-white matter in the brain. Additionally, micro-columns with thin CMC-coating upon mild dehydration were able to withstand a force of 893 ± 457 mN before buckling, whereas a solid agarose cylinder of similar dimensions was predicted to withstand less than 150 μN of force. This thin CMC coating increased the stiffness by three orders of magnitude, enabling needle-less insertion into brain while significantly reducing the footprint of previous needle-based delivery methods to minimize insertion trauma. SIGNIFICANCE Our novel micro-TENNs are the first strategy designed for minimally invasive implantation to facilitate nervous system repair by simultaneously providing neuronal replacement and physical reconstruction of long-distance axon pathways in the brain. The micro-TENN approach may offer the ability to treat several disorders that disrupt the connectome, including Parkinson's disease, traumatic brain injury, stroke, and brain tumor excision.
Collapse
Affiliation(s)
- J P Harris
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, USA
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the cerebral cortex. GABAergic inhibition enables synchronization of activity in cortical networks, and contributes to generation of variety of brain activity patterns. In relation to epilepsy, GABAergic inhibition has been traditionally viewed as the main mechanism counterbalancing glutamatergic excitation and preventing hypersynchronous neuronal discharges. Indeed, deficits in GABAergic functions most commonly result in a hyperexcitable epileptic state, and many of the currently used antiepileptic drugs act through enhancement of GABAergic functions. However, a number of observations show that some epileptiform activity patterns involve synchronization by GABAergic mechanisms. These include two main categories that will be reviewed here: (1) synchronization of epileptiform oscillations based on GABAergic inhibition, and (2) epileptiform events driven by depolarizing and excitatory GABA. The conclusion is reached that GABAergic control of spike timing, either through inhibition or excitation under certain conditions, may work as a powerful synchronizing mechanism during epilepsy.
Collapse
Affiliation(s)
- Roustem Khazipov
- INMED-INSERM U901, 13273 Marseille Cedex 09, France Aix-Marseille University, 13273 Marseille Cedex 09, France Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
31
|
KCC2 rescues functional deficits in human neurons derived from patients with Rett syndrome. Proc Natl Acad Sci U S A 2016; 113:751-6. [PMID: 26733678 DOI: 10.1073/pnas.1524013113] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Rett syndrome is a severe form of autism spectrum disorder, mainly caused by mutations of a single gene methyl CpG binding protein 2 (MeCP2) on the X chromosome. Patients with Rett syndrome exhibit a period of normal development followed by regression of brain function and the emergence of autistic behaviors. However, the mechanism behind the delayed onset of symptoms is largely unknown. Here we demonstrate that neuron-specific K(+)-Cl(-) cotransporter2 (KCC2) is a critical downstream gene target of MeCP2. We found that human neurons differentiated from induced pluripotent stem cells from patients with Rett syndrome showed a significant deficit in KCC2 expression and consequently a delayed GABA functional switch from excitation to inhibition. Interestingly, overexpression of KCC2 in MeCP2-deficient neurons rescued GABA functional deficits, suggesting an important role of KCC2 in Rett syndrome. We further identified that RE1-silencing transcriptional factor, REST, a neuronal gene repressor, mediates the MeCP2 regulation of KCC2. Because KCC2 is a slow onset molecule with expression level reaching maximum later in development, the functional deficit of KCC2 may offer an explanation for the delayed onset of Rett symptoms. Our studies suggest that restoring KCC2 function in Rett neurons may lead to a potential treatment for Rett syndrome.
Collapse
|
32
|
Chang HK, Kim KH, Kang KW, Kang YJ, Kim TW, Park HK, Kim SE, Kim CJ. Antidepressants modulate glycine action in rat hippocampus. J Exerc Rehabil 2015; 11:311-9. [PMID: 26730381 PMCID: PMC4697779 DOI: 10.12965/jer.150263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/13/2015] [Indexed: 11/22/2022] Open
Abstract
Antidepressants are drugs that relieve symptoms of depressive disorders. Fluoxetine, tianeptine, and milnacipran are different types of antidepressants, and they have widely been used for relieving of depression symptoms. In the present study, the effects of fluoxetine, tianeptine, and milnacipran on the glycine-induced ion current by nystatin-perforated patch clamp and on the amplitude of field potential in the hippocampal CA1 region by multichannel extracellular recording, MED64, system, were studied. In the present results, fluoxetine, tianeptine, and milnacipran reduced glycine-induced ion current in the hippocampal CA1 neurons in nystatin-perforated patch clamp method. These drugs enhanced the amplitude of the field potential in the hippocampal CA1 region in MED64 system. These results suggest that antidepressants may increase neuronal activity by enhancing field potential through inhibition on glycine-induced ion current.
Collapse
Affiliation(s)
- Hyun-Kyung Chang
- Department of Urology, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Korea
| | - Khae Hawn Kim
- Department of Urology, Gachon University Gil Medical Center, Gachon University School of Medicine, Incheon, Korea
| | - Ki-Woon Kang
- Department of Anesthesiology and Pain Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Yoo-Jin Kang
- Department of Anesthesiology and Pain Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Tae-Wook Kim
- Department of Community Sport and Recreation, College of Health Science, Jangan University, Hwasung, Korea
| | - Hun-Kyung Park
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sung-Eun Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
33
|
Glykys J, Staley KJ. Diazepam effect during early neonatal development correlates with neuronal Cl(.). Ann Clin Transl Neurol 2015; 2:1055-70. [PMID: 26734658 PMCID: PMC4693588 DOI: 10.1002/acn3.259] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 09/16/2015] [Indexed: 11/09/2022] Open
Abstract
Objective Although benzodiazepines and other GABAA receptors allosteric modulators are used to treat neonatal seizures, their efficacy may derive from actions on subcortical structures. Side effects of benzodiazepines in nonseizing human neonates include myoclonus, seizures, and abnormal movements. Excitatory actions of GABA may underlie both side effects and reduced anticonvulsant activity of benzodiazepines. Neocortical organotypic slice cultures were used to study: (1) spontaneous cortical epileptiform activity during early development; (2) developmental changes in [Cl−]i and (3) whether diazepam's anticonvulsant effect correlated with neuronal [Cl−]i. Methods Epileptiform activity in neocortical organotypic slice cultures was measured by field potential recordings. Cl− changes during development were assessed by multiphoton imaging of neurons transgenically expressing a Cl‐sensitive fluorophore. Clinically relevant concentrations of diazepam were used to test the anticonvulsant effectiveness at ages corresponding to premature neonates through early infancy. Results (1) Neocortical organotypic slices at days in vitro 5 (DIV5) exhibited spontaneous epileptiform activity. (2) Epileptiform event duration decreased with age. (3) There was a progressive decrease in [Cl−]i over the same age range. (4) Diazepam was ineffective in decreasing epileptiform activity at DIV5‐6, but progressively more effective at older ages through DIV15. (5) At DIV5‐6, diazepam worsened epileptiform activity in 50% of the slices. Interpretation The neocortical organotypic slice is a useful model to study spontaneous epileptiform activity. Decreasing [Cl−]i during development correlates with decreasing duration of spontaneous epileptiform activity and increasing anticonvulsant efficacy of diazepam. We provide a potential explanation for the reports of seizures and myoclonus induction by benzodiazepines in newborn human neonates and the limited electrographic efficacy of benzodiazepines for the treatment of neonatal seizures.
Collapse
Affiliation(s)
- Joseph Glykys
- Department of Neurology Massachusetts General Hospital Boston Massachusetts; Harvard Medical School Boston Massachusetts
| | - Kevin J Staley
- Department of Neurology Massachusetts General Hospital Boston Massachusetts; Harvard Medical School Boston Massachusetts
| |
Collapse
|
34
|
Abstract
Neurons coexpressing neuropeptide Y, agouti-related peptide, and GABA (NAG) play an important role in ingestive behavior and are located in the arcuate nucleus of the hypothalamus. NAG neurons receive both GABAergic and glutamatergic synaptic inputs, however, the developmental time course of synaptic input organization of NAG neurons in mice is unknown. In this study, we show that these neurons have low numbers of GABAergic synapses and that GABA is inhibitory to NAG neurons during early postnatal period. In contrast, glutamatergic inputs onto NAG neurons are relatively abundant by P13 and are comparatively similar to the levels observed in the adult. As mice reach adulthood (9-10 weeks), GABAergic tone onto NAG neurons increases. At this age, NAG neurons received similar numbers of inhibitory and EPSCs. To further differentiate age-associated changes in synaptic distribution, 17- to 18-week-old lean and diet-induced obesity (DIO) mice were studied. Surprisingly, NAG neurons from lean adult mice exhibit a reduction in the GABAergic synapses compared with younger adults. Conversely, DIO mice display reductions in the number of GABAergic and glutamatergic inputs onto NAG neurons. Based on these experiments, we propose that synaptic distribution in NAG neurons is continuously restructuring throughout development to accommodate the animals' energy requirements.
Collapse
|
35
|
Gackière F, Vinay L. Contribution of the potassium-chloride cotransporter KCC2 to the strength of inhibition in the neonatal rodent spinal cord in vitro. J Neurosci 2015; 35:5307-16. [PMID: 25834055 PMCID: PMC6705413 DOI: 10.1523/jneurosci.1674-14.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 02/16/2015] [Accepted: 02/26/2015] [Indexed: 11/21/2022] Open
Abstract
In healthy mature motoneurons (MNs), KCC2 cotransporters maintain the intracellular chloride concentration at low levels, a prerequisite for postsynaptic inhibition mediated by GABA and glycine. KCC2 expression in lumbar MNs is reduced after spinal cord injury (SCI) resulting in a depolarizing shift of the chloride equilibrium potential. Despite modeling studies indicating that such a downregulation of KCC2 function would reduce the strength of postsynaptic inhibition, physiological evidence is still lacking. The present study aimed at investigating the functional impact of a modification of KCC2 function. We focused on a well characterized disynaptic inhibitory pathway responsible for reciprocal inhibition between antagonistic muscles. We performed in vitro extracellular recordings on spinal cords isolated from rodents at the end of the first postnatal week. Genetic reduction of KCC2 expression, pharmacological blockade of KCC2, as well as SCI-induced downregulation of KCC2 all resulted in a reduction of the strength of reciprocal inhibition. We then tried to restore endogenous inhibition after SCI by means of zinc ions that have been shown to boost KCC2 function in other models. Zinc chloride indeed hyperpolarized the chloride equilibrium potential in MNs and increased reciprocal inhibition after neonatal SCI. This study demonstrates that the level of KCC2 function sets the strength of postsynaptic inhibition and suggests that the downregulation of KCC2 after SCI likely contributes to the high occurrence of flexor-extensor cocontractions in SCI patients.
Collapse
Affiliation(s)
- Florian Gackière
- Institut de Neurosciences de la Timone, UMR 7289, CNRS, Aix Marseille Université, 13385 Marseille cx 5, France
| | - Laurent Vinay
- Institut de Neurosciences de la Timone, UMR 7289, CNRS, Aix Marseille Université, 13385 Marseille cx 5, France
| |
Collapse
|
36
|
Jiang H, Jiang W, Zou J, Wang B, Yu M, Pan Y, Lin Y, Mao Y, Wang Y. The GluN2B subunit of N-methy-D-asparate receptor regulates the radial migration of cortical neurons in vivo. Brain Res 2015; 1610:20-32. [PMID: 25838242 DOI: 10.1016/j.brainres.2015.03.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 02/01/2015] [Accepted: 03/09/2015] [Indexed: 10/23/2022]
Abstract
The formation of layered structure of the mammalian neocortex requires a fine organized migration of post-mitotic neurons during early development. However, whether the radial migration is regulated by NMDA receptor and specific subunits remains contradictory and unknown. Here, we reported that in the developing rat cortex, migration of presumptive layer II/III neurons to their deserved destination was regulated by NMDA receptors with GluN2B but not GluN2A subunit. Using in utero electroporation of small interference RNA (siRNA) of distinct NMDA receptor subunits, we found that knockdown GluN1 and GluN2B subunits dramatically delayed the neuronal migration to proper layer II/III, while improperly stayed at lower layers or even the germinal regions, without changing the cell fate. In contrast, knockdown of GluN2A subunit did not impair the neuronal migration. Additionally, the ecotopic neurons by GluN2B RNAi developed to well dendritic differentiation, while the ones by GluN1 RNAi still kept morphology of migrating neurons. Therefore, GluN2B subunit of NMDA receptor plays an essential role in regulating proper neuronal migration and cortical lamination.
Collapse
Affiliation(s)
- Huayu Jiang
- Neurology Department, Shanghai Tenth People׳s Hospital, Tongji University, School of Medicine, Shanghai 200072, China
| | - Weiqing Jiang
- Department of Neurology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jing Zou
- Department of Neurology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Baoxiang Wang
- Department of Neurology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Mingrong Yu
- Neurology Department, Qiqihar Hospital, Heilongjiang Agriculture Reclamation Bureau, Qiqihar 161005, China
| | - Yuanmei Pan
- Department of Neurology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yan Lin
- Department of Neurology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yingwei Mao
- Department of Biology, 214 Life Sciences Building Penn State University, University Park, PA 16802, USA
| | - Yonggang Wang
- Neurology Department, Shanghai Tenth People׳s Hospital, Tongji University, School of Medicine, Shanghai 200072, China.
| |
Collapse
|
37
|
Khazipov R, Valeeva G, Khalilov I. Depolarizing GABA and developmental epilepsies. CNS Neurosci Ther 2014; 21:83-91. [PMID: 25438879 DOI: 10.1111/cns.12353] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 10/14/2014] [Accepted: 10/20/2014] [Indexed: 12/22/2022] Open
Abstract
Early in development, GABA, which is the main inhibitory neurotransmitter in adult brain, depolarizes immature neurons and exerts dual--excitatory and shunting/inhibitory--effects in the developing neuronal networks. The present review discusses some general questions, including the properties of excitation at depolarizing GABAergic synapse and shunting inhibition by depolarizing GABA; technical issues in exploration of depolarizing GABA using various techniques and preparations, including the developmental aspects of traumatic injury and what is known (or rather unknown) on the actions of GABA in vivo; complex roles of depolarizing GABA in developmental epilepsies, including a contribution of depolarizing GABA to enhanced excitability in the immature networks, caused by repetitive seizures accumulation of intracellular chloride concentration that increases excitatory GABA power and its synchronizing proconvulsive effects, and correction of chloride homeostasis as a potential strategy to treat neonatal seizures.
Collapse
Affiliation(s)
- Roustem Khazipov
- INMED-INSERM U901, Marseille, France; Aix-Marseille University, Marseille, France; Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | | | | |
Collapse
|
38
|
Kaila K, Price TJ, Payne JA, Puskarjov M, Voipio J. Cation-chloride cotransporters in neuronal development, plasticity and disease. Nat Rev Neurosci 2014; 15:637-54. [PMID: 25234263 DOI: 10.1038/nrn3819] [Citation(s) in RCA: 505] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Electrical activity in neurons requires a seamless functional coupling between plasmalemmal ion channels and ion transporters. Although ion channels have been studied intensively for several decades, research on ion transporters is in its infancy. In recent years, it has become evident that one family of ion transporters, cation-chloride cotransporters (CCCs), and in particular K(+)-Cl(-) cotransporter 2 (KCC2), have seminal roles in shaping GABAergic signalling and neuronal connectivity. Studying the functions of these transporters may lead to major paradigm shifts in our understanding of the mechanisms underlying brain development and plasticity in health and disease.
Collapse
Affiliation(s)
- Kai Kaila
- 1] Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland. [2] Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland
| | - Theodore J Price
- University of Texas at Dallas, School of Behavior and Brain Sciences, Dallas, Texas 75093, USA
| | - John A Payne
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California 95616, USA
| | - Martin Puskarjov
- 1] Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland. [2] Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland
| | - Juha Voipio
- Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
39
|
Rusanescu G, Mao J. Notch3 is necessary for neuronal differentiation and maturation in the adult spinal cord. J Cell Mol Med 2014; 18:2103-16. [PMID: 25164209 PMCID: PMC4244024 DOI: 10.1111/jcmm.12362] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Notch receptors are key regulators of nervous system development and promoters of neural stem cells renewal and proliferation. Defects in the expression of Notch genes result in severe, often lethal developmental abnormalities. Notch3 is generally thought to have a similar proliferative, anti-differentiation and gliogenic role to Notch1. However, in some cases, Notch3 has an opposite, pro-differentiation effect. Here, we show that Notch3 segregates from Notch1 and is transiently expressed in adult rat and mouse spinal cord neuron precursors and immature neurons. This suggests that during the differentiation of adult neural progenitor cells, Notch signalling may follow a modified version of the classical lateral inhibition model, involving the segregation of individual Notch receptors. Notch3 knockout mice, otherwise neurologically normal, are characterized by a reduced number of mature inhibitory interneurons and an increased number of highly excitable immature neurons in spinal cord laminae I–II. As a result, these mice have permanently lower nociceptive thresholds, similar to chronic pain. These results suggest that defective neuronal differentiation, for example as a result of reduced Notch3 expression or activation, may underlie human cases of intractable chronic pain, such as fibromyalgia and neuropathic pain.
Collapse
Affiliation(s)
- Gabriel Rusanescu
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Charlestown, MA, USA
| | | |
Collapse
|
40
|
Fukuchi M, Kirikoshi Y, Mori A, Eda R, Ihara D, Takasaki I, Tabuchi A, Tsuda M. Excitatory GABA induces BDNF transcription via CRTC1 and phosphorylated CREB-related pathways in immature cortical cells. J Neurochem 2014; 131:134-46. [PMID: 24965890 DOI: 10.1111/jnc.12801] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 06/11/2014] [Accepted: 06/22/2014] [Indexed: 11/29/2022]
Abstract
Although the excitatory action of GABA has been shown to activate the expression of brain-derived neurotrophic factor (BDNF), its molecular mechanisms remain unclear. Using cultured rat cortical cells, we here demonstrated that GABA induced Bdnf mRNA expression mainly via L-type voltage-dependent Ca(2+) channels (L-VDCC) at the early stage and inhibited it at the late stage of the culture, which corresponded to the excitatory and inhibitory states of cortical cells. The excitatory GABA-induced Bdnf mRNA expression was controlled by multiple Ca(2+) signaling pathways including Ca(2+) /calmodulin-dependent protein kinase (CaMK), mitogen-activated protein kinase (MAPK) and calcineurin (CN). The Bdnf-promoter IV (Bdnf-pIV) was activated by GABA, mainly via cAMP-response element (CRE)/CREB, and this was prevented by the over-expression of a dominant negative CREB. The nuclear translocation of CREB-regulated transcriptional coactivator 1 (CRTC1) was selectively induced by the GABA-induced CN pathway to activate Bdnf-pIV. On the other hand, GABA-induced Gal4-CREB-dependent transcription, which was controlled by multiple Ca(2+) signaling pathways, was prevented when the serine at position 133 of Gal4-CREB was mutated to alanine. Taken together, the excitatory action of GABA transcriptionally activated Bdnf expression through the combination of nuclear-localized CRTC1 and phosphorylated CREB in immature cortical cells, and may be the molecular mechanisms underlying Bdnf expression to control neuronal development. We demonstrated that GABA induced Bdnf expression at the early stage of the culture, in which GABA exerted its excitatory action. The excitatory GABA-induced Bdnf expression was controlled by multiple Ca(2+) signaling pathways evoked via L-VDCC. Both the CREB coactivator, CRTC1 and CREB phosphorylation participated in excitatory GABA-induced Bdnf transcription. Our present study indicates the mechanism underlying the excitatory GABA-induced Bdnf expression in immature neurons and provide new insights into molecular mechanisms underlying Bdnf expression to control neuronal development.
Collapse
Affiliation(s)
- Mamoru Fukuchi
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yuya Kirikoshi
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Atsumi Mori
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Reika Eda
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Daisuke Ihara
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Ichiro Takasaki
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama, Japan.,Department of Pharmacology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan
| | - Akiko Tabuchi
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Masaaki Tsuda
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
41
|
Single-neuron NMDA receptor phenotype influences neuronal rewiring and reintegration following traumatic injury. J Neurosci 2014; 34:4200-13. [PMID: 24647941 DOI: 10.1523/jneurosci.4172-13.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Alterations in the activity of neural circuits are a common consequence of traumatic brain injury (TBI), but the relationship between single-neuron properties and the aggregate network behavior is not well understood. We recently reported that the GluN2B-containing NMDA receptors (NMDARs) are key in mediating mechanical forces during TBI, and that TBI produces a complex change in the functional connectivity of neuronal networks. Here, we evaluated whether cell-to-cell heterogeneity in the connectivity and aggregate contribution of GluN2B receptors to [Ca(2+)]i before injury influenced the functional rewiring, spontaneous activity, and network plasticity following injury using primary rat cortical dissociated neurons. We found that the functional connectivity of a neuron to its neighbors, combined with the relative influx of calcium through distinct NMDAR subtypes, together contributed to the individual neuronal response to trauma. Specifically, individual neurons whose [Ca(2+)]i oscillations were largely due to GluN2B NMDAR activation lost many of their functional targets 1 h following injury. In comparison, neurons with large GluN2A contribution or neurons with high functional connectivity both independently protected against injury-induced loss in connectivity. Mechanistically, we found that traumatic injury resulted in increased uncorrelated network activity, an effect linked to reduction of the voltage-sensitive Mg(2+) block of GluN2B-containing NMDARs. This uncorrelated activation of GluN2B subtypes after injury significantly limited the potential for network remodeling in response to a plasticity stimulus. Together, our data suggest that two single-cell characteristics, the aggregate contribution of NMDAR subtypes and the number of functional connections, influence network structure following traumatic injury.
Collapse
|
42
|
Liu F, Rainosek SW, Sadovova N, Fogle CM, Patterson TA, Hanig JP, Paule MG, Slikker W, Wang C. Protective effect of acetyl-l-carnitine on propofol-induced toxicity in embryonic neural stem cells. Neurotoxicology 2014; 42:49-57. [DOI: 10.1016/j.neuro.2014.03.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 03/21/2014] [Accepted: 03/21/2014] [Indexed: 10/25/2022]
|
43
|
Sun C, Zhang L, Chen G. An unexpected role of neuroligin-2 in regulating KCC2 and GABA functional switch. Mol Brain 2013; 6:23. [PMID: 23663753 PMCID: PMC3661362 DOI: 10.1186/1756-6606-6-23] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND GABAA receptors are ligand-gated Cl- channels, and the intracellular Cl- concentration governs whether GABA function is excitatory or inhibitory. During early brain development, GABA undergoes functional switch from excitation to inhibition: GABA depolarizes immature neurons but hyperpolarizes mature neurons due to a developmental decrease of intracellular Cl- concentration. This GABA functional switch is mainly mediated by the up-regulation of KCC2, a potassium-chloride cotransporter that pumps Cl- outside neurons. However, the upstream factor that regulates KCC2 expression is unclear. RESULTS We report here that KCC2 is unexpectedly regulated by neuroligin-2 (NL2), a cell adhesion molecule specifically localized at GABAergic synapses. The expression of NL2 precedes that of KCC2 in early postnatal development. Upon knockdown of NL2, the expression level of KCC2 is significantly decreased, and GABA functional switch is significantly delayed during early development. Overexpression of shRNA-proof NL2 rescues both KCC2 reduction and delayed GABA functional switch induced by NL2 shRNAs. Moreover, NL2 appears to be required to maintain GABA inhibitory function even in mature neurons, because knockdown NL2 reverses GABA action to excitatory. Gramicidin-perforated patch clamp recordings confirm that NL2 directly regulates the GABA equilibrium potential. We further demonstrate that knockdown of NL2 decreases dendritic spines through down-regulating KCC2. CONCLUSIONS Our data suggest that in addition to its conventional role as a cell adhesion molecule to regulate GABAergic synaptogenesis, NL2 also regulates KCC2 to modulate GABA functional switch and even glutamatergic synapses. Therefore, NL2 may serve as a master regulator in balancing excitation and inhibition in the brain.
Collapse
Affiliation(s)
- Chicheng Sun
- Department of Biology, The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Lei Zhang
- Department of Biology, The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gong Chen
- Department of Biology, The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
44
|
Nondopaminergic Neurotransmission in the Pathophysiology of Tourette Syndrome. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2013; 112:95-130. [DOI: 10.1016/b978-0-12-411546-0.00004-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
45
|
Li Y, Xu Y, van den Pol AN. Reversed synaptic effects of hypocretin and NPY mediated by excitatory GABA-dependent synaptic activity in developing MCH neurons. J Neurophysiol 2012; 109:1571-8. [PMID: 23255725 DOI: 10.1152/jn.00522.2012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In mature neurons, GABA is the primary inhibitory neurotransmitter. In contrast, in developing neurons, GABA exerts excitatory actions, and in some neurons GABA-mediated excitatory synaptic activity is more prevalent than glutamate-mediated excitation. Hypothalamic neuropeptides that modulate cognitive arousal and energy homeostasis, hypocretin/orexin and neuropeptide Y (NPY), evoked reversed effects on synaptic actions that were dependent on presynaptic GABA release onto melanin-concentrating hormone (MCH) neurons. MCH neurons were identified by selective green fluorescent protein (GFP) expression in transgenic mice. In adults, hypocretin increased GABA release leading to reduced excitation. In contrast, in the developing brain as studied here with analysis of miniature excitatory postsynaptic currents, paired-pulse ratios, and evoked potentials, hypocretin acted presynaptically to enhance the excitatory actions of GABA. The ability of hypocretin to enhance GABA release increases inhibition in adult neurons but paradoxically enhances excitation in developing MCH neurons. In contrast, NPY attenuation of GABA release reduced inhibition in mature neurons but enhanced inhibition during development by attenuating GABA excitation. Both hypocretin and NPY also evoked direct actions on developing MCH neurons. Hypocretin excited MCH cells by activating a sodium-calcium exchanger and by reducing potassium currents; NPY reduced activity by increasing an inwardly rectifying potassium current. These data for the first time show that both hypocretin and NPY receptors are functional presynaptically during early postnatal hypothalamic development and that both neuropeptides modulate GABA actions during development with a valence of enhanced excitation or inhibition opposite to that of the adult state, potentially allowing neuropeptide modulation of use-dependent synapse stabilization.
Collapse
Affiliation(s)
- Ying Li
- Dept. of Neurosurgery, Yale Univ. School of Medicine, 333 Cedar St., New Haven, CT 06520, USA
| | | | | |
Collapse
|
46
|
Kunz PA, Burette AC, Weinberg RJ, Philpot BD. Glycine receptors support excitatory neurotransmitter release in developing mouse visual cortex. J Physiol 2012; 590:5749-64. [PMID: 22988142 DOI: 10.1113/jphysiol.2012.241299] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Glycine receptors (GlyRs) are found in most areas of the brain, and their dysfunction can cause severe neurological disorders. While traditionally thought of as inhibitory receptors, presynaptic-acting GlyRs (preGlyRs) can also facilitate glutamate release under certain circumstances, although the underlying molecular mechanisms are unknown. In the current study, we sought to better understand the role of GlyRs in the facilitation of excitatory neurotransmitter release in mouse visual cortex. Using whole-cell recordings, we found that preGlyRs facilitate glutamate release in developing, but not adult, visual cortex. The glycinergic enhancement of neurotransmitter release in early development depends on the high intracellular to extracellular Cl(-) gradient maintained by the Na(+)-K(+)-2Cl(-) cotransporter and requires Ca(2+) entry through voltage-gated Ca(2+) channels. The glycine transporter 1, localized to glial cells, regulates extracellular glycine concentration and the activation of these preGlyRs. Our findings demonstrate a developmentally regulated mechanism for controlling excitatory neurotransmitter release in the neocortex.
Collapse
Affiliation(s)
- Portia A Kunz
- Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Campus Box 7545, 115 Mason Farm Rd, Chapel Hill, NC 27599-7545, USA
| | | | | | | |
Collapse
|
47
|
Decreased Immunoreactivities of the Chloride Transporters, KCC2 and NKCC1, in the Lateral Superior Olive Neurons of Kanamycin-treated Rats. Clin Exp Otorhinolaryngol 2012; 5:117-21. [PMID: 22977707 PMCID: PMC3437411 DOI: 10.3342/ceo.2012.5.3.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Revised: 06/17/2012] [Accepted: 08/17/2012] [Indexed: 11/21/2022] Open
Abstract
Objectives From our previous study about the weak expressions of potassium-chloride (KCC2) and sodium-potassium-2 chloride (NKCC1) co-transporters in the lateral superior olive (LSO) in circling mice, we hypothesized that partially damaged cochlea of circling mice might be a cause of the weak expressions of KCC2 or NKCC1. To test this possibility, we reproduced the altered expressions of KCC2 and NKCC1 in the LSO of rats, whose cochleae were partially destroyed with kanamycin. Methods Rat pups were treated with kanamycin from postnatal (P)3 to P8 (700 mg/kg, subcutaneous injection, twice a day) and sacrificed for immunohistochemical analysis, scanning electron microscope (SEM) and auditory brain stem response. Results The SEM study revealed partially missing hair cells in P9 rats treated with kanamycin, and the hearing threshold was elevated to 63.8±2.5 dB SPL (4 ears) at P16. Both KCC2 and NKCC1 immunoreactivities were more prominent in control rats on P16. On 9 paired slices, the mean densities of NKCC1 immunoreactivities were 118.0±1.0 (control) and 112.2±1.2 (kanamycin treated), whereas those of KCC2 were 115.7±1.5 (control) and 112.0±0.8 (kanamycin treated). Conclusion We concluded that weak expressions of KCC2 and NKCC1 in circling mice were due to partial destruction of cochleae.
Collapse
|
48
|
Ben-Ari Y, Woodin MA, Sernagor E, Cancedda L, Vinay L, Rivera C, Legendre P, Luhmann HJ, Bordey A, Wenner P, Fukuda A, van den Pol AN, Gaiarsa JL, Cherubini E. Refuting the challenges of the developmental shift of polarity of GABA actions: GABA more exciting than ever! Front Cell Neurosci 2012; 6:35. [PMID: 22973192 PMCID: PMC3428604 DOI: 10.3389/fncel.2012.00035] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 07/29/2012] [Indexed: 12/12/2022] Open
Abstract
During brain development, there is a progressive reduction of intracellular chloride associated with a shift in GABA polarity: GABA depolarizes and occasionally excites immature neurons, subsequently hyperpolarizing them at later stages of development. This sequence, which has been observed in a wide range of animal species, brain structures and preparations, is thought to play an important role in activity-dependent formation and modulation of functional circuits. This sequence has also been considerably reinforced recently with new data pointing to an evolutionary preserved rule. In a recent “Hypothesis and Theory Article,” the excitatory action of GABA in early brain development is suggested to be “an experimental artefact” (Bregestovski and Bernard, 2012). The authors suggest that the excitatory action of GABA is due to an inadequate/insufficient energy supply in glucose-perfused slices and/or to the damage produced by the slicing procedure. However, these observations have been repeatedly contradicted by many groups and are inconsistent with a large body of evidence including the fact that the developmental shift is neither restricted to slices nor to rodents. We summarize the overwhelming evidence in support of both excitatory GABA during development, and the implications this has in developmental neurobiology.
Collapse
Affiliation(s)
- Yehezkel Ben-Ari
- INSERM Unité 901, Université de la Méditerranée, UMR S901 Aix-Marseille 2 and INMED Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Direct and glia-mediated effects of GABA on development of central olfactory neurons. ACTA ACUST UNITED AC 2012; 7:143-61. [PMID: 22874585 DOI: 10.1017/s1740925x12000075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Previously studied for its role in processing olfactory information in the antennal lobe, GABA also may shape development of the olfactory pathway, acting either through or on glial cells. Early in development, the dendrites of GABAergic neurons extend to the glial border that surrounds the nascent olfactory lobe neuropil. These neuropil glia express both GABAA and GABAB receptors, about half of the glia in acute cultures responded to GABA with small outward currents, and about a third responded with small transient increases in intracellular calcium. The neuronal classes that express GABA in vivo, the local interneurons and a subset of projection neurons, also do so in culture. Exposure to GABA in culture increased the size and complexity of local interneurons, but had no effect on glial morphology. The presence of glia alone did not affect neuronal morphology, but in the presence of both glia and GABA, the growth-enhancing effects of GABA on cultured antennal lobe neurons were eliminated. Contact between the glial cells and the neurons was not necessary. Operating in vivo, these antagonistic effects, one direct and one glia mediated, could help to sculpt the densely branched, tufted arbors that are characteristic of neurons innervating olfactory glomeruli.
Collapse
|
50
|
Boulland JL, Chaudhry FA. Ontogenetic changes in the distribution of the vesicular GABA transporter VGAT correlate with the excitation/inhibition shift of GABA action. Neurochem Int 2012; 61:506-16. [PMID: 22490609 DOI: 10.1016/j.neuint.2012.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/19/2012] [Accepted: 03/20/2012] [Indexed: 10/28/2022]
Abstract
GABA is the major inhibitory neurotransmitter in the adult CNS and is among others involved in the synchronization of large neuronal networks. During development, GABA acts as a morphogenetic factor and has transient excitatory actions in many brain regions. One distinct protein, the vesicular GABA transporter (VGAT), has been identified accumulating GABA into presynaptic vesicles prior to its exocytotic release. The function of VGAT and its distribution is well defined in the adult, but its contribution to the transient excitatory action at putative GABAergic nerve terminals in the immature brain and its potential roles in putative glutamatergic nerve terminals remain elusive. We have studied VGAT expression in the brain from late embryonic stages through several postnatal stages until adulthood. Quantitative immunoblotting and immunolabeling of tissue sections at the light microscope and the electron microscope levels show an abrupt augmentation in VGAT staining in the cerebral cortex during the first three postnatal weeks, resembling the increase in other proteins involved in GABA synthesis and recycling in the same time frame - such as GAD65, GAD67, GAT1 (Slc6a1) and SN1 (Slc38a3) - and coincides with the synaptogenetic spurt. Dynamic changes in the expression of VGAT are seen in many cellular populations and in several layers in different brain regions. However, mossy fiber terminals (MFT) elude staining for VGAT. We also demonstrate that VGAT(+) nerve terminals undergo a developmental reorganization so that from targeting primarily the dendrites of the principal neurons in several brain regions in the immature brain, they target the soma of the same cells in the adult. This shift in the targeted subcellular compartment coincides with the conversion of the chloride gradient across neuronal membranes and suggests that it may be important for the shift of GABA action from excitation to inhibition and for the establishment of the potent synchronization of neuronal networks.
Collapse
|