1
|
Fronza MG, Ferreira BF, Pavan-Silva I, Guimarães FS, Lisboa SF. "NO" Time in Fear Response: Possible Implication of Nitric-Oxide-Related Mechanisms in PTSD. Molecules 2023; 29:89. [PMID: 38202672 PMCID: PMC10779493 DOI: 10.3390/molecules29010089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric condition characterized by persistent fear responses and altered neurotransmitter functioning due to traumatic experiences. Stress predominantly affects glutamate, a neurotransmitter crucial for synaptic plasticity and memory formation. Activation of the N-Methyl-D-Aspartate glutamate receptors (NMDAR) can trigger the formation of a complex comprising postsynaptic density protein-95 (PSD95), the neuronal nitric oxide synthase (nNOS), and its adaptor protein (NOS1AP). This complex is pivotal in activating nNOS and nitric oxide (NO) production, which, in turn, activates downstream pathways that modulate neuronal signaling, including synaptic plasticity/transmission, inflammation, and cell death. The involvement of nNOS and NOS1AP in the susceptibility of PTSD and its comorbidities has been widely shown. Therefore, understanding the interplay between stress, fear, and NO is essential for comprehending the maintenance and progression of PTSD, since NO is involved in fear acquisition and extinction processes. Moreover, NO induces post-translational modifications (PTMs), including S-nitrosylation and nitration, which alter protein function and structure for intracellular signaling. Although evidence suggests that NO influences synaptic plasticity and memory processing, the specific role of PTMs in the pathophysiology of PTSD remains unclear. This review highlights pathways modulated by NO that could be relevant to stress and PTSD.
Collapse
Affiliation(s)
- Mariana G. Fronza
- Pharmacology Departament, Ribeirão Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil; (M.G.F.); (B.F.F.); (I.P.-S.)
| | - Bruna F. Ferreira
- Pharmacology Departament, Ribeirão Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil; (M.G.F.); (B.F.F.); (I.P.-S.)
| | - Isabela Pavan-Silva
- Pharmacology Departament, Ribeirão Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil; (M.G.F.); (B.F.F.); (I.P.-S.)
| | - Francisco S. Guimarães
- Pharmacology Departament, Ribeirão Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil; (M.G.F.); (B.F.F.); (I.P.-S.)
| | - Sabrina F. Lisboa
- Pharmacology Departament, Ribeirão Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil; (M.G.F.); (B.F.F.); (I.P.-S.)
- Biomolecular Sciences Department, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo 14040-903, Brazil
| |
Collapse
|
2
|
Cai CY, Tao Y, Zhou Y, Yang D, Qin C, Bian XL, Xian JY, Cao B, Chang L, Wu HY, Luo CX, Zhu DY. Nos1 + and Nos1 - excitatory neurons in the BLA regulate anxiety- and depression-related behaviors oppositely. J Affect Disord 2023; 333:181-192. [PMID: 37080493 DOI: 10.1016/j.jad.2023.04.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/31/2023] [Accepted: 04/14/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND The basolateral amygdala (BLA) neurons are primarily glutamatergic and have been associated with emotion regulation. However, little is known about the roles of BLA neurons expressing neuronal nitric oxide synthase (nNOS, Nos1) in the regulation of emotional behaviors. METHODS Using Nos1-cre mice and chemogenetic and optogenetic manipulations, we specifically silenced or activated Nos1+ or Nos1- neurons in the BLA, or silenced their projections to the anterdorsal bed nucleus of the stria terminalis (adBNST) and ventral hippocampus (vHPC). We measured anxiety behaviors in elevated plus maze (EPM) and open-field test (OFT), and measured depression behaviors in forced swimming test (FST) and tail suspension test (TST). RESULTS BLA Nos1+ neurons were predominantly glutamatergic, and glutamatergic but not GABAergic Nos1+ neurons were involved in controlling anxiety- and depression-related behaviors. Interestingly, by selectively manipulating the activities of BLA Nos1+ and Nos1- excitatory neurons, we found that they had opposing effects on anxiety- and depression-related behaviors. BLA Nos1+ excitatory neurons projected to the adBNST, this BLA-adBNST circuit controlled the expression of anxiety- and depression-related behaviors, while BLA Nos1- excitatory neurons projected to vHPC, this BLA-vHPC circuit contributed to the expression of anxiety- and depression-related behaviors. Moreover, excitatory vHPC-adBNST circuit antagonized the role of BLA-adBNST circuit in regulating anxiety- and depression-related behaviors. CONCLUSIONS BLA Nos1+ and Nos1- excitatory neuron subpopulations exert different effects on anxiety- and depression-related behaviors through distinct projection circuits, providing a new insight of BLA excitatory neurons in emotional regulation. LIMITATIONS We did not perform retrograde labeling from adBNST and vHPC regions.
Collapse
Affiliation(s)
- Cheng-Yun Cai
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yan Tao
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ying Zhou
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Di Yang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Cheng Qin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xin-Lan Bian
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jia-Yun Xian
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Bo Cao
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lei Chang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hai-Yin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chun-Xia Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Dong-Ya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing 211166, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, China.
| |
Collapse
|
3
|
Candemir E, Fattakhov N, Leary AO, Slattery DA, Courtney MJ, Reif A, Freudenberg F. Disrupting the nNOS/NOS1AP interaction in the medial prefrontal cortex impairs social recognition and spatial working memory in mice. Eur Neuropsychopharmacol 2023; 67:66-79. [PMID: 36513018 DOI: 10.1016/j.euroneuro.2022.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/10/2022] [Accepted: 11/13/2022] [Indexed: 12/14/2022]
Abstract
The neuronal isoform of nitric oxide synthase (nNOS) and its interacting protein NOS1AP have been linked to several mental disorders including schizophrenia and depression. An increase in the interaction between nNOS and NOS1AP in the frontal cortex has been suggested to contribute to the emergence of these disorders. Here we aimed to uncover whether disruption of their interactions in the frontal cortex leads to mental disorder endophenotypes. Targeting the medial prefrontal cortex (mPFC), we stereotaxically injected wild-type C57BL/6J mice with recombinant adeno-associated virus (rAAV) expressing either full-length NOS1AP, the nNOS binding region of NOS1AP (i.e. NOS1AP396-503), or the nNOS amino-terminus (i.e. nNOS1-133), which was shown to disrupt the interaction of endogenous nNOS with PSD-95. We tested these mice in a comprehensive behavioural battery, assessing different endophenotypes related to mental disorders. We found no differences in anxiety-related and exploratory behaviours. Likewise, social interaction was comparable in all groups. However, social recognition was impaired in NOS1AP and NOS1AP396-503 mice. These mice, as well as mice overexpressing nNOS1-133 also displayed impaired spatial working memory (SWM) capacity, while spatial reference memory (SRM) remained intact. Finally, mice overexpressing NOS1AP and nNOS1-133, but not NOS1AP396-503, failed to habituate to the startling pulses in an acoustic startle response (ASR) paradigm, though we found no difference in overall startle intensity or prepulse inhibition (PPI) of the ASR. Our findings indicate a distinct role of NOS1AP/nNOS/PSD-95 interactions in the mPFC to contribute to specific endophenotypic changes observed in different mental disorders.
Collapse
Affiliation(s)
- Esin Candemir
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany; Graduate School of Life Sciences, University of Würzburg, Würzburg, Germany
| | - Nikolai Fattakhov
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany
| | - Aet O Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany
| | - Michael J Courtney
- Neuronal Signalling Laboratory, Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany
| | - Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Laboratory of Translational Psychiatry, Heinrich-Hoffmann-Straße 10, 60528 Frankfurt am Main, Germany.
| |
Collapse
|
4
|
Stieve BJ, Smith MM, Krook-Magnuson E. LINCs Are Vulnerable to Epileptic Insult and Fail to Provide Seizure Control via On-Demand Activation. eNeuro 2023; 10:ENEURO.0195-22.2022. [PMID: 36725340 PMCID: PMC9933934 DOI: 10.1523/eneuro.0195-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 02/03/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is notoriously pharmacoresistant, and identifying novel therapeutic targets for controlling seizures is crucial. Long-range inhibitory neuronal nitric oxide synthase-expressing cells (LINCs), a population of hippocampal neurons, were recently identified as a unique source of widespread inhibition in CA1, able to elicit both GABAA-mediated and GABAB-mediated postsynaptic inhibition. We therefore hypothesized that LINCs could be an effective target for seizure control. LINCs were optogenetically activated for on-demand seizure intervention in the intrahippocampal kainate (KA) mouse model of chronic TLE. Unexpectedly, LINC activation at 1 month post-KA did not substantially reduce seizure duration in either male or female mice. We tested two different sets of stimulation parameters, both previously found to be effective with on-demand optogenetic approaches, but neither was successful. Quantification of LINCs following intervention revealed a substantial reduction of LINC numbers compared with saline-injected controls. We also observed a decreased number of LINCs when the site of initial insult (i.e., KA injection) was moved to the amygdala [basolateral amygdala (BLA)-KA], and correspondingly, no effect of light delivery on BLA-KA seizures. This indicates that LINCs may be a vulnerable population in TLE, regardless of the site of initial insult. To determine whether long-term circuitry changes could influence outcomes, we continued testing once a month for up to 6 months post-KA. However, at no time point did LINC activation provide meaningful seizure suppression. Altogether, our results suggest that LINCs are not a promising target for seizure inhibition in TLE.
Collapse
Affiliation(s)
- Bethany J Stieve
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Madison M Smith
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Esther Krook-Magnuson
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
5
|
Haider AA, Rex TS, Wareham LK. cGMP Signaling in the Neurovascular Unit-Implications for Retinal Ganglion Cell Survival in Glaucoma. Biomolecules 2022; 12:1671. [PMID: 36421684 PMCID: PMC9687235 DOI: 10.3390/biom12111671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Glaucoma is a progressive age-related disease of the visual system and the leading cause of irreversible blindness worldwide. Currently, intraocular pressure (IOP) is the only modifiable risk factor for the disease, but even as IOP is lowered, the pathology of the disease often progresses. Hence, effective clinical targets for the treatment of glaucoma remain elusive. Glaucoma shares comorbidities with a multitude of vascular diseases, and evidence in humans and animal models demonstrates an association between vascular dysfunction of the retina and glaucoma pathology. Integral to the survival of retinal ganglion cells (RGCs) is functional neurovascular coupling (NVC), providing RGCs with metabolic support in response to neuronal activity. NVC is mediated by cells of the neurovascular unit (NVU), which include vascular cells, glial cells, and neurons. Nitric oxide-cyclic guanosine monophosphate (NO-cGMP) signaling is a prime mediator of NVC between endothelial cells and neurons, but emerging evidence suggests that cGMP signaling is also important in the physiology of other cells of the NVU. NO-cGMP signaling has been implicated in glaucomatous neurodegeneration in humans and mice. In this review, we explore the role of cGMP signaling in the different cell types of the NVU and investigate the potential links between cGMP signaling, breakdown of neurovascular function, and glaucoma pathology.
Collapse
Affiliation(s)
| | | | - Lauren K. Wareham
- Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
6
|
Sun N, Qin YJ, Xu C, Xia T, Du ZW, Zheng LP, Li AA, Meng F, Zhang Y, Zhang J, Liu X, Li TY, Zhu DY, Zhou QG. Design of fast-onset antidepressant by dissociating SERT from nNOS in the DRN. Science 2022; 378:390-398. [PMID: 36302033 DOI: 10.1126/science.abo3566] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Major depressive disorder (MDD) is one of the most common mental disorders. We designed a fast-onset antidepressant that works by disrupting the interaction between the serotonin transporter (SERT) and neuronal nitric oxide synthase (nNOS) in the dorsal raphe nucleus (DRN). Chronic unpredictable mild stress (CMS) selectively increased the SERT-nNOS complex in the DRN in mice. Augmentation of SERT-nNOS interactions in the DRN caused a depression-like phenotype and accounted for the CMS-induced depressive behaviors. Disrupting the SERT-nNOS interaction produced a fast-onset antidepressant effect by enhancing serotonin signaling in forebrain circuits. We discovered a small-molecule compound, ZZL-7, that elicited an antidepressant effect 2 hours after treatment without undesirable side effects. This compound, or analogous reagents, may serve as a new, rapidly acting treatment for MDD.
Collapse
Affiliation(s)
- Nan Sun
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Ya-Juan Qin
- Department of Pharmacochemistry, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chu Xu
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Institute of Dermatology, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| | - Tian Xia
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Zi-Wei Du
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Li-Ping Zheng
- Department of Pharmacochemistry, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - An-An Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xu Zhou 221004, China
| | - Fan Meng
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yu Zhang
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jing Zhang
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiao Liu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ting-You Li
- Department of Pharmacochemistry, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Dong-Ya Zhu
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- The Key Center of Gene Technology Drugs of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Qi-Gang Zhou
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- The Key Center of Gene Technology Drugs of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
- Department of Clinic Pharmacology, Sir runrun Hospital, Nanjing Medical University, Nanjing 211167, China
| |
Collapse
|
7
|
da Silva LA, Diniz CRAF, Uliana DL, da Silva-Júnior AF, Bertacchini GL, Resstel LBM. The interaction between hippocampal cholinergic and nitrergic neurotransmission coordinates NMDA-dependent behavior and autonomic changes induced by contextual fear retrieval. Psychopharmacology (Berl) 2022; 239:3297-3311. [PMID: 35978221 DOI: 10.1007/s00213-022-06213-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022]
Abstract
RATIONALE Re-exposing an animal to an environment previously paired with an aversive stimulus evokes large alterations in behavioral and cardiovascular parameters. Dorsal hippocampus (dHC) receives important cholinergic inputs from the basal forebrain, and respective acetylcholine (ACh) levels are described to influence defensive behavior. Activation of muscarinic M1 and M3 receptors facilitates autonomic and behavioral responses along threats. Evidence show activation of cholinergic receptors promoting formation of nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) in dHC. Altogether, the action of ACh and NO on conditioned responses appears to converge within dHC. OBJECTIVES As answer about how ACh and NO interact to modulate defensive responses has so far been barely addressed, we aimed to shed additional light on this topic. METHODS Male Wistar rats had guide cannula implanted into the dHC before being submitted to the contextual fear conditioning (3footshocks/085 mA/2 s). A catheter was implanted in the femoral artery the next day for cardiovascular recordings. Drugs were delivered into dHC 10 min before contextual re-exposure, which occurred 48 h after the conditioning procedure. RESULTS Neostigmine (Neo) amplified the retrieval of conditioned responses. Neo effects (1 nmol) were prevented by the prior infusion of a M1-M3 antagonist (fumarate), a neuronal nitric oxide synthase inhibitor (NPLA), a NO scavenger (cPTIO), a guanylyl cyclase inhibitor (ODQ), and a NMDA antagonist (AP-7). Pretreatment with a selective M1 antagonist (pirenzepine) only prevented the increase in autonomic responses induced by Neo. CONCLUSION The results show that modulation in the retrieval of contextual fear responses involves coordination of the dHC M1-M3/NO/cGMP/NMDA pathway.
Collapse
Affiliation(s)
- Leandro Antero da Silva
- Department of Pharmacology, School of Medicine, Universidade de Sao Paulo, Campus USP, Bandeirantes Avenue, Monte Alegre, Ribeirão Preto, SP, 14049-900, Brazil
- State University of Mato Grosso Do Sul - Medicine UEMS, Mato Grosso Do Sul, Campo Grande, Brazil
| | - Cassiano Ricardo Alves Faria Diniz
- Department of Pharmacology, School of Medicine, Universidade de Sao Paulo, Campus USP, Bandeirantes Avenue, Monte Alegre, Ribeirão Preto, SP, 14049-900, Brazil
| | - Daniela Lescano Uliana
- Department of Pharmacology, School of Medicine, Universidade de Sao Paulo, Campus USP, Bandeirantes Avenue, Monte Alegre, Ribeirão Preto, SP, 14049-900, Brazil
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA, 15260, USA
| | - Antonio Furtado da Silva-Júnior
- Department of Pharmacology, School of Medicine, Universidade de Sao Paulo, Campus USP, Bandeirantes Avenue, Monte Alegre, Ribeirão Preto, SP, 14049-900, Brazil
| | - Gabriela Luiz Bertacchini
- Department of Pharmacology, School of Medicine, Universidade de Sao Paulo, Campus USP, Bandeirantes Avenue, Monte Alegre, Ribeirão Preto, SP, 14049-900, Brazil
| | - Leonardo Barbosa Moraes Resstel
- Department of Pharmacology, School of Medicine, Universidade de Sao Paulo, Campus USP, Bandeirantes Avenue, Monte Alegre, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
8
|
Sadeghi MA, Hemmati S, Nassireslami E, Yousefi Zoshk M, Hosseini Y, Abbasian K, Chamanara M. Targeting neuronal nitric oxide synthase and the nitrergic system in post-traumatic stress disorder. Psychopharmacology (Berl) 2022; 239:3057-3082. [PMID: 36029333 DOI: 10.1007/s00213-022-06212-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/04/2022] [Indexed: 12/22/2022]
Abstract
RATIONALE Current pharmacological approaches to treatment of post-traumatic stress disorder (PTSD) lack adequate effectiveness. As a result, identifying new molecular targets for drug development is necessary. Furthermore, fear learning and memory in PTSD can undergo different phases, such as fear acquisition, consolidation, and extinction. Each phase may involve different cellular pathways and brain regions. As a result, effective management of PTSD requires mindfulness of the timing of drug administration. One of the molecular targets currently under intense investigation is the N-methyl-D-aspartate (NMDA)-type glutamate receptor (NMDAR). However, despite the therapeutic efficacy of drugs targeting NMDAR, their translation into clinical use has been challenging due to their various side effects. One possible solution to this problem is to target signaling proteins downstream to NMDAR to improve targeting specificity. One of these proteins is the neuronal nitric oxide synthase (nNOS), which is activated following calcium influx through the NMDAR. OBJECTIVE In this paper, we review the literature on the pharmacological modulation of nNOS in animal models of PTSD to evaluate its therapeutic potential. Furthermore, we attempt to decipher the inconsistencies observed between the findings of these studies based on the specific phase of fear learning which they had targeted. RESULTS Inhibition of nNOS may inhibit fear acquisition and recall, while not having a significant effect on fear consolidation and extinction. However, it may improve extinction consolidation or reconsolidation blockade. CONCLUSIONS Modulation of nNOS has therapeutic potential against PTSD and warrants further development for use in the clinical setting.
Collapse
Affiliation(s)
- Mohammad Amin Sadeghi
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Sara Hemmati
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Nassireslami
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | | - Yasaman Hosseini
- Cognitive Neuroscience Center, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Kourosh Abbasian
- Management and Health Economics Department, AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran. .,Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Jehle A, Garaschuk O. The Interplay between cGMP and Calcium Signaling in Alzheimer's Disease. Int J Mol Sci 2022; 23:7048. [PMID: 35806059 PMCID: PMC9266933 DOI: 10.3390/ijms23137048] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/31/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Cyclic guanosine monophosphate (cGMP) is a ubiquitous second messenger and a key molecule in many important signaling cascades in the body and brain, including phototransduction, olfaction, vasodilation, and functional hyperemia. Additionally, cGMP is involved in long-term potentiation (LTP), a cellular correlate of learning and memory, and recent studies have identified the cGMP-increasing drug Sildenafil as a potential risk modifier in Alzheimer's disease (AD). AD development is accompanied by a net increase in the expression of nitric oxide (NO) synthases but a decreased activity of soluble guanylate cyclases, so the exact sign and extent of AD-mediated imbalance remain unclear. Moreover, human patients and mouse models of the disease present with entangled deregulation of both cGMP and Ca2+ signaling, e.g., causing changes in cGMP-mediated Ca2+ release from the intracellular stores as well as Ca2+-mediated cGMP production. Still, the mechanisms governing such interplay are poorly understood. Here, we review the recent data on mechanisms underlying the brain cGMP signaling and its interconnection with Ca2+ signaling. We also discuss the recent evidence stressing the importance of such interplay for normal brain function as well as in Alzheimer's disease.
Collapse
Affiliation(s)
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany;
| |
Collapse
|
10
|
Hashemi M, Karami M, Zarrindast MR. The regulatory role of nitric oxide in morphine-induced analgesia in the descending path of pain from the dorsal hippocampus to the dorsolateral periaqueductal gray. Eur J Pain 2022; 26:888-901. [PMID: 35090066 DOI: 10.1002/ejp.1916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 12/30/2021] [Accepted: 01/23/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Nitric oxide (NO) levels in brain nuclei, such as the hippocampus and brainstem, are involved in morphine analgesia, but the relationship between the dorsal hippocampus (dH) and the dorsolateral periaqueductal gray matter (dlPAG) needs to be clarified, which is our goal. METHODS Wistar rats were simultaneously equipped with a stereotaxic device with unilateral guide cannula at dH and dlPAG. After recovery, they were divided into control and experimental groups. Formalin (50 μL of 2.5%) was inoculated into the left hind paw of rat. Morphine (6 mg/kg) was administered intraperitoneally (i.p.) 10 min before formalin injection. L-Arginine (0.25, 0.5, 1 and 2 μg/rat), and L-NAME (0.25, 0.5, 1 and 2 μg/rat), unrelatedly or with the respect in the order of injection were used in the nuclei before morphine injection (i.p.). Activation of the neuronal NO synthase (nNOS) in the brains of all animals was measured using NADPH-diaphorase, a selective biochemical marker of nNOS. RESULTS Morphine reduced inflammatory pain in the early and late stages of the rat formalin test. The morphine response was attenuated by before injection of single L-arginine but not L-NAME in the two target areas. However, the acute phase result was stopped due to L-NAME pretreatment. When L-NAME was injected into dlPAG before injecting L-arginine at dH, the morphine response did not decrease at all, indicating a modulatory role of NO in dlPAG, which was confirmed by NADPH-d staining. CONCLUSIONS High levels of NO in dlPAG may regulate pain process in downward synaptic interactions.
Collapse
Affiliation(s)
- Mahboobeh Hashemi
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | - Manizheh Karami
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | | |
Collapse
|
11
|
Al-Amin MM, Sullivan RKP, Alexander S, Carter DA, Bradford D, Burne THJ, Burne THJ. Impaired spatial memory in adult vitamin D deficient BALB/c mice is associated with reductions in spine density, nitric oxide, and neural nitric oxide synthase in the hippocampus. AIMS Neurosci 2022; 9:31-56. [PMID: 35434279 PMCID: PMC8941191 DOI: 10.3934/neuroscience.2022004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Vitamin D deficiency is prevalent in adults and is associated with cognitive impairment. However, the mechanism by which adult vitamin D (AVD) deficiency affects cognitive function remains unclear. We examined spatial memory impairment in AVD-deficient BALB/c mice and its underlying mechanism by measuring spine density, long term potentiation (LTP), nitric oxide (NO), neuronal nitric oxide synthase (nNOS), and endothelial NOS (eNOS) in the hippocampus. Adult male BALB/c mice were fed a control or vitamin D deficient diet for 20 weeks. Spatial memory performance was measured using an active place avoidance (APA) task, where AVD-deficient mice had reduced latency entering the shock zone compared to controls. We characterised hippocampal spine morphology in the CA1 and dentate gyrus (DG) and made electrophysiological recordings in the hippocampus of behaviourally naïve mice to measure LTP. We next measured NO, as well as glutathione, lipid peroxidation and oxidation of protein products and quantified hippocampal immunoreactivity for nNOS and eNOS. Spine morphology analysis revealed a significant reduction in the number of mushroom spines in the CA1 dendrites but not in the DG. There was no effect of diet on LTP. However, hippocampal NO levels were depleted whereas other oxidation markers were unaltered by AVD deficiency. We also showed a reduced nNOS, but not eNOS, immunoreactivity. Finally, vitamin D supplementation for 10 weeks to AVD-deficient mice restored nNOS immunoreactivity to that seen in in control mice. Our results suggest that lower levels of NO and reduced nNOS immunostaining contribute to hippocampal-dependent spatial learning deficits in AVD-deficient mice.
Collapse
Affiliation(s)
- Md. Mamun Al-Amin
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia
| | | | - Suzy Alexander
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia,Queensland Centre for Mental Health Research, Wacol 4076, Australia
| | - David A. Carter
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia
| | - DanaKai Bradford
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia,Australian E-Health Research Centre, CSIRO, Pullenvale 4069, Australia
| | - Thomas H. J. Burne
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia,Queensland Centre for Mental Health Research, Wacol 4076, Australia,* Correspondence: ; Tel: +61 733466371; Fax: +61 733466301
| | | | | | | |
Collapse
|
12
|
Hippocampal overexpression of NOS1AP promotes endophenotypes related to mental disorders. EBioMedicine 2021; 71:103565. [PMID: 34455393 PMCID: PMC8403735 DOI: 10.1016/j.ebiom.2021.103565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/03/2021] [Accepted: 08/17/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Nitric oxide synthase 1 adaptor protein (NOS1AP; previously named CAPON) is linked to the glutamatergic postsynaptic density through interaction with neuronal nitric oxide synthase (nNOS). NOS1AP and its interaction with nNOS have been associated with several mental disorders. Despite the high levels of NOS1AP expression in the hippocampus and the relevance of this brain region in glutamatergic signalling as well as mental disorders, a potential role of hippocampal NOS1AP in the pathophysiology of these disorders has not been investigated yet. METHODS To uncover the function of NOS1AP in hippocampus, we made use of recombinant adeno-associated viruses to overexpress murine full-length NOS1AP or the NOS1AP carboxyterminus in the hippocampus of mice. We investigated these mice for changes in gene expression, neuronal morphology, and relevant behavioural phenotypes. FINDINGS We found that hippocampal overexpression of NOS1AP markedly increased the interaction of nNOS with PSD-95, reduced dendritic spine density, and changed dendritic spine morphology at CA1 synapses. At the behavioural level, we observed an impairment in social memory and decreased spatial working memory capacity. INTERPRETATION Our data provide a mechanistic explanation for a highly selective and specific contribution of hippocampal NOS1AP and its interaction with the glutamatergic postsynaptic density to cross-disorder pathophysiology. Our findings allude to therapeutic relevance due to the druggability of this molecule. FUNDING This study was funded in part by the DFG, the BMBF, the Academy of Finland, the NIH, the Japanese Society of Clinical Neuropsychopharmacology, the Ministry of Education of the Russian Federation, and the European Community.
Collapse
|
13
|
Presynaptic NK1 Receptor Activation by Substance P Suppresses EPSCs via Nitric Oxide Synthesis in the Rat Insular Cortex. Neuroscience 2021; 455:151-164. [DOI: 10.1016/j.neuroscience.2020.12.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 01/28/2023]
|
14
|
McLeod F, Boyle K, Marzo A, Martin-Flores N, Moe TZ, Palomer E, Gibb AJ, Salinas PC. Wnt Signaling Through Nitric Oxide Synthase Promotes the Formation of Multi-Innervated Spines. Front Synaptic Neurosci 2020; 12:575863. [PMID: 33013349 PMCID: PMC7509412 DOI: 10.3389/fnsyn.2020.575863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/13/2020] [Indexed: 01/06/2023] Open
Abstract
Structural plasticity of synapses correlates with changes in synaptic strength. Dynamic modifications in dendritic spine number and size are crucial for long-term potentiation (LTP), the cellular correlate of learning and memory. Recent studies have suggested the generation of multi-innervated spines (MIS), in the form of several excitatory presynaptic inputs onto one spine, are crucial for hippocampal memory storage. However, little is known about the molecular mechanisms underlying MIS formation and their contribution to LTP. Using 3D enhanced resolution confocal images, we examined the contribution of Wnt synaptic modulators in MIS formation in the context of LTP. We show that blockage of endogenous Wnts with specific Wnt antagonists supresses the formation of MIS upon chemical LTP induction in cultured hippocampal neurons. Gain- and loss-of-function studies demonstrate that Wnt7a signaling promotes MIS formation through the postsynaptic Wnt scaffold protein Disheveled 1 (Dvl1) by stimulating neuronal nitric oxide (NO) synthase (nNOS). Subsequently, NO activates soluble guanylyl cyclase (sGC) to increase MIS formation. Consistently, we observed an enhanced frequency and amplitude of excitatory postsynaptic currents. Collectively, our findings identify a unique role for Wnt secreted proteins through nNOS/NO/sGC signaling to modulate MIS formation during LTP.
Collapse
Affiliation(s)
- Faye McLeod
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Kieran Boyle
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Aude Marzo
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Nuria Martin-Flores
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Thaw Zin Moe
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Ernest Palomer
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Alasdair J Gibb
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Patricia C Salinas
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
15
|
Yılmaz O, Soygüder Z, Keleş ÖF, Yaman T, Yener Z, Uyar A, Çakır T. An Immunohistochemical Study on the Presence of Nitric Oxide Synthase Isoforms (nNOS, iNOS, eNOS) in the Spinal Cord and Nodose Ganglion of Rats Receiving Ionising Gamma Radiation to their Liver. J Vet Res 2020; 64:445-453. [PMID: 32984637 PMCID: PMC7497755 DOI: 10.2478/jvetres-2020-0059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 08/25/2020] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION This study determined the presence of nitric oxide synthesis isoforms (nNOS, iNOS, and eNOS) in thoracic spinal cord segments and nodose ganglia of rats with gamma-irradiated livers. MATERIAL AND METHODS Male rats (n = 32) were divided into equal groups A, B, C, and D. In group A, the controls, no radiation was applied, while groups B, C, and D received 10 Gy of ionising gamma radiation. The rats of group B were euthanized at the end of the first day (d1), those of group C on the second day (d2), and those of group D on the third day (d3). The liver, spinal cord segments, and nodose ganglion tissues were dissected and fixed, and the liver sections were examined histopathologically. The other tissues were observed through a light microscope. RESULTS Regeneration occurred at the end of d3 in hepatocytes which were radiation-damaged at the end of d1 and d2. On d1, some nNOS-positive staining was found in the neuronal cells of laminae I-III of the spinal cord and in neurons of the nodose ganglion, and on d3, some staining was observed in lamina X of the spinal cord, while none of note was in the nodose ganglion. Dense iNOS-positive staining was seen on d1 in the ependymal cells of the spinal cord and in the glial cells of the nodose ganglion, and on d3, there was still considerable iNOS staining in both tissues. There was clear eNOS-positive staining in the capillary endothelial cells of the spinal cord and light diffuse cytoplasmic staining in the neurons of the nodose ganglion on d1, and on d3, intense eNOS-positive staining was visible in several endothelial cells of the spinal cord, while light nuclear staining was recognised in the neurons of the nodose ganglion. CONCLUSION The nNOS, iNOS, and eNOS isoforms are activated in the spinal cord and nodose ganglion of rats after ionising radiation insult to the liver.
Collapse
Affiliation(s)
| | | | - Ömer Faruk Keleş
- Department of Pathology, Faculty of Veterinary Medicine, University of Van Yüzüncü Yıl, 65080, Van, Turkey
| | - Turan Yaman
- Department of Pathology, Faculty of Veterinary Medicine, University of Van Yüzüncü Yıl, 65080, Van, Turkey
| | - Zabit Yener
- Department of Pathology, Faculty of Veterinary Medicine, University of Van Yüzüncü Yıl, 65080, Van, Turkey
| | - Ahmet Uyar
- Department of Pathology, Faculty of Veterinary Medicine, University of Hatay Mustafa Kemal, 31040, Hatay, Turkey
| | - Tahir Çakır
- Department of Medical Physics, Faculty of Medicine, University of Van Yüzüncü Yıl, 65080, Van, Turkey
| |
Collapse
|
16
|
Kelly MP, Heckman PRA, Havekes R. Genetic manipulation of cyclic nucleotide signaling during hippocampal neuroplasticity and memory formation. Prog Neurobiol 2020; 190:101799. [PMID: 32360536 DOI: 10.1016/j.pneurobio.2020.101799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/14/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Decades of research have underscored the importance of cyclic nucleotide signaling in memory formation and synaptic plasticity. In recent years, several new genetic techniques have expanded the neuroscience toolbox, allowing researchers to measure and modulate cyclic nucleotide gradients with high spatiotemporal resolution. Here, we will provide an overview of studies using genetic approaches to interrogate the role cyclic nucleotide signaling plays in hippocampus-dependent memory processes and synaptic plasticity. Particular attention is given to genetic techniques that measure real-time changes in cyclic nucleotide levels as well as newly-developed genetic strategies to transiently manipulate cyclic nucleotide signaling in a subcellular compartment-specific manner with high temporal resolution.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, VA Bldg1, 3(rd) Fl, D-12, Columbia, 29209, SC, USA.
| | - Pim R A Heckman
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| |
Collapse
|
17
|
Argyrousi EK, Heckman PRA, Prickaerts J. Role of cyclic nucleotides and their downstream signaling cascades in memory function: Being at the right time at the right spot. Neurosci Biobehav Rev 2020; 113:12-38. [PMID: 32044374 DOI: 10.1016/j.neubiorev.2020.02.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/23/2020] [Accepted: 02/03/2020] [Indexed: 01/23/2023]
Abstract
A plethora of studies indicate the important role of cAMP and cGMP cascades in neuronal plasticity and memory function. As a result, altered cyclic nucleotide signaling has been implicated in the pathophysiology of mnemonic dysfunction encountered in several diseases. In the present review we provide a wide overview of studies regarding the involvement of cyclic nucleotides, as well as their upstream and downstream molecules, in physiological and pathological mnemonic processes. Next, we discuss the regulation of the intracellular concentration of cyclic nucleotides via phosphodiesterases, the enzymes that degrade cAMP and/or cGMP, and via A-kinase-anchoring proteins that refine signal compartmentalization of cAMP signaling. We also provide an overview of the available data pointing to the existence of specific time windows in cyclic nucleotide signaling during neuroplasticity and memory formation and the significance to target these specific time phases for improving memory formation. Finally, we highlight the importance of emerging imaging tools like Förster resonance energy transfer imaging and optogenetics in detecting, measuring and manipulating the action of cyclic nucleotide signaling cascades.
Collapse
Affiliation(s)
- Elentina K Argyrousi
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands
| | - Pim R A Heckman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands.
| |
Collapse
|
18
|
Ivanova VO, Balaban PM, Bal NV. Modulation of AMPA Receptors by Nitric Oxide in Nerve Cells. Int J Mol Sci 2020; 21:ijms21030981. [PMID: 32024149 PMCID: PMC7038066 DOI: 10.3390/ijms21030981] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) is a gaseous molecule with a large number of functions in living tissue. In the brain, NO participates in numerous intracellular mechanisms, including synaptic plasticity and cell homeostasis. NO elicits synaptic changes both through various multi-chain cascades and through direct nitrosylation of targeted proteins. Along with the N-methyl-d-aspartate (NMDA) glutamate receptors, one of the key components in synaptic functioning are α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptors—the main target for long-term modifications of synaptic effectivity. AMPA receptors have been shown to participate in most of the functions important for neuronal activity, including memory formation. Interactions of NO and AMPA receptors were observed in important phenomena, such as glutamatergic excitotoxicity in retinal cells, synaptic plasticity, and neuropathologies. This review focuses on existing findings that concern pathways by which NO interacts with AMPA receptors, influences properties of different subunits of AMPA receptors, and regulates the receptors’ surface expression.
Collapse
|
19
|
Gambino G, Rizzo V, Giglia G, Ferraro G, Sardo P. Cannabinoids, TRPV and nitric oxide: the three ring circus of neuronal excitability. Brain Struct Funct 2019; 225:1-15. [PMID: 31792694 DOI: 10.1007/s00429-019-01992-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022]
Abstract
Endocannabinoid system is considered a relevant player in the regulation of neuronal excitability, since it contributes to maintaining the balance of the synaptic ionic milieu. Perturbations to bioelectric conductances have been implicated in the pathophysiological processes leading to hyperexcitability and epileptic seizures. Cannabinoid influence on neurosignalling is exerted on classic receptor-mediated mechanisms or on further molecular targets. Among these, transient receptor potential vanilloid (TRPV) are ionic channels modulated by cannabinoids that are involved in the transduction of a plethora of stimuli and trigger fundamental downstream pathways in the post-synaptic site. In this review, we aim at providing a brief summary of the most recent data about the cross-talk between cannabinoid system and TRPV channels, drawing attention on their role on neuronal hyperexcitability. Then, we aim to unveil a plausible point of interaction between these neural signalling systems taking into consideration nitric oxide, a gaseous molecule inducing profound modifications to neural performances. From this novel perspective, we struggle to propose innovative cellular mechanisms in the regulation of hyperexcitability phenomena, with the goal of exploring plausible CB-related mechanisms underpinning epileptic seizures.
Collapse
Affiliation(s)
- Giuditta Gambino
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy.
| | - Valerio Rizzo
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Giuseppe Giglia
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Giuseppe Ferraro
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Pierangelo Sardo
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| |
Collapse
|
20
|
Giesen J, Füchtbauer EM, Füchtbauer A, Funke K, Koesling D, Russwurm M. AMPA Induces NO-Dependent cGMP Signals in Hippocampal and Cortical Neurons via L-Type Voltage-Gated Calcium Channels. Cereb Cortex 2019; 30:2128-2143. [DOI: 10.1093/cercor/bhz227] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/28/2019] [Accepted: 08/28/2019] [Indexed: 02/06/2023] Open
Abstract
AbstractThe nitric oxide (NO)/cGMP signaling cascade has an established role in synaptic plasticity. However, with conventional methods, the underlying cGMP signals were barely detectable. Here, we set out to confirm the well-known NMDA-induced cGMP increases, to test the impact of AMPA on those signals, and to identify the relevant phosphodiesterases (PDEs) using a more sensitive fluorescence resonance energy transfer (FRET)-based method. Therefore, a “knock-in” mouse was generated that expresses a FRET-based cGMP indicator (cGi-500) allowing detection of cGMP concentrations between 100 nM and 3 μM. Measurements were performed in cultured hippocampal and cortical neurons as well as acute hippocampal slices. In hippocampal and cortical neurons, NMDA elicited cGMP signals half as high as the ones elicited by exogenous NO. Interestingly, AMPA increased cGMP independently of NMDA receptors and dependent on NO synthase (NOS) activation. NMDA- and AMPA-induced cGMP signals were not additive indicating that both pathways converge on the level of NOS. Accordingly, the same PDEs, PDE1 and PDE2, were responsible for degradation of NMDA- as well as AMPA-induced cGMP signals. Mechanistically, AMPAR induced calcium influx through L-type voltage-gated calcium channels leading to NOS and finally NO-sensitive guanylyl cyclase activation. Our results demonstrate that in addition to NMDA also AMPA triggers endogenous NO formation and hence cGMP production.
Collapse
Affiliation(s)
- Jan Giesen
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Ernst-Martin Füchtbauer
- Molecular Cell and Developmental Biology, Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Annette Füchtbauer
- Molecular Cell and Developmental Biology, Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Klaus Funke
- Department of Neurophysiology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Doris Koesling
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Michael Russwurm
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, 44780 Bochum, Germany
| |
Collapse
|
21
|
Multi-input Synapses, but Not LTP-Strengthened Synapses, Correlate with Hippocampal Memory Storage in Aged Mice. Curr Biol 2019; 29:3600-3610.e4. [PMID: 31630953 PMCID: PMC6839404 DOI: 10.1016/j.cub.2019.08.064] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/05/2019] [Accepted: 08/22/2019] [Indexed: 12/18/2022]
Abstract
Long-lasting changes at synapses enable memory storage in the brain. Although aging is associated with impaired memory formation, it is not known whether the synaptic underpinnings of memory storage differ with age. Using a training schedule that results in the same behavioral memory formation in young and aged mice, we examined synapse ultrastructure and molecular signaling in the hippocampus after contextual fear conditioning. Only in young, but not old mice, contextual fear memory formation was associated with synaptic changes that characterize well-known, long-term potentiation, a strengthening of existing synapses with one input. Instead, old-age memory was correlated with generation of multi-innervated dendritic spines (MISs), which are predominantly two-input synapses formed by the attraction of an additional excitatory, presynaptic terminal onto an existing synapse. Accordingly, a blocker used to inhibit MIS generation impaired contextual fear memory only in old mice. Our results reveal how the synaptic basis of hippocampal memory storage changes with age and suggest that these distinct memory-storing mechanisms may explain impaired updating in old age. Aged mice form contextual memory like young mice, but reconsolidation is impaired Only in young mice is contextual memory formation associated with structural LTP In aged mice, contextual memory formation correlates with multi-innervated spines Inhibition of multi-innervated spines impairs memory in aged but not young mice
Collapse
|
22
|
Christenson Wick Z, Tetzlaff MR, Krook-Magnuson E. Novel long-range inhibitory nNOS-expressing hippocampal cells. eLife 2019; 8:46816. [PMID: 31609204 PMCID: PMC6839902 DOI: 10.7554/elife.46816] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022] Open
Abstract
The hippocampus, a brain region that is important for spatial navigation and episodic memory, benefits from a rich diversity of neuronal cell-types. Through the use of an intersectional genetic viral vector approach in mice, we report novel hippocampal neurons which we refer to as LINCs, as they are long-range inhibitory neuronal nitric oxide synthase (nNOS)-expressing cells. LINCs project to several extrahippocampal regions including the tenia tecta, diagonal band, and retromammillary nucleus, but also broadly target local CA1 cells. LINCs are thus both interneurons and projection neurons. LINCs display regular spiking non-pyramidal firing patterns, are primarily located in the stratum oriens or pyramidale, have sparsely spiny dendrites, and do not typically express somatostatin, VIP, or the muscarinic acetylcholine receptor M2. We further demonstrate that LINCs can strongly influence hippocampal function and oscillations, including interregional coherence. The identification and characterization of these novel cells advances our basic understanding of both hippocampal circuitry and neuronal diversity.
Collapse
Affiliation(s)
- Zoé Christenson Wick
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, United States
| | - Madison R Tetzlaff
- Neuroscience Department, University of Minnesota, Minneapolis, United States
| | | |
Collapse
|
23
|
Hernández-Melesio MA, Alcaraz-Zubeldia M, Jiménez-Capdeville ME, Martínez-Lazcano JC, Santoyo-Pérez ME, Quevedo-Corona L, Gerónimo-Olvera C, Sánchez-Mendoza A, Ríos C, Pérez-Severiano F. Nitric oxide donor molsidomine promotes retrieval of object recognition memory in a model of cognitive deficit induced by 192 IgG-saporin. Behav Brain Res 2019; 366:108-117. [PMID: 30898683 DOI: 10.1016/j.bbr.2019.03.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) plays a leading role in learning and memory processes. Previously, we showed its ability to modify the deleterious effect of immunotoxin 192 IgG-saporin (192-IgG-SAP) in the cholinergic system. The aim of this study was to analyze the potential of a NO donor (molsidomine, MOLS) to prevent the recognition memory deficits resulting from the septal cholinergic denervation by 192 IgG-SAP in rats. Quantification of neuronal and endothelial nitric oxide synthase (nNOS and eNOS, respectively) expression was evaluated in striatum, prefrontal cortex, and hippocampus. In addition, a choline acetyltransferase immunohistochemical analysis was performed in medial septum and assessed the effect of MOLS treatment on the spatial working memory of rats through a recognition memory test. Results showed that 192-IgG-SAP reduced the immunoreactivity of cholinergic septal neurons (41%), compared with PBS-receiving control rats (p < 0.05). Treatment with MOLS alone failed to antagonize the septal neuron population loss but prevented the progressive abnormal morphological changes of neurons. Those animals exposed to 192-IgG-SAP immunotoxin exhibited a reduction of cortical nNOS expression against the control group, whereas expression was enhanced in the 192-IgG-SAP + MOLS group. The most relevant finding was the recovering of the discrimination index exhibited by the 192-IgG-SAP + MOLS group. When compared with the rats exposed to the 192-IgG-SAP immunotoxin, they reached values similar to those observed in the PBS group. Our results show that although MOLS failed to block the cholinergic neurons loss induced by 192-IgG-SAP, it avoided the neuronal damage progression.
Collapse
Affiliation(s)
- M Alejandra Hernández-Melesio
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, Col. La Fama, 14269, Del. Tlalpan, Ciudad de México, Mexico; Departamento de Neuropsicofarmacología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Cda. México-Xochimilco 101, Col. Huipulco, C.P 14370, Del. Tlalpan, Ciudad de México, Mexico
| | - Mireya Alcaraz-Zubeldia
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, Col. La Fama, 14269, Del. Tlalpan, Ciudad de México, Mexico
| | - María E Jiménez-Capdeville
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Av. Venustiano Carranza # 2405, C.P. 78210, San Luis Potosí, S.L.P., Mexico
| | - Juan Carlos Martínez-Lazcano
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, Col. La Fama, 14269. Del. Tlalpan, Ciudad de México, Mexico
| | - Martha E Santoyo-Pérez
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, Col. La Fama, 14269. Del. Tlalpan, Ciudad de México, Mexico
| | - Lucía Quevedo-Corona
- Departamento de Fisiología "Mauricio Russek", Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Avenida Wilfrido Massieu esq. Cda. Miguel Stampa s/n, Col. San Pedro Zacatenco, C.P. 07738, Del. Gustavo A. Madero, Ciudad de México, Mexico
| | - Cristian Gerónimo-Olvera
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, Col. La Fama, 14269, Del. Tlalpan, Ciudad de México, Mexico
| | - Alicia Sánchez-Mendoza
- Departamento de Farmacología, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano #1, Col. Sección XVI, C.P. 14080, Del. Tlalpan, Ciudad de México, Mexico
| | - Camilo Ríos
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, Col. La Fama, 14269, Del. Tlalpan, Ciudad de México, Mexico
| | - Francisca Pérez-Severiano
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Insurgentes Sur #3877, Col. La Fama, 14269, Del. Tlalpan, Ciudad de México, Mexico.
| |
Collapse
|
24
|
More JY, Bruna BA, Lobos PE, Galaz JL, Figueroa PL, Namias S, Sánchez GL, Barrientos GC, Valdés JL, Paula-Lima AC, Hidalgo C, Adasme T. Calcium Release Mediated by Redox-Sensitive RyR2 Channels Has a Central Role in Hippocampal Structural Plasticity and Spatial Memory. Antioxid Redox Signal 2018; 29:1125-1146. [PMID: 29357673 DOI: 10.1089/ars.2017.7277] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS Previous studies indicate that hippocampal synaptic plasticity and spatial memory processes entail calcium release from intracellular stores mediated by ryanodine receptor (RyR) channels. In particular, RyR-mediated Ca2+ release is central for the dendritic spine remodeling induced by brain-derived neurotrophic factor (BDNF), a neurotrophin that stimulates complex signaling pathways leading to memory-associated protein synthesis and structural plasticity. To examine if upregulation of ryanodine receptor type-2 (RyR2) channels and the spine remodeling induced by BDNF entail reactive oxygen species (ROS) generation, and to test if RyR2 downregulation affects BDNF-induced spine remodeling and spatial memory. RESULTS Downregulation of RyR2 expression (short hairpin RNA [shRNA]) in primary hippocampal neurons, or inhibition of nitric oxide synthase (NOS) or NADPH oxidase, prevented agonist-mediated RyR-mediated Ca2+ release, whereas BDNF promoted cytoplasmic ROS generation. RyR2 downregulation or inhibitors of N-methyl-d-aspartate (NMDA) receptors, or NOS or of NADPH oxidase type-2 (NOX2) prevented RyR2 upregulation and the spine remodeling induced by BDNF, as did incubation with the antioxidant agent N-acetyl l-cysteine. In addition, intrahippocampal injection of RyR2-directed antisense oligodeoxynucleotides, which caused significant RyR2 downregulation, caused conspicuous defects in a memorized spatial memory task. INNOVATION The present novel results emphasize the key role of redox-sensitive Ca2+ release mediated by RyR2 channels for hippocampal structural plasticity and spatial memory. CONCLUSION Based on these combined results, we propose (i) that BDNF-induced RyR2-mediated Ca2+ release and ROS generation via NOS/NOX2 are strictly required for the dendritic spine remodeling and the RyR2 upregulation induced by BDNF, and (ii) that RyR2 channel expression is crucial for spatial memory processes. Antioxid. Redox Signal. 29, 1125-1146.
Collapse
Affiliation(s)
- Jamileth Y More
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Barbara A Bruna
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Pedro E Lobos
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - José L Galaz
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Paula L Figueroa
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Silvia Namias
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Gina L Sánchez
- 2 Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Genaro C Barrientos
- 2 Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - José L Valdés
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,3 Department of Neuroscience, Faculty of Medicine, Universidad de Chile , Santiago, Chile
| | - Andrea C Paula-Lima
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,4 Institute for Research in Dental Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,2 Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile .,3 Department of Neuroscience, Faculty of Medicine, Universidad de Chile , Santiago, Chile .,5 Center for Exercise , Metabolism and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Tatiana Adasme
- 1 Biomedical Neuroscience Institute , Faculty of Medicine, Universidad de Chile, Santiago, Chile .,6 Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O'Higgins , Santiago, Chile
| |
Collapse
|
25
|
Friebe A, Voußen B, Groneberg D. NO-GC in cells 'off the beaten track'. Nitric Oxide 2018; 77:12-18. [DOI: 10.1016/j.niox.2018.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/14/2018] [Accepted: 02/23/2018] [Indexed: 02/08/2023]
|
26
|
Christenson Wick Z, Krook-Magnuson E. Specificity, Versatility, and Continual Development: The Power of Optogenetics for Epilepsy Research. Front Cell Neurosci 2018; 12:151. [PMID: 29962936 PMCID: PMC6010559 DOI: 10.3389/fncel.2018.00151] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/15/2018] [Indexed: 12/19/2022] Open
Abstract
Optogenetics is a powerful and rapidly expanding set of techniques that use genetically encoded light sensitive proteins such as opsins. Through the selective expression of these exogenous light-sensitive proteins, researchers gain the ability to modulate neuronal activity, intracellular signaling pathways, or gene expression with spatial, directional, temporal, and cell-type specificity. Optogenetics provides a versatile toolbox and has significantly advanced a variety of neuroscience fields. In this review, using recent epilepsy research as a focal point, we highlight how the specificity, versatility, and continual development of new optogenetic related tools advances our understanding of neuronal circuits and neurological disorders. We additionally provide a brief overview of some currently available optogenetic tools including for the selective expression of opsins.
Collapse
Affiliation(s)
- Zoé Christenson Wick
- Graduate Program in Neuroscience and Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|
27
|
Dexras1 is a homeostatic regulator of exercise-dependent proliferation and cell survival in the hippocampal neurogenic niche. Sci Rep 2018; 8:5294. [PMID: 29593295 PMCID: PMC5871767 DOI: 10.1038/s41598-018-23673-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/19/2018] [Indexed: 02/07/2023] Open
Abstract
Adult hippocampal neurogenesis is highly responsive to exercise, which promotes the proliferation of neural progenitor cells and the integration of newborn granule neurons in the dentate gyrus. Here we show that genetic ablation of the small GTPase, Dexras1, suppresses exercise-induced proliferation of neural progenitors, alters survival of mitotic and post-mitotic cells in a stage-specific manner, and increases the number of mature newborn granule neurons. Dexras1 is required for exercise-triggered recruitment of quiescent neural progenitors into the cell cycle. Pharmacological inhibition of NMDA receptors enhances SGZ cell proliferation in wild-type but not dexras1-deficient mice, suggesting that NMDA receptor-mediated signaling is dependent on Dexras1. At the molecular level, the absence of Dexras1 abolishes exercise-dependent activation of ERK/MAPK and CREB, and inhibits the upregulation of NMDA receptor subunit NR2A, bdnf, trkB and vegf-a expression in the dentate gyrus. Our study reveals Dexras1 as an important stage-specific regulator of exercise-induced neurogenesis in the adult hippocampus by enhancing pro-mitogenic signaling to neural progenitor cells and modulating cell survival.
Collapse
|
28
|
Romano DR, Pharris MC, Patel NM, Kinzer-Ursem TL. Competitive tuning: Competition's role in setting the frequency-dependence of Ca2+-dependent proteins. PLoS Comput Biol 2017; 13:e1005820. [PMID: 29107982 PMCID: PMC5690689 DOI: 10.1371/journal.pcbi.1005820] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 11/16/2017] [Accepted: 10/13/2017] [Indexed: 01/20/2023] Open
Abstract
A number of neurological disorders arise from perturbations in biochemical signaling and protein complex formation within neurons. Normally, proteins form networks that when activated produce persistent changes in a synapse’s molecular composition. In hippocampal neurons, calcium ion (Ca2+) flux through N-methyl-D-aspartate (NMDA) receptors activates Ca2+/calmodulin signal transduction networks that either increase or decrease the strength of the neuronal synapse, phenomena known as long-term potentiation (LTP) or long-term depression (LTD), respectively. The calcium-sensor calmodulin (CaM) acts as a common activator of the networks responsible for both LTP and LTD. This is possible, in part, because CaM binding proteins are “tuned” to different Ca2+ flux signals by their unique binding and activation dynamics. Computational modeling is used to describe the binding and activation dynamics of Ca2+/CaM signal transduction and can be used to guide focused experimental studies. Although CaM binds over 100 proteins, practical limitations cause many models to include only one or two CaM-activated proteins. In this work, we view Ca2+/CaM as a limiting resource in the signal transduction pathway owing to its low abundance relative to its binding partners. With this view, we investigate the effect of competitive binding on the dynamics of CaM binding partner activation. Using an explicit model of Ca2+, CaM, and seven highly-expressed hippocampal CaM binding proteins, we find that competition for CaM binding serves as a tuning mechanism: the presence of competitors shifts and sharpens the Ca2+ frequency-dependence of CaM binding proteins. Notably, we find that simulated competition may be sufficient to recreate the in vivo frequency dependence of the CaM-dependent phosphatase calcineurin. Additionally, competition alone (without feedback mechanisms or spatial parameters) could replicate counter-intuitive experimental observations of decreased activation of Ca2+/CaM-dependent protein kinase II in knockout models of neurogranin. We conclude that competitive tuning could be an important dynamic process underlying synaptic plasticity. Learning and memory formation are likely associated with dynamic fluctuations in the connective strength of neuronal synapses. These fluctuations, called synaptic plasticity, are regulated by calcium ion (Ca2+) influx through ion channels localized to the post-synaptic membrane. Within the post-synapse, the dominant Ca2+ sensor protein, calmodulin (CaM), may activate a variety of downstream binding partners, each contributing to synaptic plasticity outcomes. The conditions at which certain binding partners most strongly activate are increasingly studied using computational models. Nearly all computational studies describe these binding partners in combinations of only one or two CaM binding proteins. In contrast, we combine seven well-studied CaM binding partners into a single model wherein they simultaneously compete for access to CaM. Our dynamic model suggests that competition narrows the window of conditions for optimal activation of some binding partners, mimicking the Ca2+-frequency dependence of some proteins in vivo. Further characterization of CaM-dependent signaling dynamics in neuronal synapses may benefit our understanding of learning and memory formation. Furthermore, we propose that competitive binding may be another framework, alongside feedback and feed-forward loops, signaling motifs, and spatial localization, that can be applied to other signal transduction networks, particularly second messenger cascades, to explain the dynamical behavior of protein activation.
Collapse
Affiliation(s)
- Daniel R. Romano
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| | - Matthew C. Pharris
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| | - Neal M. Patel
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| | - Tamara L. Kinzer-Ursem
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
- * E-mail:
| |
Collapse
|
29
|
Wang Q, Mergia E, Koesling D, Mittmann T. Nitric oxide/cGMP signaling via guanylyl cyclase isoform 1 modulates glutamate and GABA release in somatosensory cortex of mice. Neuroscience 2017; 360:180-189. [PMID: 28782641 DOI: 10.1016/j.neuroscience.2017.07.063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 11/24/2022]
Abstract
In hippocampus, two guanylyl cyclases (NO-GC1 and NO-GC2) are involved in the transduction of the effects of nitric oxide (NO) on synaptic transmission. However, the respective roles of the NO-GC isoforms on synaptic transmission are less clear in other regions of the brain. In the present study, we used knock-out mice deficient for the NO-GC1 isoform (NO-GC1 KO) to analyze its role in the glutamatergic and GABAergic neurotransmission at pyramidal neurons in layers II/III of somatosensory cortex. NO-GC1 KO slices revealed reduced frequencies of miniature excitatory- and inhibitory-postsynaptic currents, increased paired-pulse ratios and decreased input-output curves of evoked signals, which indicated the reduction of glutamate and GABA release in NO-GC1 KO mice. The functional changes in NO-GC1 KO mice were caused by the lack of cGMP as they were rescued to WT-like levels by the cGMP analog, 8-Br-PET-cGMP and conversely, mimicked by the NO-GC inhibitor, ODQ, in WT slices. In search of a cGMP target, two blockers of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels (ZD7288 and DK-AH269) reduced glutamate release in WT to the level of NO-GC1 KO mice suggesting HCN channels as possible effectors for presynaptic cGMP enhancing the glutamate release probability. By blocking postsynaptic NMDA receptors, the NMDA receptor-dependent NO signal was shown to be linked to the effect of NO-GC1 on presynaptic GABA release. Of note, the balance between glutamatergic and GABAergic inputs at individual synapses remained unaltered in the NO-GC1 KO mice. In sum, our results indicate a role for cGMP generated by presynaptic localized NO-GC1 to adjust inhibitory and excitatory inputs at individual synapses in the somatosensory cortex.
Collapse
Affiliation(s)
- Qi Wang
- Institute of Physiology, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany
| | - Evanthia Mergia
- Institute of Pharmacology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Doris Koesling
- Institute of Pharmacology, Ruhr-University Bochum, 44780 Bochum, Germany
| | - Thomas Mittmann
- Institute of Physiology, UMC of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| |
Collapse
|
30
|
Rodriguez-Grande B, Konsman JP. Gas Diffusion in the CNS. J Neurosci Res 2017; 96:207-218. [PMID: 28504343 DOI: 10.1002/jnr.24077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/23/2017] [Accepted: 04/10/2017] [Indexed: 12/21/2022]
Abstract
Gases have been long known to have essential physiological functions in the CNS such as respiration or regulation of vascular tone. Since gases have been classically considered to freely diffuse, research in gas biology has so far focused on mechanisms of gas synthesis and gas reactivity, rather than gas diffusion and transport. However, the discovery of gas pores during the last two decades and the characterization of diverse diffusion patterns through different membranes has raised the possibility that modulation of gas diffusion is also a physiologically relevant parameter. Here we review the means of gas movement into and within the brain through "free" diffusion and gas pores, notably aquaporins, discussing the role that gas diffusion may play in the modulation of gas function. We highlight how diffusion is relevant to neuronal signaling, volume transmission, and cerebrovascular control in the case of NO, one of the most extensively studied gases. We point out how facilitated transport can be especially relevant for gases with low permeability in lipid membranes like NH3 and discuss the possible implications of NH3 -permeable channels in physiology and hyperammonemic encephalopathy. We identify novel research questions about how modulation of gas diffusion could intervene in CNS pathologies. This emerging area of research can provide novel and interesting insights in the field of gas biology.
Collapse
|
31
|
Participation of hippocampal nitric oxide synthase and soluble guanylate cyclase in the modulation of behavioral responses elicited by the rat forced swimming test. Behav Pharmacol 2017; 28:19-29. [PMID: 27779493 DOI: 10.1097/fbp.0000000000000263] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
32
|
Pigott BM, Garthwaite J. Nitric Oxide Is Required for L-Type Ca(2+) Channel-Dependent Long-Term Potentiation in the Hippocampus. Front Synaptic Neurosci 2016; 8:17. [PMID: 27445786 PMCID: PMC4925670 DOI: 10.3389/fnsyn.2016.00017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/13/2016] [Indexed: 12/11/2022] Open
Abstract
Nitric oxide (NO) has long been implicated in the generation of long-term potentiation (LTP) and other types of synaptic plasticity, a role for which the intimate coupling between NMDA receptors (NMDARs) and the neuronal isoform of NO synthase (nNOS) is likely to be instrumental in many instances. While several types of synaptic plasticity depend on NMDARs, others do not, an example of which is LTP triggered by opening of L-type voltage-gated Ca2+ channels (L-VGCCs) in postsynaptic neurons. In CA3-CA1 synapses in the hippocampus, NMDAR-dependent LTP (LTPNMDAR) appears to be primarily expressed postsynaptically whereas L-VGCC-dependent LTP (LTPL−VGCC), which often coexists with LTPNMDAR, appears mainly to reflect enhanced presynaptic transmitter release. Since NO is an excellent candidate as a retrograde messenger mediating post-to-presynaptic signaling, we sought to determine if NO functions in LTPL−VGCC in mouse CA3-CA1 synapses. When elicited by a burst type of stimulation with NMDARs and the associated NO release blocked, LTPL−VGCC was curtailed by inhibition of NO synthase or of the NO-receptor guanylyl cyclase to the same extent as occurred with inhibition of L-VGCCs. Unlike LTPNMDAR at these synapses, LTPL−VGCC was unaffected in mice lacking endothelial NO synthase, implying that the major source of the NO is neuronal. Transient delivery of exogenous NO paired with tetanic synaptic stimulation under conditions of NMDAR blockade resulted in a long-lasting potentiation that was sensitive to inhibition of NO-receptor guanylyl cyclase but was unaffected by inhibition of L-VGCCs. The results indicate that NO, acting through its second messenger cGMP, plays an unexpectedly important role in L-VGCC-dependent, NMDAR-independent LTP, possibly as a retrograde messenger generated in response to opening of postsynaptic L-VGCCs and/or as a signal acting postsynaptically, perhaps to facilitate changes in gene expression.
Collapse
Affiliation(s)
- Beatrice M Pigott
- The Wolfson Institute for Biomedical Research, University College London London, UK
| | - John Garthwaite
- The Wolfson Institute for Biomedical Research, University College London London, UK
| |
Collapse
|
33
|
Sweeney Y, Clopath C. Emergent spatial synaptic structure from diffusive plasticity. Eur J Neurosci 2016; 45:1057-1067. [DOI: 10.1111/ejn.13279] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/04/2016] [Accepted: 05/13/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Yann Sweeney
- Department of Bioengineering; Imperial College London, South Kensington Campus; London SW7 2AZ UK
| | - Claudia Clopath
- Department of Bioengineering; Imperial College London, South Kensington Campus; London SW7 2AZ UK
| |
Collapse
|
34
|
Garthwaite J. From synaptically localized to volume transmission by nitric oxide. J Physiol 2015; 594:9-18. [PMID: 26486504 DOI: 10.1113/jp270297] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/13/2015] [Indexed: 12/22/2022] Open
Abstract
Nitric oxide (NO) functions widely as a transmitter/diffusible second messenger in the central nervous system, exerting physiological effects in target cells by binding to specialized guanylyl cyclase-coupled receptors, resulting in cGMP generation. Despite having many context-dependent physiological roles and being implicated in numerous disease states, there has been a lack of clarity about the ways that NO operates at the cellular and subcellular levels. Recently, several approaches have been used to try to gain a more concrete, quantitative understanding of this unique signalling pathway. These approaches have included analysing the kinetics of NO receptor function, real-time imaging of cellular NO signal transduction in target cells, and the use of ultrasensitive detector cells to record NO as it is being generated from native sources in brain tissue. The current picture is that, when formed in a synapse, NO is likely to act only very locally, probably mostly within the confines of that synapse, and to exist only in picomolar concentrations. Nevertheless, closely neighbouring synapses may also be within reach, raising the possibility of synaptic crosstalk. By engaging its enzyme-coupled receptors, the low NO concentrations are able to stimulate physiological (submicromolar) increases in cGMP concentration in an activity-dependent manner. When many NO-emitting neurones or synapses are active simultaneously in a tissue region, NO can act more like a volume transmitter to influence, and perhaps coordinate, the behaviour of cells within that region, irrespective of their identity and anatomical connectivity.
Collapse
Affiliation(s)
- John Garthwaite
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
35
|
Carletti F, Gambino G, Rizzo V, Ferraro G, Sardo P. Cannabinoid and nitric oxide signaling interplay in the modulation of hippocampal hyperexcitability: Study on electrophysiological and behavioral models of temporal lobe epilepsy in the rat. Neuroscience 2015; 303:149-59. [DOI: 10.1016/j.neuroscience.2015.06.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/10/2015] [Accepted: 06/23/2015] [Indexed: 02/04/2023]
|
36
|
Burette A, Collman F, Micheva KD, Smith SJ, Weinberg RJ. Knowing a synapse when you see one. Front Neuroanat 2015; 9:100. [PMID: 26283929 PMCID: PMC4517447 DOI: 10.3389/fnana.2015.00100] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/10/2015] [Indexed: 11/22/2022] Open
Abstract
Recent years have seen a rapidly growing recognition of the complexity and diversity of the myriad individual synaptic connections that define brain synaptic networks. It has also become increasingly apparent that the synapses themselves are a major key to understanding the development, function and adaptability of those synaptic networks. In spite of this growing appreciation, the molecular, structural and functional characteristics of individual synapses and the patterning of their diverse characteristics across functional networks have largely eluded quantitative study with available imaging technologies. Here we offer an overview of new computational imaging methods that promise to bring single-synapse analysis of synaptic networks to the fore. We focus especially on the challenges and opportunities associated with quantitative detection of individual synapses and with measuring individual synapses across network scale populations in mammalian brain.
Collapse
Affiliation(s)
- Alain Burette
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| | | | - Kristina D Micheva
- Department of Molecular and Cellular Physiology, Stanford University Stanford, CA, USA
| | | | - Richard J Weinberg
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| |
Collapse
|
37
|
Gasulla J, Calvo DJ. Enhancement of tonic and phasic GABAergic currents following nitric oxide synthase inhibition in hippocampal CA1 pyramidal neurons. Neurosci Lett 2015; 590:29-34. [PMID: 25636692 DOI: 10.1016/j.neulet.2015.01.058] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/21/2015] [Accepted: 01/22/2015] [Indexed: 01/23/2023]
Abstract
Nitric oxide (NO) is involved in synaptic plasticity in the hippocampus through different presynaptic and postsynaptic mechanisms that include the modulation of the GABAergic neurotransmission. Inhibitory synapses on hippocampal pyramidal neurons are known to possess the molecular machinery for retrograde NO-signaling, but the modulation of GABAARs function by NO in these neurons and the mechanisms of action involved have not been fully characterized. Here we show that suppression of the endogenous NO generation by the nitric oxide synthase (NOS) inhibitor L-NAME produces significant and reversible increases in the magnitude of both tonic and phasic GABAergic currents in CA1 hippocampal pyramidal neurons. GABA-evoked chloride currents were measured in the presence or absence of L-NAME using whole-cell patch-clamp recordings in acute hippocampal slices from young adult mice. Enhancement of the tonic GABA responses induced by L-NAME was insensitive to TTX and decreased by co-incubation with the NO donor DEA/NO. Applications of DEA/NO alone did not produce significant effects on tonic GABA responses. L-NAME treatment also increased the amplitude of phasic GABAergic currents evoked by GABA-puffs. Our results indicate that the extent of tonic and phasic inhibition mediated by GABAA receptors in CA1 hippocampal pyramidal neurons is affected by endogenous NO production.
Collapse
Affiliation(s)
- Javier Gasulla
- Laboratorio de Neurobiología Celular y Molecular Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI) "Dr. Héctor N. Torres", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Vuelta de Obligado 2490, Ciudad Autónoma de Buenos Aires (CP 1428), Argentina
| | - Daniel J Calvo
- Laboratorio de Neurobiología Celular y Molecular Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI) "Dr. Héctor N. Torres", Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Vuelta de Obligado 2490, Ciudad Autónoma de Buenos Aires (CP 1428), Argentina.
| |
Collapse
|
38
|
Kolarow R, Kuhlmann CRW, Munsch T, Zehendner C, Brigadski T, Luhmann HJ, Lessmann V. BDNF-induced nitric oxide signals in cultured rat hippocampal neurons: time course, mechanism of generation, and effect on neurotrophin secretion. Front Cell Neurosci 2014; 8:323. [PMID: 25426021 PMCID: PMC4224130 DOI: 10.3389/fncel.2014.00323] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 09/26/2014] [Indexed: 11/13/2022] Open
Abstract
BDNF and nitric oxide signaling both contribute to plasticity at glutamatergic synapses. However, the role of combined signaling of both pathways at the same synapse is largely unknown. Using NO imaging with diaminofluoresceine in cultured hippocampal neurons we analyzed the time course of neurotrophin-induced NO signals. Application of exogenous BDNF, NT-4, and NT-3 (but not NGF) induced NO signals in the soma and in proximal dendrites of hippocampal neurons that were sensitive to NO synthase activity, TrkB signaling, and intracellular calcium elevation. The effect of NO signaling on neurotrophin secretion was analyzed in BDNF-GFP, and NT-3-GFP transfected hippocampal neurons. Exogenous application of the NO donor sodium-nitroprusside markedly inhibited neurotrophin secretion. However, endogenously generated NO in response to depolarization and neurotrophin stimulation, both did not result in a negative feedback on neurotrophin secretion. These results suggest that a negative feedback of NO signaling on synaptic secretion of neurotrophins operates only at high intracellular levels of nitric oxide that are under physiological conditions not reached by depolarization or BDNF signaling.
Collapse
Affiliation(s)
- Richard Kolarow
- Medical Faculty, Institute of Physiology, Otto-von-Guericke-University Magdeburg, Germany ; University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Christoph R W Kuhlmann
- University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Thomas Munsch
- Medical Faculty, Institute of Physiology, Otto-von-Guericke-University Magdeburg, Germany
| | - Christoph Zehendner
- University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Tanja Brigadski
- Medical Faculty, Institute of Physiology, Otto-von-Guericke-University Magdeburg, Germany ; University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Heiko J Luhmann
- University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Volkmar Lessmann
- Medical Faculty, Institute of Physiology, Otto-von-Guericke-University Magdeburg, Germany ; University Medical Center, Institute of Physiology, Johannes Gutenberg-University Mainz Mainz, Germany
| |
Collapse
|
39
|
Bernardinelli Y, Nikonenko I, Muller D. Structural plasticity: mechanisms and contribution to developmental psychiatric disorders. Front Neuroanat 2014; 8:123. [PMID: 25404897 PMCID: PMC4217507 DOI: 10.3389/fnana.2014.00123] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/14/2014] [Indexed: 01/31/2023] Open
Abstract
Synaptic plasticity mechanisms are usually discussed in terms of changes in synaptic strength. The capacity of excitatory synapses to rapidly modify the membrane expression of glutamate receptors in an activity-dependent manner plays a critical role in learning and memory processes by re-distributing activity within neuronal networks. Recent work has however also shown that functional plasticity properties are associated with a rewiring of synaptic connections and a selective stabilization of activated synapses. These structural aspects of plasticity have the potential to continuously modify the organization of synaptic networks and thereby introduce specificity in the wiring diagram of cortical circuits. Recent work has started to unravel some of the molecular mechanisms that underlie these properties of structural plasticity, highlighting an important role of signaling pathways that are also major candidates for contributing to developmental psychiatric disorders. We review here some of these recent advances and discuss the hypothesis that alterations of structural plasticity could represent a common mechanism contributing to the cognitive and functional defects observed in diseases such as intellectual disability, autism spectrum disorders and schizophrenia.
Collapse
Affiliation(s)
- Yann Bernardinelli
- Department of Basic Neurosciences, University of Geneva Medical School Geneva, Switzerland
| | - Irina Nikonenko
- Department of Basic Neurosciences, University of Geneva Medical School Geneva, Switzerland
| | - Dominique Muller
- Department of Basic Neurosciences, University of Geneva Medical School Geneva, Switzerland
| |
Collapse
|
40
|
Rizzo V, Carletti F, Gambino G, Schiera G, Cannizzaro C, Ferraro G, Sardo P. Role of CB2 receptors and cGMP pathway on the cannabinoid-dependent antiepileptic effects in an in vivo model of partial epilepsy. Epilepsy Res 2014; 108:1711-8. [PMID: 25458534 DOI: 10.1016/j.eplepsyres.2014.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/12/2014] [Accepted: 10/01/2014] [Indexed: 10/24/2022]
Abstract
This study aimed at providing an insight on the possible role of cannabinoid (CB) type 2 receptors (CB2R) and cGMP pathway in the antiepileptic activity of WIN 55,212-2, (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl) pyrrolo[1,2,3-de]-1,4-benzoxazin-6-Yl]-1-naphthalenylmethanone, a non-selective CB agonist, in the maximal dentate activation (MDA) model of partial epilepsy in adult male rats. We evaluated the activity of a CB2 antagonist/inverse agonist AM630, [6-iodo-2-methyl-1-[2-(4-morpholinyl)ethyl]-1H-indol-3-yl](4-methoxyphenyl)methanone or 6-iodopravadoline, alone or in co-administration with WIN 55,212-2. Also, in the MDA model it was investigated the co-treatment of WIN 55,212-2 and 1H-[1,2,4]Oxadiazole[4,3-a]quinoxalin-1-one (ODQ), a specific inhibitor of the nitric oxide (NO)-activated soluble guanylyl cyclase (sGC), the cGMP producing enzyme. The WIN 55,212-2-dependent (21mg/kg) antiepileptic effects were significantly increased by the co-administration with AM630 and by the co-treatment with ODQ (10mg/kg). Whereas, the administration of AM630 (2mg/kg), alone exerts no effects on hippocampal hyperexcitability. Our data show that pharmacological blockade of CB2 receptors and of sGC seems to cooperate with WIN in its antiepileptic action. These findings shed light on CB signaling mechanisms, hinting that the modulation of the effects of CB agonist in the hyperexcitability phenomena may be exerted both by targeting CB receptors and their possible downstream effectors, such as nitrergic-dependent cGMP pathway.
Collapse
Affiliation(s)
- Valerio Rizzo
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (Bio.Ne.C.), Sezione di Fisiologia umana "G. Pagano", Università degli Studi di Palermo, Corso Tukory, 129-90134 Palermo, Italy; Department of Neuroscience, The Scripps Research Institute, Scripps Florida 130 Scripps Way, Jupiter, FL 33458.
| | - Fabio Carletti
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (Bio.Ne.C.), Sezione di Fisiologia umana "G. Pagano", Università degli Studi di Palermo, Corso Tukory, 129-90134 Palermo, Italy
| | - Giuditta Gambino
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (Bio.Ne.C.), Sezione di Fisiologia umana "G. Pagano", Università degli Studi di Palermo, Corso Tukory, 129-90134 Palermo, Italy
| | - Girolamo Schiera
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (Bio.Ne.C.), Sezione di Fisiologia umana "G. Pagano", Università degli Studi di Palermo, Corso Tukory, 129-90134 Palermo, Italy
| | - Carla Cannizzaro
- Dipartimento di Scienze per la Promozione della salute, Università degli Studi di Palermo, Via del Vespro, 133, 90100 Palermo, Italy
| | - Giuseppe Ferraro
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (Bio.Ne.C.), Sezione di Fisiologia umana "G. Pagano", Università degli Studi di Palermo, Corso Tukory, 129-90134 Palermo, Italy
| | - Pierangelo Sardo
- Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche (Bio.Ne.C.), Sezione di Fisiologia umana "G. Pagano", Università degli Studi di Palermo, Corso Tukory, 129-90134 Palermo, Italy
| |
Collapse
|
41
|
Colvin SM, Kwan KY. Dysregulated nitric oxide signaling as a candidate mechanism of fragile X syndrome and other neuropsychiatric disorders. Front Genet 2014; 5:239. [PMID: 25101118 PMCID: PMC4105824 DOI: 10.3389/fgene.2014.00239] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/03/2014] [Indexed: 12/31/2022] Open
Abstract
A mechanistic understanding of the pathophysiology underpinning psychiatric disorders is essential for the development of targeted molecular therapies. For fragile X syndrome (FXS), recent mechanistic studies have been focused on the metabotropic glutamate receptor (mGluR) signaling pathway. This line of research has led to the discovery of promising candidate drugs currently undergoing various phases of clinical trial, and represents a model of how biological insights can inform therapeutic strategies in neurodevelopmental disorders. Although mGluR signaling is a key mechanism at which targeted treatments can be directed, it is likely to be one of many mechanisms contributing to FXS. A more complete understanding of the molecular and neural underpinnings of the disorder is expected to inform additional therapeutic strategies. Alterations in the assembly of neural circuits in the neocortex have been recently implicated in genetic studies of autism and schizophrenia, and may also contribute to FXS. In this review, we explore dysregulated nitric oxide signaling in the developing neocortex as a novel candidate mechanism of FXS. This possibility stems from our previous work demonstrating that neuronal nitric oxide synthase 1 (NOS1 or nNOS) is regulated by the FXS protein FMRP in the mid-fetal human neocortex. Remarkably, in the mid-late fetal and early postnatal neocortex of human FXS patients, NOS1 expression is severely diminished. Given the role of nitric oxide in diverse neural processes, including synaptic development and plasticity, the loss of NOS1 in FXS may contribute to the etiology of the disorder. Here, we outline the genetic and neurobiological data that implicate neocortical dysfunction in FXS, review the evidence supporting dysregulated nitric oxide signaling in the developing FXS neocortex and its contribution to the disorder, and discuss the implications for targeting nitric oxide signaling in the treatment of FXS and other psychiatric illnesses.
Collapse
Affiliation(s)
- Steven M Colvin
- Department of Human Genetics - The Molecular and Behavioral Neuroscience Institute, University of Michigan Medical School Ann Arbor, MI, USA
| | - Kenneth Y Kwan
- Department of Human Genetics - The Molecular and Behavioral Neuroscience Institute, University of Michigan Medical School Ann Arbor, MI, USA
| |
Collapse
|
42
|
NO regulates the strength of synaptic inputs onto hippocampal CA1 neurons via NO-GC1/cGMP signalling. Pflugers Arch 2014; 467:1383-94. [PMID: 25010738 DOI: 10.1007/s00424-014-1571-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 06/21/2014] [Accepted: 06/30/2014] [Indexed: 12/11/2022]
Abstract
GABAergic interneurons are the predominant source of inhibition in the brain that coordinate the level of excitation and synchronization in neuronal circuitries. However, the underlying cellular mechanisms are still not fully understood. Here we report nitric oxide (NO)/NO-GC1 signalling as an important regulatory mechanism of GABAergic and glutamatergic synaptic transmission in the hippocampal CA1 region. Deletion of the NO receptor NO-GC1 induced functional alterations, indicated by a strong reduction of spontaneous and evoked inhibitory postsynaptic currents (IPSCs), which could be compensated by application of the missing second messenger cGMP. Moreover, we found a general impairment in the strength of inhibitory and excitatory synaptic inputs onto CA1 pyramidal neurons deriving from NO-GC1KO mice. Finally, we disclosed one subpopulation of GABAergic interneurons, fast-spiking interneurons, that receive less excitatory synaptic input and consequently respond with less spike output after blockage of the NO/cGMP signalling pathway. On the basis of these and previous findings, we propose NO-GC1 as the major NO receptor which transduces the NO signal into cGMP at presynaptic terminals of different neuronal subtypes in the hippocampal CA1 region. Furthermore, we suggest NO-GC1-mediated cGMP signalling as a mechanism which regulates the strength of synaptic transmission, hence being important in gating information processing between hippocampal CA3 and CA1 region.
Collapse
|
43
|
Lies B, Groneberg D, Friebe A. Toward a better understanding of gastrointestinal nitrergic neuromuscular transmission. Neurogastroenterol Motil 2014; 26:901-12. [PMID: 24827638 DOI: 10.1111/nmo.12367] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/21/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND Nitric oxide (NO) is an important inhibitory neurotransmitter in the gastrointestinal (GI) tract. The majority of nitrergic effects are transduced by NO-sensitive guanylyl cyclase (NO-GC) as the receptor for NO, and, thus, mediated by cGMP-dependent mechanisms. Work carried out during the past years has demonstrated NO to be largely involved in GI smooth muscle relaxation and motility. However, detailed investigation of nitrergic signaling has turned out to be complicated as NO-GC was identified in several different GI cell types such as smooth muscle cells, interstitial cells of Cajal and fibroblast-like cells. With regards to nitrergic neurotransmission, special focus has been placed on the role of interstitial cells of Cajal using mutant mice with reduced populations of ICC. Recently, global and cell-specific knockout mice for enzymes participating in nitrergic signaling have been generated providing a suitable approach to further examine the role of NO-mediated signaling in GI smooth muscle. PURPOSE This review discusses the current knowledge on nitrergic mechanisms in gastrointestinal neuromuscular transmission with a focus on genetic models and outlines possible further investigations to gain better understanding on NO-mediated effects in the GI tract.
Collapse
Affiliation(s)
- B Lies
- Physiologisches Institut I, Universität Würzburg, Würzburg, Germany
| | | | | |
Collapse
|
44
|
Disney AA, Reynolds JH. Expression of m1-type muscarinic acetylcholine receptors by parvalbumin-immunoreactive neurons in the primary visual cortex: a comparative study of rat, guinea pig, ferret, macaque, and human. J Comp Neurol 2014; 522:986-1003. [PMID: 23983014 PMCID: PMC3945972 DOI: 10.1002/cne.23456] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 08/05/2013] [Accepted: 08/06/2013] [Indexed: 11/10/2022]
Abstract
Cholinergic neuromodulation is a candidate mechanism for aspects of arousal and attention in mammals. We have reported previously that cholinergic modulation in the primary visual cortex (V1) of the macaque monkey is strongly targeted toward GABAergic interneurons, and in particular that the vast majority of parvalbumin-immunoreactive (PV) neurons in macaque V1 express the m1-type (pirenzepine-sensitive, Gq-coupled) muscarinic ACh receptor (m1AChR). In contrast, previous physiological data indicates that PV neurons in rats rarely express pirenzepine-sensitive muscarinic AChRs. To examine further this apparent species difference in the cholinergic effectors for the primary visual cortex, we have conducted a comparative study of the expression of m1AChRs by PV neurons in V1 of rats, guinea pigs, ferrets, macaques, and humans. We visualize PV- and mAChR-immunoreactive somata by dual-immunofluorescence confocal microscopy and find that the species differences are profound; the vast majority (>75%) of PV-ir neurons in macaques, humans, and guinea pigs express m1AChRs. In contrast, in rats only ∼25% of the PV population is immunoreactive for m1AChRs. Our data reveal that while they do so much less frequently than in primates, PV neurons in rats do express Gq-coupled muscarinic AChRs, which appear to have gone undetected in the previous in vitro studies. Data such as these are critical in determining the species that represent adequate models for the capacity of the cholinergic system to modulate inhibition in the primate cortex.
Collapse
Affiliation(s)
- Anita A Disney
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, California, 92037
| | | |
Collapse
|
45
|
Jafari-Sabet M, Khodadadnejad MA, Ghoraba S, Ataee R. Nitric oxide in the dorsal hippocampal area is involved on muscimol state-dependent memory in the step-down passive avoidance test. Pharmacol Biochem Behav 2014; 117:137-43. [DOI: 10.1016/j.pbb.2013.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 09/06/2013] [Accepted: 12/06/2013] [Indexed: 10/25/2022]
|
46
|
Lourenço CF, Ferreira NR, Santos RM, Lukacova N, Barbosa RM, Laranjinha J. The pattern of glutamate-induced nitric oxide dynamics in vivo and its correlation with nNOS expression in rat hippocampus, cerebral cortex and striatum. Brain Res 2014; 1554:1-11. [PMID: 24495843 DOI: 10.1016/j.brainres.2014.01.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 01/13/2014] [Accepted: 01/18/2014] [Indexed: 01/20/2023]
Abstract
Nitric oxide (NO) is a diffusible intercellular messenger, acting via volume signaling in the brain and, therefore, the knowledge of its temporal dynamics is determinant to the understanding of its neurobiological role. However, such an analysis in vivo is challenging and indirect or static approaches are mostly used to infer NO bioactivity. In the present work we measured the glutamate-dependent NO temporal dynamics in vivo in the hippocampus (CA1, CA3 and DG subregions), cerebral cortex and striatum, using NO selective microelectrodes. Concurrently, the immunolocalization of nNOS was evaluated in each region. A transitory increase in NO levels occurred at higher amplitudes in the striatum and hippocampus relatively to the cortex. In the hippocampus, subtle differences in the profiles of NO signals were observed along the trisynaptic loop, with CA1 exhibiting the largest signals. The topography of NO temporal dynamics did not fully overlap with the pattern of the density of nNOS expression, suggesting that, complementary to the distribution of nNOS, the local regulation of NO synthesis as well as the decay pathways critically determine the effective NO concentration sensed by a target within the diffusional spread of this free radical. In sum, the rate and pattern of NO changes here shown, by incorporating regulatory mechanisms and processes that affect NO synthesis and decay, provide refined information critical for the understanding of NO multiple actions in the brain.
Collapse
Affiliation(s)
- C F Lourenço
- Faculty of Pharmacy and Center for Neurosciences and Cell Biology, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - N R Ferreira
- Faculty of Pharmacy and Center for Neurosciences and Cell Biology, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - R M Santos
- Faculty of Pharmacy and Center for Neurosciences and Cell Biology, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - N Lukacova
- Institute of Neurobiology, Slovak Academy of Sciences, Soltésovej 4, 040 01 Kosice, Slovak Republic
| | - R M Barbosa
- Faculty of Pharmacy and Center for Neurosciences and Cell Biology, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - J Laranjinha
- Faculty of Pharmacy and Center for Neurosciences and Cell Biology, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
47
|
Muñoz FJ, Godoy JA, Cerpa W, Poblete IM, Huidobro-Toro JP, Inestrosa NC. Wnt-5a increases NO and modulates NMDA receptor in rat hippocampal neurons. Biochem Biophys Res Commun 2014; 444:189-94. [PMID: 24440698 DOI: 10.1016/j.bbrc.2014.01.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/10/2014] [Indexed: 10/25/2022]
Abstract
Wnt signaling has a crucial role in synaptic function at the central nervous system. Here we evaluate whether Wnts affect nitric oxide (NO) generation in hippocampal neurons. We found that non-canonical Wnt-5a triggers NO production; however, Wnt-3a a canonical ligand did not exert the same effect. Co-administration of Wnt-5a with the soluble Frizzled related protein-2 (sFRP-2) a Wnt antagonist blocked the NO production. Wnt-5a activates the non-canonical Wnt/Ca(2+) signaling through a mechanism that depends on Ca(2+) release from Ryanodine-sensitive internal stores. The increase in NO levels evoked by Wnt-5a promotes the insertion of the GluN2B subunit of the NMDA receptor (NMDAR) into the neuronal cell surface. To the best of our knowledge, this is the first time that Wnt-5a signaling is related to NO production, which in turn increases NMDARs trafficking to the cell surface.
Collapse
Affiliation(s)
- Francisco J Muñoz
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago de Chile, Chile; Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Parc de Recerca Biomédica de Barcelona, Spain
| | - Juan A Godoy
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago de Chile, Chile
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago de Chile, Chile
| | - Inés M Poblete
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago de Chile, Chile; Departamento de Fisiología, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago de Chile, Chile
| | - Juan Pablo Huidobro-Toro
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago de Chile, Chile; Departamento de Fisiología, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago de Chile, Chile; Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago, Santiago de Chile, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago de Chile, Chile; Centre for Healthy Brain Ageing, School of Psychiatry, UNSW, Faculty of Medicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|
48
|
Nitric oxide mediates local activity-dependent excitatory synapse development. Proc Natl Acad Sci U S A 2013; 110:E4142-51. [PMID: 24127602 DOI: 10.1073/pnas.1311927110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Learning related paradigms play an important role in shaping the development and specificity of synaptic networks, notably by regulating mechanisms of spine growth and pruning. The molecular events underlying these synaptic rearrangements remain poorly understood. Here we identify NO signaling as a key mediator of activity-dependent excitatory synapse development. We find that chronic blockade of NO production in vitro and in vivo interferes with the development of hippocampal and cortical excitatory spine synapses. The effect results from a selective loss of activity-mediated spine growth mechanisms and is associated with morphological and functional alterations of remaining synapses. These effects of NO are mediated by a cGMP cascade and can be reproduced or prevented by postsynaptic expression of vasodilator-stimulated phosphoprotein phospho-mimetic or phospho-resistant mutants. In vivo analyses show that absence of NO prevents the increase in excitatory synapse density induced by environmental enrichment and interferes with the formation of local clusters of excitatory synapses. We conclude that NO plays an important role in regulating the development of excitatory synapses by promoting local activity-dependent spine-growth mechanisms.
Collapse
|
49
|
Rocha-de-Melo AP, Picanço-Diniz CW, Borba JMC, Santos-Monteiro J, Guedes RCA. NADPH-diaphorase Histochemical Labeling Patterns in the Hippocampal Neuropil and Visual Cortical Neurons in Weaned Rats Reared during Lactation on Different Litter Sizes. Nutr Neurosci 2013; 7:207-16. [PMID: 15682647 DOI: 10.1080/10284150400001961] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Tissue distribution of nitric oxide-synthases was investigated in the rat hippocampus and visual cortex under nutritional changes induced by modification of the litter size. Young (30-45-days-old) rats, suckled in litters formed by 3,6 or 12 pups (called small, medium and large litters, respectively), were studied by using nicotine-adenine-dinucleotide phosphate-diaphorase histochemistry (shortly, diaphorase), a simple and robust procedure to characterize tissue distribution of nitric oxide-synthases. We assessed morphometric features of the diaphorase-positive cells in visual cortex, and the neuropil histochemical activity in hippocampal CA1 and dentate gyrus using densitometry analysis. In the large-litter group, the labeled-cell density in white matter of area 17 was higher, as compared to the small-litter group. There was a clear trend, in the large-litter group, to lower values of soma area, dendritic field and branches per neuron, but the differences were not significant. Densitometry analysis of hippocampus revealed a significant increase in the relative neuropil histochemical activity of the dentate gyrus molecular layer in the larger litters, which may be associated to increased compensatory blood flow in the hippocampus. The pathophysiological mechanisms of the observed changes remain to be investigated.
Collapse
Affiliation(s)
- Ana Paula Rocha-de-Melo
- Departamento de Nutrição, Universidade Federal de Pernambuco, BR-50670-901, Recife, PE, Brazil
| | | | | | | | | |
Collapse
|
50
|
Abstract
In the hippocampus, as in many other CNS areas, nitric oxide (NO) participates in synaptic plasticity, manifested as changes in pre- and/or postsynaptic function. While it is known that these changes are brought about by cGMP following activation of guanylyl cyclase-coupled NO receptors attempts to locate cGMP by immunocytochemistry in hippocampal slices in response to NO have failed to detect the cGMP elevation where expected, i.e. in the pyramidal neurones. Instead, astrocytes, unidentified varicose fibres and GABA-ergic nerve terminals are reported to be the prominent NO targets, raising the possibility that NO acts indirectly via other cells. We have re-investigated the distribution of cGMP generated in response to endogenous and exogenous NO in hippocampal slices using immunohistochemistry and new conditions designed to optimise cGMP accumulation and, hence, its detectability. The conditions included use of tissue from the developing rat hippocampus, a potent inhibitor of phosphodiesterase-2, and an allosteric enhancer of the NO-receptive guanylyl cyclase. Under these conditions, cGMP was formed in response to endogenous NO and was found in a population of pyramidal cell somata in area CA3 and subiculum as well as in structures described previously. The additional presence of exogenous NO resulted in hippocampal cGMP reaching the highest level recorded for brain tissue (1700 pmol/mg protein) and in cGMP immunolabelling throughout the pyramidal cell layer. Populations of axons and interneurones were also stained. According with these results, immunohistochemistry for the common NO receptor β1-subunit indicated widespread expression. A similar staining pattern for the α1-subunit with an antibody used previously in the hippocampus and elsewhere, however, proved to be artefactual. The results indicate that the targets of NO in the hippocampus are more varied and extensive than previous evidence had suggested and, in particular, that the pyramidal neurones participating in NO-dependent synaptic plasticity are direct NO targets.
Collapse
|