1
|
Yang J, Kong L, Zou L, Liu Y. The role of synaptic protein NSF in the development and progression of neurological diseases. Front Neurosci 2024; 18:1395294. [PMID: 39498393 PMCID: PMC11532144 DOI: 10.3389/fnins.2024.1395294] [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: 03/03/2024] [Accepted: 10/04/2024] [Indexed: 11/07/2024] Open
Abstract
This document provides a comprehensive examination of the pivotal function of the N-ethylmaleimide-sensitive factor (NSF) protein in synaptic function. The NSF protein directly participates in critical biological processes, including the cyclic movement of synaptic vesicles (SVs) between exocytosis and endocytosis, the release and transmission of neurotransmitters, and the development of synaptic plasticity through interactions with various proteins, such as SNARE proteins and neurotransmitter receptors. This review also described the multiple functions of NSF in intracellular membrane fusion events and its close associations with several neurological disorders, such as Parkinson's disease, Alzheimer's disease, and epilepsy. Subsequent studies should concentrate on determining high-resolution structures of NSF in different domains, identifying its specific alterations in various diseases, and screening small molecule regulators of NSF from multiple perspectives. These research endeavors aim to reveal new therapeutic targets associated with the biological functions of NSF and disease mechanisms.
Collapse
Affiliation(s)
- Jingyue Yang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingyue Kong
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Li Zou
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yumin Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
2
|
Lorenz-Guertin JM, Povysheva N, Chapman CA, MacDonald ML, Fazzari M, Nigam A, Nuwer JL, Das S, Brady ML, Vajn K, Bambino MJ, Weintraub ST, Johnson JW, Jacob TC. Inhibitory and excitatory synaptic neuroadaptations in the diazepam tolerant brain. Neurobiol Dis 2023; 185:106248. [PMID: 37536384 PMCID: PMC10578451 DOI: 10.1016/j.nbd.2023.106248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023] Open
Abstract
Benzodiazepine (BZ) drugs treat seizures, anxiety, insomnia, and alcohol withdrawal by potentiating γ2 subunit containing GABA type A receptors (GABAARs). BZ clinical use is hampered by tolerance and withdrawal symptoms including heightened seizure susceptibility, panic, and sleep disturbances. Here, we investigated inhibitory GABAergic and excitatory glutamatergic plasticity in mice tolerant to benzodiazepine sedation. Repeated diazepam (DZP) treatment diminished sedative effects and decreased DZP potentiation of GABAAR synaptic currents without impacting overall synaptic inhibition. While DZP did not alter γ2-GABAAR subunit composition, there was a redistribution of extrasynaptic GABAARs to synapses, resulting in higher levels of synaptic BZ-insensitive α4-containing GABAARs and a concomitant reduction in tonic inhibition. Conversely, excitatory glutamatergic synaptic transmission was increased, and NMDAR subunits were upregulated at synaptic and total protein levels. Quantitative proteomics further revealed cortex neuroadaptations of key pro-excitatory mediators and synaptic plasticity pathways highlighted by Ca2+/calmodulin-dependent protein kinase II (CAMKII), MAPK, and PKC signaling. Thus, reduced inhibitory GABAergic tone and elevated glutamatergic neurotransmission contribute to disrupted excitation/inhibition balance and reduced BZ therapeutic power with benzodiazepine tolerance.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nadya Povysheva
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Caitlyn A Chapman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew L MacDonald
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marco Fazzari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Aparna Nigam
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jessica L Nuwer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sabyasachi Das
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Megan L Brady
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katarina Vajn
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew J Bambino
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antoni, TX, USA
| | - Jon W Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Chapman CA, Nuwer JL, Jacob TC. The Yin and Yang of GABAergic and Glutamatergic Synaptic Plasticity: Opposites in Balance by Crosstalking Mechanisms. Front Synaptic Neurosci 2022; 14:911020. [PMID: 35663370 PMCID: PMC9160301 DOI: 10.3389/fnsyn.2022.911020] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 04/26/2022] [Indexed: 01/12/2023] Open
Abstract
Synaptic plasticity is a critical process that regulates neuronal activity by allowing neurons to adjust their synaptic strength in response to changes in activity. Despite the high proximity of excitatory glutamatergic and inhibitory GABAergic postsynaptic zones and their functional integration within dendritic regions, concurrent plasticity has historically been underassessed. Growing evidence for pathological disruptions in the excitation and inhibition (E/I) balance in neurological and neurodevelopmental disorders indicates the need for an improved, more "holistic" understanding of synaptic interplay. There continues to be a long-standing focus on the persistent strengthening of excitation (excitatory long-term potentiation; eLTP) and its role in learning and memory, although the importance of inhibitory long-term potentiation (iLTP) and depression (iLTD) has become increasingly apparent. Emerging evidence further points to a dynamic dialogue between excitatory and inhibitory synapses, but much remains to be understood regarding the mechanisms and extent of this exchange. In this mini-review, we explore the role calcium signaling and synaptic crosstalk play in regulating postsynaptic plasticity and neuronal excitability. We examine current knowledge on GABAergic and glutamatergic synapse responses to perturbances in activity, with a focus on postsynaptic plasticity induced by short-term pharmacological treatments which act to either enhance or reduce neuronal excitability via ionotropic receptor regulation in neuronal culture. To delve deeper into potential mechanisms of synaptic crosstalk, we discuss the influence of synaptic activity on key regulatory proteins, including kinases, phosphatases, and synaptic structural/scaffolding proteins. Finally, we briefly suggest avenues for future research to better understand the crosstalk between glutamatergic and GABAergic synapses.
Collapse
Affiliation(s)
| | | | - Tija C. Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
4
|
Qin J, Wei T, Chen H, Lin X, Qin D, Wei F, Liu P, Ye W, Su J. Salicylate Induced GABAAR Internalization by Dopamine D1-Like Receptors Involving Protein Kinase C (PKC) in Spiral Ganglion Neurons. Med Sci Monit 2021; 27:e933278. [PMID: 34657931 PMCID: PMC8532520 DOI: 10.12659/msm.933278] [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] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Sodium salicylate (SS) induces excitotoxicity of spiral ganglion neurons (SGNs) by inhibiting the response of γ-aminobutyric acid type A receptors (GABAARs). Our previous studies have shown that SS can increase the internalization of GABAARs on SGNs, which involves dopamine D1-like receptors (D1Rs) and related signaling pathways. In this study, we aimed to explore the role of D1Rs and their downstream molecule protein kinase C (PKC) in the process of SS inhibiting GABAARs. MATERIAL AND METHODS The expression of D1Rs and GABARγ2 on rat cochlear SGNs cultured in vitro was tested by immunofluorescence. Then, the SGNs were exposed to SS, D1R agonist (SKF38393), D1R antagonist (SCH23390), clathrin/dynamin-mediated endocytosis inhibitor (dynasore), and PKC inhibitor (Bisindolylmaleimide I). Western blotting and whole-cell patch clamp technique were used to assess the changes of surface and total protein of GABARγ2 and GABA-activated currents. RESULTS Immunofluorescence showed that D1 receptors (DRD1) were expressed on SGNs. Data from western blotting showed that SS promoted the internalization of cell surface GABAARs, and activating D1Rs had the same result. Inhibiting D1Rs and PKC decreased the internalization of GABAARs. Meanwhile, the phosphorylation level of GABAARγ2 S327 affected by PKC was positively correlated with the degree of internalization of GABAARs. Moreover, whole-cell patch clamp recording showed that inhibition of D1Rs or co-inhibition of D1Rs and PKC attenuated the inhibitory effect of SS on GABA-activated currents. CONCLUSIONS D1Rs mediate the GABAAR internalization induced by SS via a PKC-dependent manner and participate in the excitotoxic process of SGNs.
Collapse
Affiliation(s)
- Jiangyuan Qin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Tingjia Wei
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Huiying Chen
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Xiaoyu Lin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Danxue Qin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Fangyu Wei
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Peiqiang Liu
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Wenhua Ye
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jiping Su
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
5
|
Di Cerbo A, Roncati L, Marini C, Carnevale G, Zavatti M, Avallone R, Corsi L. Possible Association Between DHEA and PKCε in Hepatic Encephalopathy Amelioration: A Pilot Study. Front Vet Sci 2021; 8:695375. [PMID: 34651032 PMCID: PMC8505975 DOI: 10.3389/fvets.2021.695375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/25/2021] [Indexed: 12/18/2022] Open
Abstract
Objective: Hepatic encephalopathy (HE) is a neuropsychiatric syndrome caused by liver failure and by an impaired neurotransmission and neurological function caused by hyperammonemia (HA). HE, in turn, decreases the phosphorylation of protein kinase C epsilon (PKCε), contributing to the impairment of neuronal functions. Dehydroepiandrosterone (DHEA) exerts a neuroprotective effect by increasing the GABAergic tone through GABAA receptor stimulation. Therefore, we investigated the protective effect of DHEA in an animal model of HE, and the possible modulation of PKCε expression in different brain area. Methods: Fulminant hepatic failure was induced in 18 male, Sprague–Dawley rats by i.p. administration of 3 g/kg D-galactosamine, and after 30 min, a group of animals received a subcutaneous injection of 25 mg/kg (DHEA) repeated twice a day (3 days). Exploratory behavior and general activity were evaluated 24 h and 48 h after the treatments by the open field test. Then, brain cortex and cerebellum were used for immunoblotting analysis of PKCε level. Results: DHEA administration showed a significant improvement of locomotor activity both 24 and 48 h after D-galactosamine treatment (****p < 0.0001) but did not ameliorate liver parenchymal degeneration. Western blot analysis revealed a reduced immunoreactivity of PKCε (*p < 0.05) following D-galactosamine treatment in rat cortex and cerebellum. After the addition of DHEA, PKCε increased in the cortex in comparison with the D-galactosamine-treated (***p < 0.001) and control group (*p < 0.05), but decreased in the cerebellum (*p < 0.05) with respect to the control group. PKCε decreased after treatment with NH4Cl alone and in combination with DHEA in both cerebellum and cortex (****p < 0.0001). MTS assay demonstrated the synergistic neurotoxic action of NH4Cl and glutamate pretreatment in cerebellum and cortex along with an increased cell survival after DHEA pretreatment, which was significant only in the cerebellum (*p < 0.05). Conclusion: An association between the DHEA-mediated increase of PKCε expression and the improvement of comatose symptoms was observed. PKCε activation and expression in the brain could inhibit GABA-ergic tone counteracting HE symptoms. In addition, DHEA seemed to ameliorate the symptoms of HE and to increase the expression of PKCε in cortex and cerebellum.
Collapse
Affiliation(s)
- Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | - Luca Roncati
- Institute of Pathology, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlotta Marini
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | - Gianluca Carnevale
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Manuela Zavatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Rossella Avallone
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Lorenzo Corsi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.,National Institute of Biostructure and Biosystems, Rome, Italy
| |
Collapse
|
6
|
Weng YC, Huang YT, Chiang IC, Tsai PJ, Su YW, Chou WH. Lipocalin-2 mediates the rejection of neural transplants. FASEB J 2021; 35:e21317. [PMID: 33421207 DOI: 10.1096/fj.202001018r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 11/19/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Lipocalin-2 (LCN2) has been implicated in promoting apoptosis and neuroinflammation in neurological disorders; however, its role in neural transplantation remains unknown. In this study, we cultured and differentiated Lund human mesencephalic (LUHMES) cells into human dopaminergic-like neurons and found that LCN2 mRNA was progressively induced in mouse brain after the intrastriatal transplantation of human dopaminergic-like neurons. The induction of LCN2 protein was detected in a subset of astrocytes and neutrophils infiltrating the core of the engrafted sites, but not in neurons and microglia. LCN2-immunoreactive astrocytes within the engrafted sites expressed lower levels of A1 and A2 astrocytic markers. Recruitment of microglia, neutrophils, and monocytes after transplantation was attenuated in LCN2 deficiency mice. The expression of M2 microglial markers was significantly elevated and survival of engrafted neurons was markedly improved after transplantation in LCN2 deficiency mice. Brain type organic cation transporter (BOCT), the cell surface receptor for LCN2, was induced in dopaminergic-like neurons after differentiation, and treatment with recombinant LCN2 protein directly induced apoptosis in dopaminergic-like neurons in a dose-dependent manner. Our results, therefore, suggested that LCN2 is a neurotoxic factor for the engrafted neurons and a modulator of neuroinflammation. LCN2 inhibition may be useful in reducing rejection after neural transplantation.
Collapse
Affiliation(s)
- Yi-Chinn Weng
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yu-Ting Huang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - I-Chen Chiang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Pei-Ju Tsai
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Yu-Wen Su
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Wen-Hai Chou
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
7
|
Minutillo A, Panza G, Mauri MC. Musical practice and BDNF plasma levels as a potential marker of synaptic plasticity: an instrument of rehabilitative processes. Neurol Sci 2020; 42:1861-1867. [PMID: 32940801 PMCID: PMC8043880 DOI: 10.1007/s10072-020-04715-9] [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: 05/07/2020] [Accepted: 09/08/2020] [Indexed: 01/06/2023]
Abstract
Background and objectives The aim of the study was to investigate the influence of musical practice on brain plasticity. BDNF (brain-derived neurotrophic factor) is a neurotrophin involved in neuroplasticity and synaptic function. Materials and methods We recruited 48 healthy subjects of equal age and sex (21 musicians and 27 non-musicians). All subjects were administered the AQ (Autism-Spectrum Questionnaire) and plasma levels (PLs) of BDNF, oxytocin (OT), and vasopressin (VP) were measured in the blood sample of every participant. Results. The difference between BDNF PLs in the two groups was found to be statistically significant (t = − 2.214, p = 0.03). Furthermore, oxytocin (OT) PLs and musical practice were found to be independent positive predictors of BDNF PLs (p < 0.04). We also found a negative correlation between BDNF PLs and AD (attention to detail) sub-scale score of AQ throughout the whole sample. Assuming BDNF PLs to be a marker of synaptic plasticity, higher PLs could be associated with the activation of alternative neural pathways: a lower score in the “attention to detail” sub-scale could imply greater flexibility of higher cerebral functions among musicians. Further researches should be conducted to assess the rehabilitative usefulness of these findings among patients affected by psychiatric disorders.
Collapse
Affiliation(s)
- Alessandro Minutillo
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy. .,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| | - Gabriele Panza
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Carlo Mauri
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
8
|
Neutralization of Lipocalin-2 Diminishes Stroke-Reperfusion Injury. Int J Mol Sci 2020; 21:ijms21176253. [PMID: 32872405 PMCID: PMC7503651 DOI: 10.3390/ijms21176253] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 01/18/2023] Open
Abstract
Oxidative stress is a key contributor to the pathogenesis of stroke-reperfusion injury. Neuroinflammatory peptides released after ischemic stroke mediate reperfusion injury. Previous studies, including ours, have shown that lipocalin-2 (LCN2) is secreted in response to cerebral ischemia to promote reperfusion injury. Genetic deletion of LCN2 significantly reduces brain injury after stroke, suggesting that LCN2 is a mediator of reperfusion injury and a potential therapeutic target. Immunotherapy has the potential to harness neuroinflammatory responses and provides neuroprotection against stroke. Here we report that LCN2 was induced on the inner surface of cerebral endothelial cells, neutrophils, and astrocytes that gatekeep the blood–brain barrier (BBB) after stroke. LCN2 monoclonal antibody (mAb) specifically targeted LCN2 in vitro and in vivo, attenuating the induction of LCN2 and pro-inflammatory mediators (iNOS, IL-6, CCL2, and CCL9) after stroke. Administration of LCN2 mAb at 4 h after stroke significantly reduced neurological deficits, cerebral infarction, edema, BBB leakage, and infiltration of neutrophils. The binding epitope of LCN2 mAb was mapped to the β3 and β4 strands, which are responsible for maintaining the integrity of LCN2 cup-shaped structure. These data indicate that LCN2 can be pharmacologically targeted using a specific mAb to reduce reperfusion injury after stroke.
Collapse
|
9
|
Schwann Cell Autocrine and Paracrine Regulatory Mechanisms, Mediated by Allopregnanolone and BDNF, Modulate PKCε in Peripheral Sensory Neurons. Cells 2020; 9:cells9081874. [PMID: 32796542 PMCID: PMC7465687 DOI: 10.3390/cells9081874] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023] Open
Abstract
Protein kinase type C-ε (PKCε) plays important roles in the sensitization of primary afferent nociceptors, such as ion channel phosphorylation, that in turn promotes mechanical hyperalgesia and pain chronification. In these neurons, PKCε is modulated through the local release of mediators by the surrounding Schwann cells (SCs). The progesterone metabolite allopregnanolone (ALLO) is endogenously synthesized by SCs, whereas it has proven to be a crucial mediator of neuron-glia interaction in peripheral nerve fibers. Biomolecular and pharmacological studies on rat primary SCs and dorsal root ganglia (DRG) neuronal cultures were aimed at investigating the hypothesis that ALLO modulates neuronal PKCε, playing a role in peripheral nociception. We found that SCs tonically release ALLO, which, in turn, autocrinally upregulated the synthesis of the growth factor brain-derived neurotrophic factor (BDNF). Subsequently, glial BDNF paracrinally activates PKCε via trkB in DRG sensory neurons. Herein, we report a novel mechanism of SCs-neuron cross-talk in the peripheral nervous system, highlighting a key role of ALLO and BDNF in nociceptor sensitization. These findings emphasize promising targets for inhibiting the development and chronification of neuropathic pain.
Collapse
|
10
|
Cilleros-Mañé V, Just-Borràs L, Tomàs M, Garcia N, Tomàs JM, Lanuza MA. The M 2 muscarinic receptor, in association to M 1 , regulates the neuromuscular PKA molecular dynamics. FASEB J 2020; 34:4934-4955. [PMID: 32052889 DOI: 10.1096/fj.201902113r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/23/2019] [Accepted: 01/20/2020] [Indexed: 01/13/2023]
Abstract
Muscarinic acetylcholine receptor 1 subtype (M1 ) and muscarinic acetylcholine receptor 2 subtype (M2 ) presynaptic muscarinic receptor subtypes increase and decrease, respectively, neurotransmitter release at neuromuscular junctions. M2 involves protein kinase A (PKA), although the muscarinic regulation to form and inactivate the PKA holoenzyme is unknown. Here, we show that M2 signaling inhibits PKA by downregulating Cβ subunit, upregulating RIIα/β and liberating RIβ and RIIα to the cytosol. This promotes PKA holoenzyme formation and reduces the phosphorylation of the transmitter release target synaptosome-associated protein 25 and the gene regulator cAMP response element binding. Instead, M1 signaling, which is downregulated by M2 , opposes to M2 by recruiting R subunits to the membrane. The M1 and M2 reciprocal actions are performed through the anchoring protein A kinase anchor protein 150 as a common node. Interestingly, M2 modulation on protein expression needs M1 signaling. Altogether, these results describe the dynamics of PKA subunits upon M2 muscarinic signaling in basal and under presynaptic nerve activity, uncover a specific involvement of the M1 receptor and reveal the M1 /M2 balance to activate PKA to regulate neurotransmission. This provides a molecular mechanism to the PKA holoenzyme formation and inactivation which could be general to other synapses and cellular models.
Collapse
Affiliation(s)
- Víctor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - Josep Maria Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - Maria Angel Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
11
|
Wang JJ, Liu F, Yang F, Wang YZ, Qi X, Li Y, Hu Q, Zhu MX, Xu TL. Disruption of auto-inhibition underlies conformational signaling of ASIC1a to induce neuronal necroptosis. Nat Commun 2020; 11:475. [PMID: 31980622 PMCID: PMC6981194 DOI: 10.1038/s41467-019-13873-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
We reported previously that acid-sensing ion channel 1a (ASIC1a) mediates acidic neuronal necroptosis via recruiting receptor-interacting protein kinase 1 (RIPK1) to its C terminus (CT), independent of its ion-conducting function. Here we show that the N-terminus (NT) of ASIC1a interacts with its CT to form an auto-inhibition that prevents RIPK1 recruitment/activation under resting conditions. The interaction involves glutamate residues at distal NT and is disrupted by acidosis. Expression of mutant ASIC1a bearing truncation or glutamate-to-alanine substitutions at distal NT causes constitutive cell death. The NT-CT interaction is further disrupted by N-ethylmaleimide-sensitive fusion ATPase (NSF), which associates with ASIC1a-NT under acidosis, facilitating RIPK1 interaction with ASIC1a-CT. Importantly, a membrane-penetrating synthetic peptide representing the distal 20 ASIC1a NT residues, NT1–20, reduced neuronal damage in both in vitro model of acidotoxicity and in vivo mouse model of ischemic stroke, demonstrating the therapeutic potential of targeting the auto-inhibition of ASIC1a for neuroprotection against acidotoxicity. Acid-sensing ion channel 1a (ASIC1a) mediates acidic neuronal necroptosis via recruiting receptor-interacting protein kinase 1 (RIPK1). Here authors show that auto-inhibition of ASICa prevents RIPK1 recruitment and demonstrate that targeting the auto-inhibition has therapeutic potential to prevent acidotoxicity.
Collapse
Affiliation(s)
- Jing-Jing Wang
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Liu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Yang
- Department of Biophysics and Kidney Disease Center, First Affiliated Hospital, Institute of Neuroscience, National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi-Zhi Wang
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Qi
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Li
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Hu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, USA.
| | - Tian-Le Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Kumar V, Weng YC, Wu YC, Huang YT, Chou WH. PKCε phosphorylation regulates the mitochondrial translocation of ATF2 in ischemia-induced neurodegeneration. BMC Neurosci 2018; 19:76. [PMID: 30497386 PMCID: PMC6267029 DOI: 10.1186/s12868-018-0479-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/27/2018] [Indexed: 11/10/2022] Open
Abstract
Background Global cerebral ischemia triggers neurodegeneration in the hippocampal CA1 region, but the mechanism of neuronal death remains elusive. The epsilon isoform of protein kinase C (PKCε) has recently been identified as a master switch that controls the nucleocytoplasmic trafficking of ATF2 and the survival of melanoma cells. It is of interest to assess the role of PKCε–ATF2 signaling in neurodegeneration. Results Phosphorylation of ATF2 at Thr-52 was reduced in the hippocampus of PKCε null mice, suggesting that ATF2 is a phosphorylation substrate of PKCε. PKCε protein concentrations were significantly reduced 4, 24, 48 and 72 h after transient global cerebral ischemia, resulting in translocation of nuclear ATF2 to the mitochondria. Degenerating neurons staining positively with Fluoro-Jade C exhibited cytoplasmic ATF2. Conclusions Our results support the hypothesis that PKCε regulates phosphorylation and nuclear sequestration of ATF2 in hippocampal neurons during ischemia-induced neurodegeneration.
Collapse
Affiliation(s)
- Varun Kumar
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH, 44242, USA
| | - Yi-Chinn Weng
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, 35053, Taiwan, ROC
| | - Yu-Chieh Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, 35053, Taiwan, ROC
| | - Yu-Ting Huang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, 35053, Taiwan, ROC
| | - Wen-Hai Chou
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH, 44242, USA. .,Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, 35053, Taiwan, ROC.
| |
Collapse
|
13
|
Kumar V, Weng YC, Wu YC, Huang YT, Liu TH, Kristian T, Liu YL, Tsou HH, Chou WH. Genetic inhibition of PKCε attenuates neurodegeneration after global cerebral ischemia in male mice. J Neurosci Res 2018; 97:444-455. [PMID: 30488977 DOI: 10.1002/jnr.24362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/31/2018] [Accepted: 11/13/2018] [Indexed: 11/08/2022]
Abstract
Global cerebral ischemia that accompanies cardiac arrest is a major cause of morbidity and mortality. Protein Kinase C epsilon (PKCε) is a member of the novel PKC subfamily and plays a vital role in ischemic preconditioning. Pharmacological activation of PKCε before cerebral ischemia confers neuroprotection. The role of endogenous PKCε after cerebral ischemia remains elusive. Here we used male PKCε-null mice to assess the effects of PKCε deficiency on neurodegeneration after transient global cerebral ischemia (tGCI). We found that the cerebral vasculature, blood flow, and the expression of other PKC isozymes were not altered in the PKCε-null mice. Spatial learning and memory was impaired after tGCI, but the impairment was attenuated in male PKCε-null mice as compared to male wild-type controls. A significant reduction in Fluoro-Jade C labeling and mitochondrial release of cytochrome C in the hippocampus was found in male PKCε-null mice after tGCI. Male PKCε-null mice expressed increased levels of PKCδ in the mitochondria, which may prevent the translocation of PKCδ from the cytosol to the mitochondria after tGCI. Our results demonstrate the neuroprotective effects of PKCε deficiency on neurodegeneration after tGCI, and suggest that reduced mitochondrial translocation of PKCδ may contribute to the neuroprotective action in male PKCε-null mice.
Collapse
Affiliation(s)
- Varun Kumar
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Yi-Chinn Weng
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Chieh Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Ting Huang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Tung-Hsia Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Tibor Kristian
- Department of Anesthesiology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Hsiao-Hui Tsou
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan.,Graduate Institute of Biostatistics, College of Public Health, China Medical University, Taichung, Taiwan
| | - Wen-Hai Chou
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio.,Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
14
|
Lorenz-Guertin JM, Bambino MJ, Jacob TC. γ2 GABA AR Trafficking and the Consequences of Human Genetic Variation. Front Cell Neurosci 2018; 12:265. [PMID: 30190672 PMCID: PMC6116786 DOI: 10.3389/fncel.2018.00265] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/02/2018] [Indexed: 11/13/2022] Open
Abstract
GABA type A receptors (GABAARs) mediate the majority of fast inhibitory neurotransmission in the central nervous system (CNS). Most prevalent as heteropentamers composed of two α, two β, and a γ2 subunit, these ligand-gated ionotropic chloride channels are capable of extensive genetic diversity (α1-6, β1-3, γ1-3, δ, 𝜀, 𝜃, π, ρ1-3). Part of this selective GABAAR assembly arises from the critical role for γ2 in maintaining synaptic receptor localization and function. Accordingly, mutations in this subunit account for over half of the known epilepsy-associated genetic anomalies identified in GABAARs. Fundamental structure-function studies and cellular pathology investigations have revealed dynamic GABAAR trafficking and synaptic scaffolding as critical regulators of GABAergic inhibition. Here, we introduce in vitro and in vivo findings regarding the specific role of the γ2 subunit in receptor trafficking. We then examine γ2 subunit human genetic variation and assess disease related phenotypes and the potential role of altered GABAAR trafficking. Finally, we discuss new-age imaging techniques and their potential to provide novel insight into critical regulatory mechanisms of GABAAR function.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Matthew J Bambino
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
15
|
Simó A, Just-Borràs L, Cilleros-Mañé V, Hurtado E, Nadal L, Tomàs M, Garcia N, Lanuza MA, Tomàs J. BDNF-TrkB Signaling Coupled to nPKCε and cPKCβI Modulate the Phosphorylation of the Exocytotic Protein Munc18-1 During Synaptic Activity at the Neuromuscular Junction. Front Mol Neurosci 2018; 11:207. [PMID: 29946239 PMCID: PMC6007318 DOI: 10.3389/fnmol.2018.00207] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/25/2018] [Indexed: 11/13/2022] Open
Abstract
Munc18-1, a neuron-specific member of the Sec1/Munc18 family, is involved in neurotransmitter release by binding tightly to syntaxin. Munc18-1 is phosphorylated by PKC on Ser-306 and Ser-313 in vitro which reduces the amount of Munc18-1 able to bind syntaxin. We have previously identified that PKC is involved in neurotransmitter release when continuous electrical stimulation imposes a moderate activity on the NMJ and that muscle contraction through TrkB has an important impact on presynaptic PKC isoforms levels, specifically cPKCβI and nPKCε. Therefore, the present study was designed to understand how Munc18-1 phosphorylation is affected by (1) synaptic activity at the neuromuscular junction, (2) nPKCε and cPKCβI isoforms activity, (3) muscle contraction per se, and (4) the BDNF/TrkB signaling in a neuromuscular activity-dependent manner. We performed immunohistochemistry and confocal techniques to evidence the presynaptic location of Munc18-1 in the rat diaphragm muscle. To study synaptic activity, we stimulated the phrenic nerve (1 Hz, 30 min) with or without contraction (abolished by μ-conotoxin GIIIB). Specific inhibitory reagents were used to block nPKCε and cPKCβI activity and to modulate the tropomyosin receptor kinase B (TrkB). Main results obtained from Western blot experiments showed that phosphorylation of Munc18-1 at Ser-313 increases in response to a signaling mechanism initiated by synaptic activity and directly mediated by nPKCε. Otherwise, cPKCβI and TrkB activities work together to prevent this synaptic activity-induced Munc18-1 phosphorylation by a negative regulation of cPKCβI over nPKCε. Therefore, a balance between the activities of these PKC isoforms could be a relevant cue in the regulation of the exocytotic apparatus. The results also demonstrate that muscle contraction prevents the synaptic activity-induced Munc18-1 phosphorylation through a mechanism that opposes the TrkB/cPKCβI/nPKCε signaling.
Collapse
Affiliation(s)
- Anna Simó
- Unitat d'Histologia i Neurobiologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Víctor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Laura Nadal
- Unitat d'Histologia i Neurobiologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
16
|
Pakri Mohamed RM, Mokhtar MH, Yap E, Hanim A, Abdul Wahab N, Jaffar FHF, Kumar J. Ethanol-Induced Changes in PKCε: From Cell to Behavior. Front Neurosci 2018; 12:244. [PMID: 29706864 PMCID: PMC5907685 DOI: 10.3389/fnins.2018.00244] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/28/2018] [Indexed: 11/20/2022] Open
Abstract
The long-term binge intake of ethanol causes neuroadaptive changes that lead to drinkers requiring higher amounts of ethanol to experience its effects. This neuroadaptation can be partly attributed to the modulation of numerous neurotransmitter receptors by the various protein kinases C (PKCs). PKCs are enzymes that control cellular activities by regulating other proteins via phosphorylation. Among the various isoforms of PKC, PKCε is the most implicated in ethanol-induced biochemical and behavioral changes. Ethanol exposure causes changes to PKCε expression and localization in various brain regions that mediate addiction-favoring plasticity. Ethanol works in conjunction with numerous upstream kinases and second messenger activators to affect cellular PKCε expression. Chauffeur proteins, such as receptors for activated C kinase (RACKs), cause the translocation of PKCε to aberrant sites and mediate ethanol-induced changes. In this article, we aim to review the following: the general structure and function of PKCε, ethanol-induced changes in PKCε expression, the regulation of ethanol-induced PKCε activities in DAG-dependent and DAG-independent environments, the mechanisms underlying PKCε-RACKε translocation in the presence of ethanol, and the existing literature on the role of PKCε in ethanol-induced neurobehavioral changes, with the goal of creating a working model upon which further research can build.
Collapse
Affiliation(s)
| | - Mohd H. Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ernie Yap
- Department of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Athirah Hanim
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Norhazlina Abdul Wahab
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Farah H. F. Jaffar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
17
|
Lorenz-Guertin JM, Jacob TC. GABA type a receptor trafficking and the architecture of synaptic inhibition. Dev Neurobiol 2018; 78:238-270. [PMID: 28901728 PMCID: PMC6589839 DOI: 10.1002/dneu.22536] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/08/2017] [Accepted: 09/08/2017] [Indexed: 12/21/2022]
Abstract
Ubiquitous expression of GABA type A receptors (GABAA R) in the central nervous system establishes their central role in coordinating most aspects of neural function and development. Dysregulation of GABAergic neurotransmission manifests in a number of human health disorders and conditions that in certain cases can be alleviated by drugs targeting these receptors. Precise changes in the quantity or activity of GABAA Rs localized at the cell surface and at GABAergic postsynaptic sites directly impact the strength of inhibition. The molecular mechanisms constituting receptor trafficking to and from these compartments therefore dictate the efficacy of GABAA R function. Here we review the current understanding of how GABAA Rs traffic through biogenesis, plasma membrane transport, and degradation. Emphasis is placed on discussing novel GABAergic synaptic proteins, receptor and scaffolding post-translational modifications, activity-dependent changes in GABAA R confinement, and neuropeptide and neurosteroid mediated changes. We further highlight modern techniques currently advancing the knowledge of GABAA R trafficking and clinically relevant neurodevelopmental diseases connected to GABAergic dysfunction. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 238-270, 2018.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261
| |
Collapse
|
18
|
Liu M, Fitzgibbon M, Wang Y, Reilly J, Qian X, O'Brien T, Clapcote S, Shen S, Roche M. Ulk4 regulates GABAergic signaling and anxiety-related behavior. Transl Psychiatry 2018; 8:43. [PMID: 29391390 PMCID: PMC5804027 DOI: 10.1038/s41398-017-0091-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/09/2017] [Accepted: 11/30/2017] [Indexed: 01/15/2023] Open
Abstract
Excitation/inhibition imbalance has been proposed as a fundamental mechanism in the pathogenesis of neuropsychiatric and neurodevelopmental disorders, in which copy number variations of the Unc-51 like kinase 4 (ULK4) gene encoding a putative Serine/Threonine kinase have been reported in approximately 1/1000 of patients suffering pleiotropic clinical conditions of schizophrenia, depression, autistic spectrum disorder (ASD), developmental delay, language delay, intellectual disability, or behavioral disorder. The current study characterized behavior of heterozygous Ulk4 +/tm1a mice, demonstrating that Ulk4 +/tm1a mice displayed no schizophrenia-like behavior in acoustic startle reactivity and prepulse inhibition tests or depressive-like behavior in the Porsolt swim or tail suspension tests. However, Ulk4 +/tm1a mice exhibited an anxiety-like behavioral phenotype in several tests. Previously identified hypo-anxious (Atp1a2, Ptn, and Mdk) and hyper-anxious (Gria1, Syngap1, and Npy2r) genes were found to be dysregulated accordingly in Ulk4 mutants. Ulk4 was found to be expressed in GABAergic neurons and the Gad67+ interneurons were significantly reduced in the hippocampus and basolateral amygdala of Ulk4 +/tm1a mice. Transcriptome analyses revealed a marked reduction of GABAergic neuronal subtypes, including Pvalb, Sst, Cck, Npy, and Nos3, as well as significant upregulation of GABA receptors, including Gabra1, Gabra3, Gabra4, Gabra5, and Gabrb3. This is the first evidence that Ulk4 plays a major role in regulating GABAergic signaling and anxiety-like behavior, which may have implications for the development of novel anxiolytic treatments.
Collapse
Affiliation(s)
- Min Liu
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Marie Fitzgibbon
- Physiology, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, National University of Ireland Galway, Galway, Ireland
| | - Yanqin Wang
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Physiology, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Jamie Reilly
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Xiaohong Qian
- National Center for Protein Sciences, Beijing Proteome Research Center, National Engineering Research Center for Protein Drugs, Beijing Institute of Radiation Medicine, Beijing, China
| | - Timothy O'Brien
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Steve Clapcote
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, National University of Ireland Galway, Galway, Ireland.
| | - Michelle Roche
- Physiology, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
19
|
Fang H, Wang ZH, Bu YJ, Yuan ZJ, Wang GQ, Guo Y, Cheng XY, Qiu WJ. Repeated inhalation of sevoflurane inhibits the information transmission of Purkinje cells and delays motor development via the GABAA receptor ε subunit in neonatal mice. Mol Med Rep 2017; 17:1083-1092. [PMID: 29115488 PMCID: PMC5780070 DOI: 10.3892/mmr.2017.7941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 09/19/2017] [Indexed: 11/21/2022] Open
Abstract
General anesthesia is widely used in pediatric surgery, although the influence of general anesthesia on cerebellar information transmission and motor function is unclear. In the present study, neonatal mice received repeated inhalation of sevoflurane, and electrophysiological alterations in Purkinje cells (PCs) and the development of motor functions were detected. In addition, γ-aminobutyric acidA receptor ε (GABAA-R ε) subunit knockout mice were used to investigate the mechanism of action of sevoflurane on cerebellar function. In the neonatal mice, the field potential response of PCs induced by sensory stimulation and the motor function indices were markedly inhibited by sevoflurane, and the inhibitory effect was positively associated with the number of repetitions of anesthesia. In additional the GABAA-R ε subunit level of PCs was promoted by sevoflurane in a dose-dependent manner, and the inhibitory effects of sevoflurane on PC field potential response and motor function were alleviated in GABAA-R ε subunit knockout mice. The GABAA-R ε subunit was activated by sevoflurane, leading to inhibition of sensory information transmission in the cerebellar cortex, field potential responses of PCs and the development of cerebellar motor function. The present study provided experimental evidence for the safe usage of sevoflurane in clinical anesthesia, and suggested that GABAA-R ε subunit antagonists may be considered for combined application with general anesthesia with repeated inhalation of sevoflurane, for adverse effect prevention in the clinic.
Collapse
Affiliation(s)
- Hong Fang
- Department of Anesthesiology, The Affiliated Heji Hospital of Changzhi Medical College, Changzhi, Shanxi 046011, P.R. China
| | - Ze-Hua Wang
- Department of Anesthesiology, The Affiliated Heji Hospital of Changzhi Medical College, Changzhi, Shanxi 046011, P.R. China
| | - Ying-Jiang Bu
- Department of Anesthesiology, The Affiliated Heji Hospital of Changzhi Medical College, Changzhi, Shanxi 046011, P.R. China
| | - Zhi-Jun Yuan
- Department of Anesthesiology, The Affiliated Heji Hospital of Changzhi Medical College, Changzhi, Shanxi 046011, P.R. China
| | - Guo-Qiang Wang
- Department of Anesthesiology, The Affiliated Heji Hospital of Changzhi Medical College, Changzhi, Shanxi 046011, P.R. China
| | - Yan Guo
- Department of Anesthesiology, The Affiliated Heji Hospital of Changzhi Medical College, Changzhi, Shanxi 046011, P.R. China
| | - Xiao-Yun Cheng
- Department of Anesthesiology, The Suburban People's Hospital, Changzhi, Shanxi 046011, P.R. China
| | - Wen-Jie Qiu
- Department of Anesthesiology, The Suburban People's Hospital, Changzhi, Shanxi 046011, P.R. China
| |
Collapse
|
20
|
Trexler AJ, Taraska JW. Regulation of insulin exocytosis by calcium-dependent protein kinase C in beta cells. Cell Calcium 2017; 67:1-10. [PMID: 29029784 DOI: 10.1016/j.ceca.2017.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 12/27/2022]
Abstract
The control of insulin release from pancreatic beta cells helps ensure proper blood glucose level, which is critical for human health. Protein kinase C has been shown to be one key control mechanism for this process. After glucose stimulation, calcium influx into beta cells triggers exocytosis of insulin-containing dense-core granules and activates protein kinase C via calcium-dependent phospholipase C-mediated generation of diacylglycerol. Activated protein kinase C potentiates insulin release by enhancing the calcium sensitivity of exocytosis, likely by affecting two main pathways that could be linked: (1) the reorganization of the cortical actin network, and (2) the direct phosphorylation of critical exocytotic proteins such as munc18, SNAP25, and synaptotagmin. Here, we review what is currently known about the molecular mechanisms of protein kinase C action on each of these pathways and how these effects relate to the control of insulin release by exocytosis. We identify remaining challenges in the field and suggest how these challenges might be addressed to advance our understanding of the regulation of insulin release in health and disease.
Collapse
Affiliation(s)
- Adam J Trexler
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Justin W Taraska
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
21
|
The Neuropeptide Orexin-A Inhibits the GABAA Receptor by PKC and Ca2+/CaMKII-Dependent Phosphorylation of Its β1 Subunit. J Mol Neurosci 2017; 61:459-467. [DOI: 10.1007/s12031-017-0886-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022]
|
22
|
A Highly Polymorphic Copy Number Variant in the NSF Gene is Associated with Cocaine Dependence. Sci Rep 2016; 6:31033. [PMID: 27498889 PMCID: PMC4976312 DOI: 10.1038/srep31033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 07/12/2016] [Indexed: 12/25/2022] Open
Abstract
Cocaine dependence is a complex psychiatric disorder involving both genetic and environmental factors. Several neurotransmitter systems mediate cocaine’s effects, dependence and relapse, being the components of the neurotransmitter release machinery good candidates for the disorder. Previously, we identified a risk haplotype for cocaine dependence in the NSF gene, encoding the protein N-Ethylmaleimide-Sensitive Factor essential for synaptic vesicle turnover. Here we examined the possible contribution to cocaine dependence of a large copy number variant (CNV) that encompasses part of the NSF gene. We performed a case-control association study in a discovery sample (359 cases and 356 controls) and identified an association between cocaine dependence and the CNV (P = 0.013), that was confirmed in the replication sample (508 cases and 569 controls, P = 7.1e-03) and in a pooled analysis (P = 1.8e-04), with an over-representation of low number of copies in cases. Subsequently, we studied the functional impact of the CNV on gene expression and found that the levels of two NSF transcripts were significantly increased in peripheral blood mononuclear cells (PBMC) along with the number of copies of the CNV. These results, together with a previous study from our group, support the role of NSF in the susceptibility to cocaine dependence.
Collapse
|
23
|
Nishizaki T. N-Ethylmaleimide Dissociates α7 ACh Receptor from a Complex with NSF and Promotes Its Delivery to the Presynaptic Membrane. Neurochem Res 2016; 41:2043-8. [PMID: 27105867 DOI: 10.1007/s11064-016-1915-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 11/29/2022]
Abstract
N-Ethylmaleimide (NEM)-sensitive factor (NSF) associates with soluble NSF attachment protein (SNAP), that binds to SNAP receptors (SNAREs) including syntaxin, SNAP25, and synaptobrevin. The complex of NSF/SNAP/SNAREs plays a critical role in the regulation of vesicular traffic. The present study investigated NEM-regulated α7 ACh receptor translocation. NSF associated with β-SNAP and the SNAREs syntaxin 1 and synaptobrevin 2 in the rat hippocampus. NSF also associated with the α7 ACh receptor subunit, the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunits GluA1 and GluA2, and the γ-aminobutyric acid A (GABAA) receptor γ2 subunit. NEM, an inhibitor of NSF, significantly dissociated the α7 ACh receptor subunit from a complex with NSF and increased cell surface localization of the receptor subunit, but such effect was not obtained with the GluA1, GluA2 or γ2 subunits. NEM, alternatively, dissociated synaptobrevin 2 from an assembly of NSF/β-SNAP/syntaxin 1/synaptobrevin 2. NEM significantly increased the rate of nicotine-triggered AMPA receptor-mediated miniature excitatory postsynaptic currents, without affecting the amplitude, in rat hippocampal slices. The results of the present study indicate that NEM releases the α7 ACh receptor subunit and synaptobrevin 2 from an assembly of α7 ACh receptor subunit/NSF/β-SNAP/syntaxin 1/synaptobrevin 2, thereby promoting delivery of the α7 ACh receptor subunit to presynaptic membrane.
Collapse
|
24
|
Maiya R, McMahon T, Wang D, Kanter B, Gandhi D, Chapman HL, Miller J, Messing RO. Selective chemical genetic inhibition of protein kinase C epsilon reduces ethanol consumption in mice. Neuropharmacology 2016; 107:40-48. [PMID: 26947945 DOI: 10.1016/j.neuropharm.2016.02.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/18/2016] [Accepted: 02/27/2016] [Indexed: 11/19/2022]
Abstract
Reducing expression or inhibiting translocation of protein kinase C epsilon (PKCε) prolongs ethanol intoxication and decreases ethanol consumption in mice. However, we do not know if this phenotype is due to reduced PKCε kinase activity or to impairment of kinase-independent functions. In this study, we used a chemical-genetic strategy to determine whether a potent and highly selective inhibitor of PKCε catalytic activity reduces ethanol consumption. We generated ATP analog-specific PKCε (AS-PKCε) knock-in mice harboring a point mutation in the ATP binding site of PKCε that renders the mutant kinase highly sensitive to inhibition by 1-tert-butyl-3-naphthalen-1-ylpyrazolo[3,4-d]pyrimidin-4-amine (1-NA-PP1). Systemically administered 1-NA-PP1 readily crossed the blood brain barrier and inhibited PKCε-mediated phosphorylation. 1-NA-PP1 reversibly reduced ethanol consumption by AS-PKCε mice but not by wild type mice lacking the AS-PKCε mutation. These results support the development of inhibitors of PKCε catalytic activity as a strategy to reduce ethanol consumption, and they demonstrate that the AS- PKCε mouse is a useful tool to study the role of PKCε in behavior.
Collapse
Affiliation(s)
- Rajani Maiya
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 78712, USA
| | - Thomas McMahon
- The Ernest Gallo Clinic and Research Center, University of California San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA
| | - Dan Wang
- The Ernest Gallo Clinic and Research Center, University of California San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA
| | - Benjamin Kanter
- The Ernest Gallo Clinic and Research Center, University of California San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA
| | - Dev Gandhi
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 78712, USA
| | - Holly L Chapman
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 78712, USA
| | - Jacklyn Miller
- The Ernest Gallo Clinic and Research Center, University of California San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA
| | - Robert O Messing
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 78712, USA; The Ernest Gallo Clinic and Research Center, University of California San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA.
| |
Collapse
|
25
|
Andersson H, Björnström K, Eintrei C, Sundqvist T. Orexin a phosphorylates the γ-Aminobutyric acid type A receptor β2 subunit on a serine residue and changes the surface expression of the receptor in SH-SY5Y cells exposed to propofol. J Neurosci Res 2015; 93:1748-55. [PMID: 26283475 DOI: 10.1002/jnr.23631] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/03/2015] [Accepted: 08/03/2015] [Indexed: 12/17/2022]
Abstract
Propofol activates the γ-aminobutyric acid type A receptor (GABAA R) and causes a reversible neurite retraction, leaving a thin, thread-like structure behind; it also reverses the transport of vesicles in rat cortical neurons. The awakening peptide orexin A (OA) inhibits this retraction via phospholipase D (PLD) and protein kinase Cɛ (PKCɛ). The human SH-SY5Y cells express both GABAA Rs and orexin 1 and 2 receptors. These cells are used to examine the interaction between OA and the GABAA R. The effects of OA are studied with flow cytometry and immunoblotting. This study shows that OA stimulates phosphorylation on the serine residues of the GABAA R β2 subunit and that the phosphorylation is caused by the activation of PLD and PKCɛ. OA administration followed by propofol reduces the cell surface expression of the GABAA R, whereas propofol stimulation before OA increases the surface expression. The GABAA R β2 subunit is important for receptor recirculation, and the effect of OA on propofol-stimulated cells may be due to a disturbed recirculation of the GABAA R.
Collapse
Affiliation(s)
- Henrik Andersson
- Department of Anaesthesiology and Intensive Care, Linköping University, Linköping, Sweden.,Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Karin Björnström
- Department of Anaesthesiology and Intensive Care, Linköping University, Linköping, Sweden.,Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Christina Eintrei
- Department of Anaesthesiology and Intensive Care, Linköping University, Linköping, Sweden.,Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Tommy Sundqvist
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
26
|
Weng YC, Wang G, Messing RO, Chou WH. Identification of lipocalin-2 as a PKCδ phosphorylation substrate in neutrophils. J Biomed Sci 2015; 22:21. [PMID: 25890235 PMCID: PMC4396066 DOI: 10.1186/s12929-015-0129-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/13/2015] [Indexed: 01/05/2023] Open
Abstract
Background PKCδ expressed in neutrophils is implicated in promoting reperfusion injury after ischemic stroke. To understand the molecular and cellular actions of PKCδ, we employed a chemical-genetics approach to identify PKCδ substrates in neutrophils. Results We recently generated knock-in mice endogenously expressing analog-specific PKCδ (AS-PKCδ) that can utilize ATP analogs as phosphate donors. Using neutrophils isolated from the knock-in mice, we identified several PKCδ substrates, one of which was lipocalin-2 (LCN2), which is an iron-binding protein that can trigger apoptosis by reducing intracellular iron concentrations. We found that PKCδ phosphorylated LCN2 at T115 and this phosphorylation was reduced in Prkcd−/− mice. PKCδ colocalized with LCN2 in resting and stimulated neutrophils. LCN2 release from neutrophils after cerebral ischemia was reduced in PKCδ null mice. Conclusions These findings suggest that PKCδ phosphorylates LCN2 and mediates its release from neutrophils during ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Yi-Chinn Weng
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH, 44224, USA.
| | - Guona Wang
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH, 44224, USA.
| | - Robert O Messing
- Department of Neurology, University of California, San Francisco, CA, 94608, USA. .,Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Wen-Hai Chou
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH, 44224, USA. .,Department of Neurology, University of California, San Francisco, CA, 94608, USA.
| |
Collapse
|
27
|
Puia G, Ravazzini F, Castelnovo LF, Magnaghi V. PKCε and allopregnanolone: functional cross-talk at the GABAA receptor level. Front Cell Neurosci 2015; 9:83. [PMID: 25852476 PMCID: PMC4365694 DOI: 10.3389/fncel.2015.00083] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/23/2015] [Indexed: 11/13/2022] Open
Abstract
Changes in GABAergic inhibition occur during physiological processes, during response to drugs and in various pathologies. These changes can be achieved through direct allosteric modifications at the γ-amino butyric acid (GABA) type A (GABAA) receptor protein level, or by altering the synthesis, trafficking and stability of the receptor. Neurosteroids (NSs) and protein kinase C (PKC) are potent modulators of GABAA receptors and their effects are presumably intermingled, even though evidence for this hypothesis is only partially explored. However, several PKC isoforms are able to phosphorylate the GABAA receptor, producing different functional effects. We focused on the ε isoform, that has been correlated to the sensitivity of the GABAA receptor to allosteric modulators and whose expression may be regulated in peripheral sensory neurons by NSs. The cross-talk between PKC-ε and NSs, leading to changes in GABAA receptor functionality, is considered and discussed in this perspective.
Collapse
Affiliation(s)
- Giulia Puia
- Department of Life Science, University of Modena and Reggio Emilia Modena, Italy
| | - Federica Ravazzini
- Department of Life Science, University of Modena and Reggio Emilia Modena, Italy
| | - Luca Franco Castelnovo
- Department of Pharmacological and Biomolecular Sciences, University of Milan Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences, University of Milan Milan, Italy
| |
Collapse
|
28
|
Wang G, Weng YC, Han X, Whaley JD, McCrae KR, Chou WH. Lipocalin-2 released in response to cerebral ischaemia mediates reperfusion injury in mice. J Cell Mol Med 2015; 19:1637-45. [PMID: 25702801 PMCID: PMC4511361 DOI: 10.1111/jcmm.12538] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 12/17/2014] [Indexed: 12/04/2022] Open
Abstract
Thrombolysis remains the only effective therapy to reverse acute ischaemic stroke. However, delayed treatment may cause serious complications including hemorrhagic transformation and reperfusion injury. The level of lipocalin-2 (LCN2) is elevated in the plasma of ischaemic stroke patients, but its role in stroke is unknown. Here, we show that LCN2 was acutely induced in mice after ischaemic stroke and is an important mediator of reperfusion injury. Increased levels of LCN2 were observed in mouse serum as early as 1 hr after transient middle cerebral artery occlusion (tMCAO), reaching peak levels at 23 hrs. LCN2 was also detected in neutrophils infiltrating into the ipsilateral hemisphere, as well as a subset of astrocytes after tMCAO, but not in neurons and microglia. Stroke injury, neurological deficits and infiltration of immune cells were markedly diminished in LCN2 null mice after tMCAO, but not after permanent MCAO (pMCAO). In vitro, recombinant LCN2 protein induced apoptosis in primary cultured neurons in a dose-dependent manner. Our results demonstrate that LCN2 is a neurotoxic factor secreted rapidly in response to cerebral ischaemia, suggesting its potential usage as an early stroke biomarker and a novel therapeutic target to reduce stroke-reperfusion injury.
Collapse
Affiliation(s)
- Guona Wang
- Department of Biological Sciences and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Yi-Chinn Weng
- Department of Biological Sciences and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Xiqian Han
- Department of Biological Sciences and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - James D Whaley
- Department of Biological Sciences and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Keith R McCrae
- Department of Cellular and Molecular Medicine and Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Wen-Hai Chou
- Department of Biological Sciences and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
29
|
Wan Y, Wang Q, Prud’homme GJ. GABAergic system in the endocrine pancreas: a new target for diabetes treatment. Diabetes Metab Syndr Obes 2015; 8:79-87. [PMID: 25678807 PMCID: PMC4322886 DOI: 10.2147/dmso.s50642] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Excessive loss of functional pancreatic β-cell mass, mainly due to apoptosis, is a major factor in the development of hyperglycemia in both type 1 and type 2 diabetes (T1D and T2D). In T1D, β-cells are destroyed by immunological mechanisms. In T2D, while metabolic factors are known to contribute to β-cell failure and subsequent apoptosis, mounting evidence suggests that islet inflammation also plays an important role in the loss of β-cell mass. Therefore, it is of great importance for clinical intervention to develop new therapies. γ-Aminobutyric acid (GABA), a major neurotransmitter, is also produced by islet β-cells, where it functions as an important intraislet transmitter in regulating islet-cell secretion and function. Importantly, recent studies performed in rodents, including in vivo studies of xenotransplanted human islets, reveal that GABA exerts β-cell regenerative effects. Moreover, it protects β-cells against apoptosis induced by cytokines, drugs, and other stresses, and has anti-inflammatory and immunoregulatory activities. It ameliorates the manifestations of diabetes in preclinical models, suggesting potential applications for the treatment of diabetic patients. This review outlines the actions of GABA relevant to β-cell regeneration, including its signaling mechanisms and potential interactions with other mediators. These studies increase our understanding of the regenerative processes of pancreatic β-cells, and help pave the way for the development of regenerative medicine for diabetes.
Collapse
Affiliation(s)
- Yun Wan
- Department of Endocrinology and Metabolism, Huashan Hospital, Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Medical College, Fudan University, Shanghai, People’s Republic of China
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St Michael’s Hospital, Toronto, ON, Canada
- Departments of Physiology and Medicine, Faculty of Medicine, Toronto, ON, Canada
- Correspondence: Qinghua Wang, Division of Endocrinology and Metabolism, St Michael’s Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada, Tel +1 416 864 6060 ext 77 610, Fax +1 416 864 5140, Email
| | - Gerald J Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
30
|
Nakamura Y, Darnieder LM, Deeb TZ, Moss SJ. Regulation of GABAARs by phosphorylation. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 72:97-146. [PMID: 25600368 PMCID: PMC5337123 DOI: 10.1016/bs.apha.2014.11.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
γ-Aminobutyric acid type A receptors (GABAARs) are the principal mediators of fast synaptic inhibition in the brain as well as the low persistent extrasynaptic inhibition, both of which are fundamental to proper brain function. Thus unsurprisingly, deficits in GABAARs are implicated in a number of neurological disorders and diseases. The complexity of GABAAR regulation is determined not only by the heterogeneity of these receptors but also by its posttranslational modifications, the foremost, and best characterized of which is phosphorylation. This review will explore the details of this dynamic process, our understanding of which has barely scratched the surface. GABAARs are regulated by a number of kinases and phosphatases, and its phosphorylation plays an important role in governing its trafficking, expression, and interaction partners. Here, we summarize the progress in understanding the role phosphorylation plays in the regulation of GABAARs. This includes how phosphorylation can affect the allosteric modulation of GABAARs, as well as signaling pathways that affect GABAAR phosphorylation. Finally, we discuss the dysregulation of GABAAR phosphorylation and its implication in disease processes.
Collapse
|
31
|
Kumar V, Weng YC, Geldenhuys WJ, Wang D, Han X, Messing RO, Chou WH. Generation and characterization of ATP analog-specific protein kinase Cδ. J Biol Chem 2014; 290:1936-51. [PMID: 25505183 DOI: 10.1074/jbc.m114.598698] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To better study the role of PKCδ in normal function and disease, we developed an ATP analog-specific (AS) PKCδ that is sensitive to specific kinase inhibitors and can be used to identify PKCδ substrates. AS PKCδ showed nearly 200 times higher affinity (Km) and 150 times higher efficiency (kcat/Km) than wild type (WT) PKCδ toward N(6)-(benzyl)-ATP. AS PKCδ was uniquely inhibited by 1-(tert-butyl)-3-(1-naphthyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine (1NA-PP1) and 1-(tert-butyl)-3-(2-methylbenzyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine (2MB-PP1) but not by other 4-amino-5-(4-methylphenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP1) analogs tested, whereas WT PKCδ was insensitive to all PP1 analogs. To understand the mechanisms for specificity and affinity of these analogs, we created in silico WT and AS PKCδ homology models based on the crystal structure of PKCι. N(6)-(Benzyl)-ATP and ATP showed similar positioning within the purine binding pocket of AS PKCδ, whereas N(6)-(benzyl)-ATP was displaced from the pocket of WT PKCδ and was unable to interact with the glycine-rich loop that is required for phosphoryl transfer. The adenine rings of 1NA-PP1 and 2MB-PP1 matched the adenine ring of ATP when docked in AS PKCδ, and this interaction prevented the potential interaction of ATP with Lys-378, Glu-428, Leu-430, and Phe-633 residues. 1NA-PP1 failed to effectively dock within WT PKCδ. Other PP1 analogs failed to interact with either AS PKCδ or WT PKCδ. These results provide a structural basis for the ability of AS PKCδ to efficiently and specifically utilize N(6)-(benzyl)-ATP as a phosphate donor and for its selective inhibition by 1NA-PP1 and 2MB-PP1. Such homology modeling could prove useful in designing molecules to target PKCδ and other kinases to understand their function in cell signaling and to identify unique substrates.
Collapse
Affiliation(s)
- Varun Kumar
- From the Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio 44242
| | - Yi-Chinn Weng
- From the Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio 44242
| | - Werner J Geldenhuys
- the Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Dan Wang
- the Ernest Gallo Clinic and Research Center, Department of Neurology, University of California, San Francisco, Emeryville, California 94608, and
| | - Xiqian Han
- From the Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio 44242
| | - Robert O Messing
- the Ernest Gallo Clinic and Research Center, Department of Neurology, University of California, San Francisco, Emeryville, California 94608, and the Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas, Austin, Texas 78712
| | - Wen-Hai Chou
- From the Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, Ohio 44242, the Ernest Gallo Clinic and Research Center, Department of Neurology, University of California, San Francisco, Emeryville, California 94608, and
| |
Collapse
|
32
|
Schuster DJ, Metcalf MD, Kitto KF, Messing RO, Fairbanks CA, Wilcox GL. Ligand requirements for involvement of PKCε in synergistic analgesic interactions between spinal μ and δ opioid receptors. Br J Pharmacol 2014; 172:642-53. [PMID: 24827408 DOI: 10.1111/bph.12774] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE We recently found that PKCε was required for spinal analgesic synergy between two GPCRs, δ opioid receptors and α2 A adrenoceptors, co-located in the same cellular subpopulation. We sought to determine if co-delivery of μ and δ opioid receptor agonists would similarly result in synergy requiring PKCε. EXPERIMENTAL APPROACH Combinations of μ and δ opioid receptor agonists were co-administered intrathecally by direct lumbar puncture to PKCε-wild-type (PKCε-WT) and -knockout (PKCε-KO) mice. Antinociception was assessed using the hot-water tail-flick assay. Drug interactions were evaluated by isobolographic analysis. KEY RESULTS All agonists produced comparable antinociception in both PKCε-WT and PKCε-KO mice. Of 19 agonist combinations that produced analgesic synergy, only 3 required PKCε for a synergistic interaction. In these three combinations, one of the agonists was morphine, although not all combinations involving morphine required PKCε. Morphine + deltorphin II and morphine + deltorphin I required PKCε for synergy, whereas a similar combination, morphine + deltorphin, did not. Additionally, morphine + oxymorphindole required PKCε for synergy, whereas a similar combination, morphine + oxycodindole, did not. CONCLUSIONS AND IMPLICATIONS We discovered biased agonism for a specific signalling pathway at the level of spinally co-delivered opioid agonists. As the bias is only revealed by an appropriate ligand combination and cannot be accounted for by a single drug, it is likely that the receptors these agonists act on are interacting with each other. Our results support the existence of μ and δ opioid receptor heteromers at the spinal level in vivo. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- D J Schuster
- Department of Neuroscience, Medical School, University of Minnesota, Minneapolis, MN, USA
| | | | | | | | | | | |
Collapse
|
33
|
Xu F, Luk CC, Wiersma-Meems R, Baehre K, Herman C, Zaidi W, Wong N, Syed NI. Neuronal somata and extrasomal compartments play distinct roles during synapse formation between Lymnaea neurons. J Neurosci 2014; 34:11304-15. [PMID: 25143611 PMCID: PMC6615512 DOI: 10.1523/jneurosci.1651-14.2014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/07/2014] [Accepted: 07/15/2014] [Indexed: 12/21/2022] Open
Abstract
Proper synapse formation is pivotal for all nervous system functions. However, the precise mechanisms remain elusive. Moreover, compared with the neuromuscular junction, steps regulating the synaptogenic program at central cholinergic synapses remain poorly defined. In this study, we identified different roles of neuronal compartments (somal vs extrasomal) in chemical and electrical synaptogenesis. Specifically, the electrically synapsed Lymnaea pedal dorsal A cluster neurons were used to study electrical synapses, whereas chemical synaptic partners, visceral dorsal 4 (presynaptic, cholinergic), and left pedal dorsal 1 (LPeD1; postsynaptic) were explored for chemical synapse formation. Neurons were cultured in a soma-soma or soma-axon configuration and synapses explored electrophysiologically. We provide the first direct evidence that electrical synapses develop in a soma-soma, but not soma-axon (removal of soma) configuration, indicating the requirement of gene transcription regulation in the somata of both synaptic partners. In addition, the soma-soma electrical coupling was contingent upon trophic factors present in Lymnaea brain-conditioned medium. Further, we demonstrate that chemical (cholinergic) synapses between soma-soma and soma-axon pairs were indistinguishable, with both exhibiting a high degree of contact site and target cell type specificity. We also provide direct evidence that presynaptic cell contact-mediated, clustering of postsynaptic cholinergic receptors at the synaptic site requires transmitter-receptor interaction, receptor internalization, and a protein kinase C-dependent lateral migration toward the contact site. This study provides novel insights into synaptogenesis between central neurons revealing both distinct and synergistic roles of cell-cell signaling and extrinsic trophic factors in executing the synaptogenic program.
Collapse
Affiliation(s)
- Fenglian Xu
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Alberta T2N 4Z6, Canada
| | - Collin C Luk
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Alberta T2N 4Z6, Canada
| | - Ryanne Wiersma-Meems
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Alberta T2N 4Z6, Canada
| | - Kelly Baehre
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Alberta T2N 4Z6, Canada
| | - Cameron Herman
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Alberta T2N 4Z6, Canada
| | - Wali Zaidi
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Alberta T2N 4Z6, Canada
| | - Noelle Wong
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Alberta T2N 4Z6, Canada
| | - Naweed I Syed
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Alberta T2N 4Z6, Canada
| |
Collapse
|
34
|
Chabot-Doré AJ, Schuster DJ, Stone LS, Wilcox GL. Analgesic synergy between opioid and α2 -adrenoceptors. Br J Pharmacol 2014; 172:388-402. [PMID: 24641506 DOI: 10.1111/bph.12695] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 03/08/2014] [Accepted: 03/12/2014] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Opioid and α2 -adrenoceptor agonists are potent analgesic drugs and their analgesic effects can synergize when co-administered. These supra-additive interactions are potentially beneficial clinically; by increasing efficacy and/or reducing the total drug required to produce sufficient pain relief, undesired side effects can be minimized. However, combination therapies of opioids and α2 -adrenoceptor agonists remain underutilized clinically, in spite of a large body of preclinical evidence describing their synergistic interaction. One possible obstacle to the translation of preclinical findings to clinical applications is a lack of understanding of the mechanisms underlying the synergistic interactions between these two drug classes. In this review, we provide a detailed overview of the interactions between different opioid and α2 -adrenoceptor agonist combinations in preclinical studies. These studies have identified the spinal cord as an important site of action of synergistic interactions, provided insights into which receptors mediate these interactions and explored downstream signalling events enabling synergy. It is now well documented that the activation of both μ and δ opioid receptors can produce synergy with α2 -adrenoceptor agonists and that α2 -adrenoceptor agonists can mediate synergy through either the α2A or the α2C adrenoceptor subtypes. Current hypotheses surrounding the cellular mechanisms mediating opioid-adrenoceptor synergy, including PKC signalling and receptor oligomerization, and the evidence supporting them are presented. Finally, the implications of these findings for clinical applications and drug discovery are discussed. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- A-J Chabot-Doré
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | | | | |
Collapse
|
35
|
Wibowo A, Peters EC, Hsieh-Wilson LC. Photoactivatable glycopolymers for the proteome-wide identification of fucose-α(1-2)-galactose binding proteins. J Am Chem Soc 2014; 136:9528-31. [PMID: 24937314 PMCID: PMC4105059 DOI: 10.1021/ja502482a] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
![]()
Although
fucose-α(1-2)-galactose (Fucα(1-2)Gal)-containing
glycans have been implicated in cognitive processes such as learning
and memory, their precise molecular mechanisms are poorly understood.
Here we employed the use of multivalent glycopolymers to enable the
first proteome-wide identification of weak affinity, low abundance
Fucα(1-2)Gal glycan-binding proteins (GBPs). Biotin-terminated
glycopolymers containing photoactivatable cross-linking groups were
designed to capture and enrich GBPs from rat brain lysates. Candidate
proteins were tested for their ability to bind Fucα(1-2)Gal,
and the functional significance of the interaction was investigated
for the synaptic vesicle protein SV2a using a knockout mouse system.
The results suggest a role for SV2a-Fucα(1-2)Gal interactions
in SV2a trafficking and synaptic vesicle recycling. More broadly,
our studies outline a general chemical approach for the systems-level
discovery of novel GBPs.
Collapse
Affiliation(s)
- Arif Wibowo
- Division of Chemistry and Chemical Engineering, California Institute of Technology and Howard Hughes Medical Institute , 1200 East California Boulevard, Pasadena, California 91125, United States
| | | | | |
Collapse
|
36
|
Björnström K, Turina D, Strid T, Sundqvist T, Eintrei C. Orexin A inhibits propofol-induced neurite retraction by a phospholipase D/protein kinase Cε-dependent mechanism in neurons. PLoS One 2014; 9:e97129. [PMID: 24828410 PMCID: PMC4020800 DOI: 10.1371/journal.pone.0097129] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 04/15/2014] [Indexed: 12/16/2022] Open
Abstract
Background The intravenous anaesthetic propofol retracts neurites and reverses the transport of vesicles in rat cortical neurons. Orexin A (OA) is an endogenous neuropeptide regulating wakefulness and may counterbalance anaesthesia. We aim to investigate if OA interacts with anaesthetics by inhibition of the propofol-induced neurite retraction. Methods In primary cortical cell cultures from newborn rats’ brains, live cell light microscopy was used to measure neurite retraction after propofol (2 µM) treatment with or without OA (10 nM) application. The intracellular signalling involved was tested using a protein kinase C (PKC) activator [phorbol 12-myristate 13-acetate (PMA)] and inhibitors of Rho-kinase (HA-1077), phospholipase D (PLD) [5-fluoro-2-indolyl des-chlorohalopemide (FIPI)], PKC (staurosporine), and a PKCε translocation inhibitor peptide. Changes in PKCε Ser729 phosphorylation were detected with Western blot. Results The neurite retraction induced by propofol is blocked by Rho-kinase and PMA. OA blocks neurite retraction induced by propofol, and this inhibitory effect could be prevented by FIPI, staurosporine and PKCε translocation inhibitor peptide. OA increases via PLD and propofol decreases PKCε Ser729 phosphorylation, a crucial step in the activation of PKCε. Conclusions Rho-kinase is essential for propofol-induced neurite retraction in cortical neuronal cells. Activation of PKC inhibits neurite retraction caused by propofol. OA blocks propofol-induced neurite retraction by a PLD/PKCε-mediated pathway, and PKCε maybe the key enzyme where the wakefulness and anaesthesia signal pathways converge.
Collapse
Affiliation(s)
- Karin Björnström
- Department of Medical and Health Sciences, division of Anaesthesiology, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- Clinic of Anaesthesiology, Östergötland County Council UHL, Linköping, Sweden
- * E-mail:
| | - Dean Turina
- Department of Medical and Health Sciences, division of Anaesthesiology, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- Clinic of Anaesthesiology, Östergötland County Council UHL, Linköping, Sweden
| | - Tobias Strid
- Department of Medical and Health Sciences, division of Anaesthesiology, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Tommy Sundqvist
- Department of Clinical and Experimental Medicine, division of Medical Microbiology, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Christina Eintrei
- Department of Medical and Health Sciences, division of Anaesthesiology, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- Clinic of Anaesthesiology, Östergötland County Council UHL, Linköping, Sweden
| |
Collapse
|
37
|
Iwata K, Matsuzaki H, Tachibana T, Ohno K, Yoshimura S, Takamura H, Yamada K, Matsuzaki S, Nakamura K, Tsuchiya KJ, Matsumoto K, Tsujii M, Sugiyama T, Katayama T, Mori N. N-ethylmaleimide-sensitive factor interacts with the serotonin transporter and modulates its trafficking: implications for pathophysiology in autism. Mol Autism 2014; 5:33. [PMID: 24834316 PMCID: PMC4022412 DOI: 10.1186/2040-2392-5-33] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 04/24/2014] [Indexed: 01/23/2023] Open
Abstract
Background Changes in serotonin transporter (SERT) function have been implicated in autism. SERT function is influenced by the number of transporter molecules present at the cell surface, which is regulated by various cellular mechanisms including interactions with other proteins. Thus, we searched for novel SERT-binding proteins and investigated whether the expression of one such protein was affected in subjects with autism. Methods Novel SERT-binding proteins were examined by a pull-down system. Alterations of SERT function and membrane expression upon knockdown of the novel SERT-binding protein were studied in HEK293-hSERT cells. Endogenous interaction of SERT with the protein was evaluated in mouse brains. Alterations in the mRNA expression of SERT (SLC6A4) and the SERT-binding protein in the post-mortem brains and the lymphocytes of autism patients were compared to nonclinical controls. Results N-ethylmaleimide-sensitive factor (NSF) was identified as a novel SERT-binding protein. NSF was co-localized with SERT at the plasma membrane, and NSF knockdown resulted in decreased SERT expression at the cell membranes and decreased SERT uptake function. NSF was endogenously co-localized with SERT and interacted with SERT. While SLC6A4 expression was not significantly changed, NSF expression tended to be reduced in post-mortem brains, and was significantly reduced in lymphocytes of autistic subjects, which correlated with the severity of the clinical symptoms. Conclusions These data clearly show that NSF interacts with SERT under physiological conditions and is required for SERT membrane trafficking and uptake function. A possible role for NSF in the pathophysiology of autism through modulation of SERT trafficking, is suggested.
Collapse
Affiliation(s)
- Keiko Iwata
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan ; Department of Development of Functional Brain Activities, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Fukui, Japan
| | - Hideo Matsuzaki
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan ; Department of Development of Functional Brain Activities, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Fukui, Japan ; Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Taro Tachibana
- Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka, Japan
| | - Koji Ohno
- Department of Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Saori Yoshimura
- Department of Bioengineering, Graduate School of Engineering, Osaka City University, Osaka, Japan
| | - Hironori Takamura
- Department of Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, Japan ; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan
| | - Kohei Yamada
- Department of Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, Japan ; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan
| | - Shinsuke Matsuzaki
- Department of Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, Japan
| | - Kazuhiko Nakamura
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kenji J Tsuchiya
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kaori Matsumoto
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masatsugu Tsujii
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan ; Faculty of Contemporary Sociology, Chukyo University, Toyota, Japan
| | - Toshirou Sugiyama
- Department of Child and Adolescent Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Taiichi Katayama
- Department of Molecular Brain Science, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, Japan
| | - Norio Mori
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan ; Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
38
|
Neurosteroids promote phosphorylation and membrane insertion of extrasynaptic GABAA receptors. Proc Natl Acad Sci U S A 2014; 111:7132-7. [PMID: 24778259 DOI: 10.1073/pnas.1403285111] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neurosteroids are synthesized within the brain and act as endogenous anxiolytic, anticonvulsant, hypnotic, and sedative agents, actions that are principally mediated via their ability to potentiate phasic and tonic inhibitory neurotransmission mediated by γ-aminobutyric acid type A receptors (GABAARs). Although neurosteroids are accepted allosteric modulators of GABAARs, here we reveal they exert sustained effects on GABAergic inhibition by selectively enhancing the trafficking of GABAARs that mediate tonic inhibition. We demonstrate that neurosteroids potentiate the protein kinase C-dependent phosphorylation of S443 within α4 subunits, a component of GABAAR subtypes that mediate tonic inhibition in many brain regions. This process enhances insertion of α4 subunit-containing GABAAR subtypes into the membrane, resulting in a selective and sustained elevation in the efficacy of tonic inhibition. Therefore, the ability of neurosteroids to modulate the phosphorylation and membrane insertion of α4 subunit-containing GABAARs may underlie the profound effects these endogenous signaling molecules have on neuronal excitability and behavior.
Collapse
|
39
|
Brain-derived neurotrophic factor promotes gephyrin protein expression and GABAA receptor clustering in immature cultured hippocampal cells. Neurochem Int 2014; 72:14-21. [PMID: 24747341 DOI: 10.1016/j.neuint.2014.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 12/31/2022]
Abstract
Fast synaptic inhibition in the adult brain is largely mediated by GABAA receptors (GABAAR). GABAAR are anchored to synaptic sites by gephyrin, a scaffolding protein that appears to be assembled as a hexagonal lattice beneath the plasma membrane. Brain derived neurotrophic factor (BDNF) alters the clustering and synaptic distribution of GABAAR but mechanisms behind this regulation are just starting to emerge. The current study was aimed to examine if BDNF alters the protein levels and/or clustering of gephyrin and to investigate whether the modulation of gephyrin is accompanied by changes in the distribution and/or clustering of GABAAR. Exogenous application of BDNF to immature neuronal cultures from rat hippocampus increased the protein levels and clustering of gephyrin. BDNF also augmented the association of gephyrin with GABAAR and promoted the formation of GABAAR clusters. Together, these observations indicate that BDNF might regulate the assembly of GABAergic synapses by promoting the association of GABAAR with gephyrin.
Collapse
|
40
|
Saroja SR, Kim EJ, Shanmugasundaram B, Höger H, Lubec G. Hippocampal monoamine receptor complex levels linked to spatial memory decline in the aging C57BL/6J. Behav Brain Res 2014; 264:1-8. [PMID: 24508236 DOI: 10.1016/j.bbr.2014.01.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 01/25/2014] [Accepted: 01/28/2014] [Indexed: 01/09/2023]
Abstract
Although a large series of reports on monoamine receptor (MAR) biochemistry and pharmacology in aging are available, work on MAR complexes rather than subunits is limited. It was the aim of the study to determine MAR complexes in hippocampi of three different age groups (3-12 and 18 months) in the mouse and to link MAR changes to spatial memory retrieval in the Morris water maze (MWM). MAR complexes were separated by blue native electrophoresis. Immunohistochemistry was performed in order to show the pattern of dopamine receptors and its colocalizations. D1R, D2R and 5-HT7R containing receptor complex levels were decreasing with age while 5-HT1AR-containing complex levels were increasing with age. D1R, 5-HT7R and 5-HT1AR were significantly correlating with the time spent in the target quadrant, representing retrieval in the MWM. D1R and D2R immunoreactivity was decreasing in an area-dependent pattern and D1R and D2R were colocalized. Individual monoamine receptors are linked to spatial memory retrieval and are modulated by age. The findings are relevant for interpretation of previous and design of future work on brain receptors, spatial memory and aging.
Collapse
Affiliation(s)
- Sivaprakasam R Saroja
- Department of Paediatrics, Medical University of Vienna, Währinger Gürtel 18, 1090 Vienna, Austria
| | - Eun-Jung Kim
- Department of Paediatrics, Medical University of Vienna, Währinger Gürtel 18, 1090 Vienna, Austria
| | | | - Harald Höger
- Core Unit of Biomedical Research, Division of Laboratory Animal Science and Genetics, Medical University of Vienna, Brauhausgasse 34, A-2325 Himberg, Austria
| | - Gert Lubec
- Department of Paediatrics, Medical University of Vienna, Währinger Gürtel 18, 1090 Vienna, Austria.
| |
Collapse
|
41
|
Tabakoff B, Hoffman PL. The neurobiology of alcohol consumption and alcoholism: an integrative history. Pharmacol Biochem Behav 2013; 113:20-37. [PMID: 24141171 PMCID: PMC3867277 DOI: 10.1016/j.pbb.2013.10.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 10/09/2013] [Accepted: 10/10/2013] [Indexed: 01/17/2023]
Abstract
Studies of the neurobiological predisposition to consume alcohol (ethanol) and to transition to uncontrolled drinking behavior (alcoholism), as well as studies of the effects of alcohol on brain function, started a logarithmic growth phase after the repeal of the 18th Amendment to the United States Constitution. Although the early studies were primitive by current technological standards, they clearly demonstrated the effects of alcohol on brain structure and function, and by the end of the 20th century left little doubt that alcoholism is a "disease" of the brain. This review traces the history of developments in the understanding of ethanol's effects on the most prominent inhibitory and excitatory systems of brain (GABA and glutamate neurotransmission). This neurobiological information is integrated with knowledge of ethanol's actions on other neurotransmitter systems to produce an anatomical and functional map of ethanol's properties. Our intent is limited in scope, but is meant to provide context and integration of the actions of ethanol on the major neurobiologic systems which produce reinforcement for alcohol consumption and changes in brain chemistry that lead to addiction. The developmental history of neurobehavioral theories of the transition from alcohol drinking to alcohol addiction is presented and juxtaposed to the neurobiological findings. Depending on one's point of view, we may, at this point in history, know more, or less, than we think we know about the neurobiology of alcoholism.
Collapse
Affiliation(s)
- Boris Tabakoff
- University of Colorado School of Medicine, MS8303, 12800 E. 19 Ave., Aurora, CO 80045 U.S.A
| | - Paula L. Hoffman
- University of Colorado School of Medicine, MS8303, 12800 E. 19 Ave., Aurora, CO 80045 U.S.A
| |
Collapse
|
42
|
Abstract
Ethanol's effects on intracellular signaling pathways contribute to acute effects of ethanol as well as to neuroadaptive responses to repeated ethanol exposure. In this chapter we review recent discoveries that demonstrate how ethanol alters signaling pathways involving several receptor tyrosine kinases and intracellular tyrosine and serine-threonine kinases, with consequences for regulation of cell surface receptor function, gene expression, protein translation, neuronal excitability and animal behavior. We also describe recent work that demonstrates a key role for ethanol in regulating the function of scaffolding proteins that organize signaling complexes into functional units. Finally, we review recent exciting studies demonstrating ethanol modulation of DNA and histone modification and the expression of microRNAs, indicating epigenetic mechanisms by which ethanol regulates neuronal gene expression and addictive behaviors.
Collapse
Affiliation(s)
- Dorit Ron
- Ernest Gallo Clinic and Research Center, University of California San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA
| | - Robert O. Messing
- Ernest Gallo Clinic and Research Center, University of California San Francisco, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA
| |
Collapse
|
43
|
Stabilization of GABA(A) receptors at endocytic zones is mediated by an AP2 binding motif within the GABA(A) receptor β3 subunit. J Neurosci 2012; 32:2485-98. [PMID: 22396422 DOI: 10.1523/jneurosci.1622-11.2011] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The strength of synaptic inhibition can be controlled by the stability and endocytosis of surface and synaptic GABA(A) receptors (GABA(A)Rs), but the surface receptor dynamics that underpin GABA(A)R recruitment to dendritic endocytic zones (EZs) have not been investigated. Stabilization of GABA(A)Rs at EZs is likely to be regulated by receptor interactions with the clathrin-adaptor AP2, but the molecular determinants of these associations remain poorly understood. Moreover, although surface GABA(A)R downmodulation plays a key role in pathological disinhibition in conditions such as ischemia and epilepsy, whether this occurs in an AP2-dependent manner also remains unclear. Here we report the characterization of a novel motif containing three arginine residues (405RRR407) within the GABA(A)R β3-subunit intracellular domain (ICD), responsible for the interaction with AP2 and GABA(A)R internalization. When this motif is disrupted, binding to AP2 is abolished in vitro and in rat brain. Using single-particle tracking, we reveal that surface β3-subunit-containing GABA(A)Rs exhibit highly confined behavior at EZs, which is dependent on AP2 interactions via this motif. Reduced stabilization of mutant GABA(A)Rs at EZs correlates with their reduced endocytosis and increased steady-state levels at synapses. By imaging wild-type or mutant super-ecliptic pHluorin-tagged GABA(A)Rs in neurons, we also show that, under conditions of oxygen-glucose deprivation to mimic cerebral ischemia, GABA(A)Rs are depleted from synapses in dendrites, depending on the 405RRR407 motif. Thus, AP2 binding to an RRR motif in the GABA(A)R β3-subunit ICD regulates GABA(A)R residency time at EZs, steady-state synaptic receptor levels, and pathological loss of GABA(A)Rs from synapses during simulated ischemia.
Collapse
|
44
|
Maggio N, Segal M. Stress and corticosteroid modulation of seizures and synaptic inhibition in the hippocampus. Exp Neurol 2011; 234:200-7. [PMID: 22227061 DOI: 10.1016/j.expneurol.2011.12.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 12/07/2011] [Accepted: 12/19/2011] [Indexed: 12/28/2022]
Abstract
The role of stress hormones in the initiation of epileptic seizures has been studied extensively in the past decade, with conflicting observations, from suppression to exacerbation of spontaneous seizures. We have now studied the effects of an acute stress on reactivity of juvenile rats to kainic acid (KA), which produces epileptic seizures. With a short (30s) stress-KA delay, stress exacerbated epilepsy via activation of mineralocorticosterone receptors (MR). With a long (60 min) stress-KA delay, seizures were suppressed through activation of a glucocorticosterone receptor (GR). In a parallel study with CA1 pyramidal neurons in acute hippocampal slices, activation of MRs reduced the frequency of mIPSCs, whereas activation of GRs produced a slow onset, 2.5 fold increase in amplitudes of mIPSCs. GR effects were not mediated by protein synthesis, but did require activation of some protein kinases. These experiments suggest that stress can either facilitate or suppress seizures, in a time and receptor dependent manner.
Collapse
Affiliation(s)
- Nicola Maggio
- Department of Neurobiology, The Weizmann Institute of Science, 76100 Rehovot, Israel.
| | | |
Collapse
|
45
|
Luscher B, Fuchs T, Kilpatrick CL. GABAA receptor trafficking-mediated plasticity of inhibitory synapses. Neuron 2011; 70:385-409. [PMID: 21555068 DOI: 10.1016/j.neuron.2011.03.024] [Citation(s) in RCA: 334] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2011] [Indexed: 12/22/2022]
Abstract
Proper developmental, neural cell-type-specific, and activity-dependent regulation of GABAergic transmission is essential for virtually all aspects of CNS function. The number of GABA(A) receptors in the postsynaptic membrane directly controls the efficacy of GABAergic synaptic transmission. Thus, regulated trafficking of GABA(A) receptors is essential for understanding brain function in both health and disease. Here we summarize recent progress in the understanding of mechanisms that allow dynamic adaptation of cell surface expression and postsynaptic accumulation and function of GABA(A) receptors. This includes activity-dependent and cell-type-specific changes in subunit gene expression, assembly of subunits into receptors, as well as exocytosis, endocytic recycling, diffusion dynamics, and degradation of GABA(A) receptors. In particular, we focus on the roles of receptor-interacting proteins, scaffold proteins, synaptic adhesion proteins, and enzymes that regulate the trafficking and function of receptors and associated proteins. In addition, we review neuropeptide signaling pathways that affect neural excitability through changes in GABA(A)R trafficking.
Collapse
Affiliation(s)
- Bernhard Luscher
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA.
| | | | | |
Collapse
|
46
|
Werner DF, Kumar S, Criswell HE, Suryanarayanan A, Fetzer JA, Comerford CE, Morrow AL. PKCγ is required for ethanol-induced increases in GABA(A) receptor α4 subunit expression in cultured cerebral cortical neurons. J Neurochem 2011; 116:554-63. [PMID: 21155805 DOI: 10.1111/j.1471-4159.2010.07140.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ethanol exposure produces alterations in GABA(A) receptor function and expression associated with CNS hyperexcitability, but the mechanisms of these effects are unknown. Ethanol is known to increase both GABA(A) receptor α4 subunits and protein kinase C (PKC) isozymes in vivo and in vitro. Here, we investigated ethanol regulation of GABA(A) receptor α4 subunit expression in cultured cortical neurons to delineate the role of PKC. Cultured neurons were prepared from rat pups on postnatal day 0-1 and tested after 18 days. GABA(A) receptor α4 subunit surface expression was assessed using P2 fractionation and surface biotinylation following ethanol exposure for 4 h. Miniature inhibitory post-synaptic currents were measured using whole cell patch clamp recordings. Ethanol increased GABA(A) receptor α4 subunit expression in both the P2 and biotinylated fractions, while reducing the decay time constant in miniature inhibitory post-synaptic currents, with no effect on γ2 or δ subunits. PKC activation mimicked ethanol effects, while the PKC inhibitor calphostin C prevented ethanol-induced increases in GABA(A) receptor α4 subunit expression. PKCγ siRNA knockdown prevented ethanol-induced increases in GABA(A) receptor α4 subunit expression, but inhibition of the PKCβ isoform with PKCβ pseudosubstrate had no effect. We conclude that PKCγ regulates ethanol-induced alterations in α4-containing GABA(A) receptors.
Collapse
Affiliation(s)
- David F Werner
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | |
Collapse
|