1
|
Sose PM, Kale PP, Doshi GM. Deciphering the Role of Peroxisome Proliferator-activated Receptor α and Phosphodiesterase Type 5 Targets in Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:956-970. [PMID: 37670711 DOI: 10.2174/1871527323666230904150841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/05/2023] [Accepted: 07/20/2023] [Indexed: 09/07/2023]
Abstract
The most prevalent cause of dementia is Alzheimer's disease (AD). Although the global AD rate is on a constant rise, medical research is yet to find a cure for this neurological condition. Current available therapeutic drugs for AD treatment only provide symptomatic alleviation. Therefore, it is essential to establish effective AD treatment strategies in addressing clinical needs. The development of disease-modifying treatments for use in the disease's early stages and the advancement of symptomatic drugs principally used in the disease's later stages are priorities in AD research. Given that the etiology of AD is difficult to comprehend, using a multimodal therapy intervention that targets molecular targets of AD-related degenerative processes is a practical strategy to change the course of AD progression. The current review article discussed PPAR-α (Peroxisome proliferator-activated receptor-α) and PDE5 (Phosphodiesterase type 5) targets with evidence for their preclinical and clinical importance. Furthermore, we support the targets with AD-related processes, functions, and remedial measures. A unique synergistic method for treating AD may involve the beneficial combinatorial targeting of these two receptors. Furthermore, we reviewed different PDE chemical families in this research and identified PDE5 inhibitors as one of the promising AD-related experimental and clinical disease-modifying medications. Lastly, we suggest jointly targeting these two pathways would be more beneficial than monotherapy in AD treatments.
Collapse
Affiliation(s)
- Parnika M Sose
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle West, Mumbai-400056, India
| | - Pravin P Kale
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle West, Mumbai-400056, India
| | - Gaurav M Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle West, Mumbai-400056, India
| |
Collapse
|
2
|
Liu Z, Liu Q, Zhang B, Liu Q, Fang L, Gou S. Blood-Brain Barrier Permeable and NO-Releasing Multifunctional Nanoparticles for Alzheimer's Disease Treatment: Targeting NO/cGMP/CREB Signaling Pathways. J Med Chem 2021; 64:13853-13872. [PMID: 34517696 DOI: 10.1021/acs.jmedchem.1c01240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The development of novel therapeutic strategies for combating Alzheimer's disease (AD) is challenging but imperative. Multifunctional nanoparticles are promising tools for regulating complex pathological dysfunctions for AD treatment. Herein, we constructed multifunctional nanoparticles consisting of regadenoson (Reg), nitric oxide (NO) donor, and YC-1 in a single molecular entity that can spontaneously self-assemble into nanoparticles and load donepezil to yield Reg-nanoparticles (Reg-NPs). The Reg moiety enabled the Reg-NPs to effectively regulate tight junction-associated proteins in the blood-brain barrier, thus facilitating the permeation of donepezil through the barrier and its accumulation in the brain. Moreover, the released NO and YC-1 activated the NO/cGMP/CREB signaling pathway by stimulating soluble guanylyl cyclase and inhibiting phosphodiesterase activity, which finally reduced cytotoxicity induced by aggregated Aβ in the neurons and was beneficial for synaptic plasticity and memory formation.
Collapse
Affiliation(s)
- Zhikun Liu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Qingqing Liu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Bin Zhang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Qiao Liu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Lei Fang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Shaohua Gou
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
3
|
Maltsev AV, Bal NV, Balaban PM. LTP suppression by protein synthesis inhibitors is NO-dependent. Neuropharmacology 2018; 146:276-288. [PMID: 30540927 DOI: 10.1016/j.neuropharm.2018.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/19/2018] [Accepted: 12/08/2018] [Indexed: 01/25/2023]
Abstract
For several decades, the ability of protein synthesis inhibitors (PSI) to suppress the long-term potentiation (LTP) of hippocampal responses is known. It is considered that mechanisms of such impairment are related to a cessation of translation and a delayed depletion of the protein pool required for maintenance of synaptic plasticity. The present study demonstrates that cycloheximide or anisomycin applications reduce amplitudes of the field excitatory postsynaptic potentials as well as the presynaptically mediated form of plasticity, the paired-pulse facilitation after LTP induction in neurons of the CA1 area of hippocampus. We showed that nitric oxide signaling could be one of the pathways that cause the LTP decrease induced by cycloheximide or anisomycin. Inhibitor of the NO synthase, L-NNA or the NO scavenger, PTIO, rescued the late-phase LTP and restored the paired-pulse facilitation up to the control levels. For the first time we have directly measured the nitric oxide production induced by application of the translation blockers in hippocampal neurons using the NO-sensitive dye DAF-FM. Inhibitory analysis demonstrated that changes during protein synthesis blockade downstream the NO signaling cascade are cGMP-independent and apparently are implemented through degradation of target proteins. Prolonged application of the NO donor SNAP impaired the LTP maintenance in the same manner as PSI.
Collapse
Affiliation(s)
- Alexander V Maltsev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerovа 5A, 117485, Moscow, Russia
| | - Natalia V Bal
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerovа 5A, 117485, Moscow, Russia.
| | - Pavel M Balaban
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerovа 5A, 117485, Moscow, Russia
| |
Collapse
|
4
|
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. [DOI: 10.1097/fbp.0000000000000263] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
5
|
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: 2.1] [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
|
6
|
Gray DT, Engle JR, Recanzone GH. Age-related neurochemical changes in the rhesus macaque superior olivary complex. J Comp Neurol 2013; 522:573-91. [PMID: 25232570 DOI: 10.1002/cne.23427] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Positive immunoreactivity to the calcium-binding protein parvalbumin (PV) and nitric oxide synthase NADPH diaphorase (NADPHd) is well documented within neurons of the central auditory system of both rodents and primates. These proteins are thought to play roles in the regulation of auditory processing. Studies examining the age-related changes in expression of these proteins have been conducted primarily in rodents but are sparse in primate models. In the brainstem, the superior olivary complex (SOC) is crucial for the computation of sound source localization in azimuth, and one hallmark of age-related hearing deficits is a reduced ability to localize sounds. To investigate how these histochemical markers change as a function of age and hearing loss, we studied eight rhesus macaques ranging in age from 12 to 35 years. Auditory brainstem responses (ABRs) were obtained in anesthetized animals for click and tone stimuli. The brainstems of the sesame animals were then stained for PV and NADPHd reactivity. Reactive neurons in the three nuclei of the SOC were counted, and the densities of each cell type were calculated. We found that PV and NADPHd expression increased with both age and ABR thresholds in the medial superior olive but not in either the medial nucleus of the trapezoid body or the lateral superior olive. Together these results suggest that the changes in protein expression employed by the SOC may compensate for the loss of efficacy of auditory sensitivity in the aged primate.
Collapse
Affiliation(s)
- Daniel T Gray
- Center for Neuroscience, University of California at Davis, Davis, California 95616
| | | | | |
Collapse
|
7
|
Nitric oxide synthase inhibition reverts muscarinic receptor down-regulation induced by pilocarpine- and kainic acid-evoked seizures in rat fronto-parietal cortex. Epilepsy Res 2013; 108:11-9. [PMID: 24246145 DOI: 10.1016/j.eplepsyres.2013.10.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 09/02/2013] [Accepted: 10/18/2013] [Indexed: 11/20/2022]
Abstract
We investigated how nitric oxide (NO) synthase inhibitor modulates muscarinic receptor expression in epileptic rats. We found that subchronic treatment (4 days) with Nω-nitro-l-arginine reduced the down-regulation of muscarinic receptors induced by pilocarpine and kainic acid in rat fronto-parietal cortex, notwithstanding the dramatic potentiation of seizures induced by both convulsants. Furthermore, functional experiments in fronto-parietal cortex slices, showed that Nω-nitro-l-arginine reduces the down-regulating effect of pilocarpine on carbachol-induced phosphoinositol hydrolysis. Finally, Nω-nitro-l-arginine greatly potentiated the induction of basic fibroblast growth factor (FGF2) by pilocarpine. These data suggest a potential role of NO in a regulatory feedback loop to control muscarinic receptor signal during seizures. The dramatic potentiation of convulsions by NO synthase inhibitors in some animal models of seizures could derive from preventing this feedback loop.
Collapse
|
8
|
Tamagnini F, Barker G, Warburton EC, Burattini C, Aicardi G, Bashir ZI. Nitric oxide-dependent long-term depression but not endocannabinoid-mediated long-term potentiation is crucial for visual recognition memory. J Physiol 2013; 591:3963-79. [PMID: 23671159 PMCID: PMC3764640 DOI: 10.1113/jphysiol.2013.254862] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Synaptic plasticity in perirhinal cortex is essential for recognition memory. Nitric oxide and endocannabinoids (eCBs), which are produced in the postsynaptic cell and act on the presynaptic terminal, are implicated in mechanisms of long-term potentiation (LTP) and long-term depression (LTD) in other brain regions. In this study, we examine these two retrograde signalling cascades in perirhinal cortex synaptic plasticity and in visual recognition memory in the rat. We show that inhibition of NO-dependent signalling prevented both carbachol- and activity (5 Hz)-dependent LTD but not activity (100 Hz theta burst)-dependent LTP in the rat perirhinal cortex in vitro. In contrast, inhibition of the eCB-dependent signalling prevented LTP but not the two forms of LTD in vitro. Local administration into perirhinal cortex of the nitric oxide synthase inhibitor NPA (2 μm) disrupted acquisition of long-term visual recognition memory. In contrast, AM251 (10 μm), a cannabinoid receptor 1 antagonist, did not impair visual recognition memory. The results of this study demonstrate dissociation between putative retrograde signalling mechanisms in LTD and LTP in perirhinal cortex. Thus, LTP relies on cannabinoid but not NO signalling, whilst LTD relies on NO- but not eCB-dependent signalling. Critically, these results also establish, for the first time, that NO- but not eCB-dependent signalling is important in perirhinal cortex-dependent visual recognition memory.
Collapse
Affiliation(s)
- Francesco Tamagnini
- School of Physiology and Pharmacology, Medical Research Council Centre for Synaptic Plasticity, Bristol University, UK
| | | | | | | | | | | |
Collapse
|
9
|
Wang G, Qi Y, Gao L, Li G, Lv X, Jin Y. Effects of subacute exposure to 1,2-dichloroethane on mouse behavior and the related mechanisms. Hum Exp Toxicol 2012; 32:983-91. [DOI: 10.1177/0960327112470270] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The aim of this study was to explore the effects of subacute exposure to 1,2-dichloroethane (1,2-DCE) on mouse behavior and the related mechanisms focusing on alteration of oxidative stress and amino acid neurotransmitters in the brain. Mouse behavior was examined by open field test. Levels of nitric oxide (NO), malondialdehyde (MDA) and nonprotein sulfhydryl (NPSH) and activity of inducible nitric oxide synthase (iNOS) and superoxide dismutase (SOD) were determined by colorimetric method. Contents of glutamate (Glu), aspartate (Asp) and gamma-aminobutyric acid (GABA) were evaluated by high-performance liquid chromatography. Reduced locomotor and exploratory activities and increased anxiety were found in 0.45 and 0.9 g/m3 1,2-DCE-treated mice. In contrast, increased excitability was found in 0.225 g/m3 1,2-DCE-treated mice. Compensatory antioxidant status and increased NOS activity and NO level in the brain were found in 1,2-DCE-treated mice. Moreover, Glu contents in 1,2-DCE-treated mice and GABA contents in 0.9 g/m3 1,2-DCE-treated mice increased, whereas GABA contents in 0.225 g/m3 1,2-DCE-treated mice decreased significantly compared with control. Taken together, our results suggested that mouse behavior could be disturbed by subacute exposure to 1,2-DCE, and the changes of amino acid neurotransmitter in the brain might be related to the behavioral effects.
Collapse
Affiliation(s)
- G. Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Y. Qi
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - L. Gao
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - G. Li
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - X. Lv
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Y.P. Jin
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People’s Republic of China
| |
Collapse
|
10
|
Zhao F, Wang Y, Jin Y, Zhong Y, Yu X, Li G, Lv X, Sun G. Effects of exogenous methionine on arsenic burden and NO metabolism in brain of mice exposed to arsenite through drinking water. ENVIRONMENTAL TOXICOLOGY 2012; 27:700-706. [PMID: 21560223 DOI: 10.1002/tox.20689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Revised: 11/24/2010] [Accepted: 11/26/2010] [Indexed: 05/30/2023]
Abstract
The aim of this study was to explore the effects of exogenous methionine (Met) on arsenic burden and metabolism of nitric oxide (NO) in the brain of mice exposed to arsenite via drinking water. Mice were exposed to sodium arsenite through drinking water contaminated with 50 mg/L arsenic for four consecutive weeks, and treated intraperitoneally with saline solution, 100 mg/kg body weight (b.w), 200 mg/kg b.w or 400 mg/kg b.w of Met, respectively at the fourth week. Levels of inorganic arsenic (iAs), monomethylarsenic acid (MMAs), and dimethylarsenic acid (DMAs) in the liver, blood and brain were determined by method of hydride generation coupled with atomic absorption spectrophotometry. Nitric oxide synthase (NOS) activities and NO levels in the brain were determined by colorimetric method. Compared with mice exposed to arsenite alone, administration of Met increased significantly the primary methylation ratio in the liver, which resulted in decrease of percent iAs and increase of percent DMAs in the liver, and decrease of iAs, MMAs and total arsenic levels (TAs) in the blood and DMAs and TAs in the brain. NOS activities and NO levels in the brain of mice exposed to arsenite alone were significantly lower than those in control, however administration of Met could increase significantly NO levels. Findings from this study suggested that exogenous Met could benefit the primary arsenic methylation in the liver, which might increase the production of methylated arsenicals and facilitate arsenic excretion. As a consequence, arsenic burden in both blood and brain was reduced, and toxic effects on NO metabolism in the brain were ameliorated.
Collapse
Affiliation(s)
- Fenghong Zhao
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Johnstone VPA, Raymond CR. A protein synthesis and nitric oxide-dependent presynaptic enhancement in persistent forms of long-term potentiation. Learn Mem 2011; 18:625-33. [PMID: 21933902 DOI: 10.1101/lm.2245911] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Long-term potentiation (LTP) is an important process underlying learning and memory in the brain. At CA3-CA1 synapses in the hippocampus, three discrete forms of LTP (LTP1, 2, and 3) can be differentiated on the basis of maintenance and induction mechanisms. However, the relative roles of pre- and post-synaptic expression mechanisms in LTP1, 2, and 3 are unknown. Neurotransmitter release in the expression of LTP1, 2, and 3 was measured via FM 1-43 destaining from CA3 terminals in hippocampal slices from male Wistar rats (7-8 wk). No difference in vesicle turnover rate was observed for LTP1 up to 160 min following induction by one train of theta-burst stimulation (1TBS). A presynaptic enhancement was found for LTP2 at 160 min after induction by 4TBS, and for LTP3 at both 80 and 160 min after induction by 8TBS. Inhibition of nitric oxide (NO) signaling blocked both LTP2 and LTP3 maintenance and the associated enhanced release. LTP2 maintenance and its presynaptic expression were dependent on protein synthesis, but not gene transcription. LTP3 maintenance was dependent on both translation and transcription, but like LTP2, the enhanced release only required translation. These data considerably strengthen the mechanistic separation of LTP1, 2, and 3, supporting a model of multiple, discrete forms of LTP at CA3-CA1 synapses rather than different temporal phases.
Collapse
Affiliation(s)
- Victoria P A Johnstone
- Department of Neuroscience, The John Curtin School of Medical Research & Eccles Institute of Neuroscience, The Australian National University, Canberra ACT 0200, Australia
| | | |
Collapse
|
12
|
Neitz A, Mergia E, Eysel UT, Koesling D, Mittmann T. Presynaptic nitric oxide/cGMP facilitates glutamate release via hyperpolarization-activated cyclic nucleotide-gated channels in the hippocampus. Eur J Neurosci 2011; 33:1611-21. [PMID: 21410795 DOI: 10.1111/j.1460-9568.2011.07654.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In hippocampal neurons, synaptic transmission is affected by a variety of modulators, including nitric oxide (NO), which was proposed as a retrograde messenger as long as two decades ago. NO signals via two NO-sensitive guanylyl cyclases (NO-GCs) (NO-GC1 and NO-GC2) and the subsequent increase in cGMP. Lack of long-term potentiation in mice deficient in either one of the two NO-GCs demonstrates the involvement of both NO-GCs in synaptic transmission. However, the physiological consequences of NO/cGMP and the cellular mechanisms involved are unknown. Here, we analyzed glutamatergic synaptic transmission, most likely reflecting glutamate release, in the hippocampal CA1 region of NO-GC knockout mice by single-cell recording, and found glutamate release to be reduced under basal and stimulated conditions in the NO-GC1 knockout mice, but restorable to wild-type-like levels with a cGMP analog. Conversely, an inhibitor of NO/cGMP signaling, ODQ, reduced glutamate release in wild-type mice to knockout-like levels; thus, we conclude that presynaptic cGMP formed by NO-GC1 facilitates glutamate release. In this pathway, NO is supplied by endothelial NO synthase. In search of a cGMP target, we found that two mechanistically distinct blockers of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels (ZD7288 and DK-AH269) abolished the cGMP-induced increase in glutamate release, suggesting that cGMP either directly or indirectly signals via HCN channels. In summary, we unravel a presynaptic role of NO/cGMP most likely in glutamate release and propose that HCN channels act as effectors for cGMP.
Collapse
Affiliation(s)
- Angela Neitz
- Department of Neurophysiology, Medical School, Ruhr-University Bochum, Germany
| | | | | | | | | |
Collapse
|
13
|
Currás-Collazo MC. Nitric oxide signaling as a common target of organohalogens and other neuroendocrine disruptors. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2011; 14:495-536. [PMID: 21790323 DOI: 10.1080/10937404.2011.578564] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Organohalogen compounds such as polychlorinated biphenyls (PCB) and polybrominated diphenyl ethers (PBDE) are global environmental pollutants and highly persistent, bioaccumulative chemicals that produce adverse effects in humans and wildlife. Because of the widespread use of these organohalogens in household items and consumer products, indoor contamination is a significant source of human exposure, especially for children. One significant concern with regard to health effects associated with exposure to organohalogens is endocrine disruption. Toxicological studies on organohalogen pollutants primarily focused on sex steroid and thyroid hormone actions, and findings have largely shaped the way one envisions their disruptive effects occurring. Organohalogens exert additional effects on other systems including other complex endocrine systems that may be disregulated at various levels of organization. Over the last 20 years evidence has mounted in favor of a critical role of nitric oxide (NO) in numerous functions ranging from neuroendocrine functions to learning and memory. With its participation in multiple systems and action at several levels of integration, NO signaling has a pervasive influence on nervous and endocrine functions. Like blockers of NO synthesis, PCBs and PBDEs produce multifaceted effects on physiological systems. Based on this unique set of converging information it is proposed that organohalogen actions occur, in part, by hijacking processes associated with this ubiquitous bioactive molecule. The current review examines the emerging evidence for NO involvement in selected organohalogen actions and includes recent progress from our laboratory that adds to our current understanding of the actions of organohalogens within hypothalamic neuroendocrine circuits. The thyroid, vasopressin, and reproductive systems as well as processes associated with long-term potentiation were selected as sample targets of organohalogens that rely on regulation by NO. Information is provided about other toxicants with demonstrated interference of NO signaling. Our focus on the convergence between NO system and organohalogen toxicity offers a novel approach to understanding endocrine and neuroendocrine disruption that is particularly problematic for developing organisms. This new working model is proposed as a way to encourage future study in elucidating common mechanisms of action that are selected with a better operational understanding of the systems affected.
Collapse
Affiliation(s)
- Margarita C Currás-Collazo
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, California 92521, USA.
| |
Collapse
|
14
|
Wang Y, Zhao F, Jin Y, Zhong Y, Yu X, Li G, Lv X, Sun G. Effects of exogenous glutathione on arsenic burden and NO metabolism in brain of mice exposed to arsenite through drinking water. Arch Toxicol 2010; 85:177-84. [DOI: 10.1007/s00204-010-0573-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 06/22/2010] [Indexed: 11/25/2022]
|
15
|
Nitric oxide neurons and neurotransmission. Prog Neurobiol 2010; 90:246-55. [DOI: 10.1016/j.pneurobio.2009.10.007] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2008] [Revised: 04/22/2009] [Accepted: 10/09/2009] [Indexed: 11/24/2022]
|
16
|
Reierson GW, Mastronardi CA, Licinio J, Wong ML. Repeated antidepressant therapy increases cyclic GMP signaling in rat hippocampus. Neurosci Lett 2009; 466:149-53. [PMID: 19788915 PMCID: PMC2804064 DOI: 10.1016/j.neulet.2009.09.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 09/16/2009] [Accepted: 09/24/2009] [Indexed: 12/31/2022]
Abstract
Cyclic adenosine monophosphpate (cAMP) signaling is thought to be involved in the pathophysiology of major depressive disorder and antidepressant action; however, relatively little is known about the possible role of cyclic guanosine monophosphate (cGMP) signaling. Accumulating evidence suggests that crosstalk occurs between cAMP and cGMP pathways. There is a need to clarify the trajectory of cAMP and cGMP concentrations, their synthesis by cyclases, and degradation by phosphodiesterases (PDEs) to understand the role of cyclic mononucleotide signaling in the effect of chronic antidepressant therapy. We used quantitative real-time PCR and enzyme immunoassay to systematically investigate the expression of intracellular signaling cascade elements in the hippocampus of rats chronically treated with the antidepressants fluoxetine and amitriptyline. We found increased cGMP levels, which were consistent with our findings of decreased PDE gene expression. Immunoassay results showed unchanged cAMP levels. We conclude that increased cGMP signaling might underlie the efficacy of chronic antidepressant treatment.
Collapse
Affiliation(s)
- Gillian W. Reierson
- Center on Pharmacogenomics, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1580 NW 10 Avenue, Miami, FL 33136, USA
| | - Claudio A. Mastronardi
- Center on Pharmacogenomics, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1580 NW 10 Avenue, Miami, FL 33136, USA
| | - Julio Licinio
- Center on Pharmacogenomics, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1580 NW 10 Avenue, Miami, FL 33136, USA
| | - Ma-Li Wong
- Center on Pharmacogenomics, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1580 NW 10 Avenue, Miami, FL 33136, USA
| |
Collapse
|
17
|
Abstract
The age of an experimental animal can be a critical variable, yet age matters are often overlooked within neuroscience. Many studies make use of young animals, without considering possible differences between immature and mature subjects. This is especially problematic when attempting to model traits or diseases that do not emerge until adulthood. In this commentary we discuss the reasons for this apparent bias in age of experimental animals, and illustrate the problem with a systematic review of published articles on long-term potentiation. Additionally, we review the developmental stages of a rat and discuss the difficulty of using the weight of an animal as a predictor of its age. Finally, we provide original data from our laboratory and review published data to emphasize that development is an ongoing process that does not end with puberty. Developmental changes can be quantitative in nature, involving gradual changes, rapid switches, or inverted U-shaped curves. Changes can also be qualitative. Thus, phenomena that appear to be unitary may be governed by different mechanisms at different ages. We conclude that selection of the age of the animals may be critically important in the design and interpretation of neurobiological studies.
Collapse
Affiliation(s)
- James Edgar McCutcheon
- Department of Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | | |
Collapse
|
18
|
Beninger RJ, Forsyth JK, Van Adel M, Reynolds JN, Boegman RJ, Jhamandas K. Subchronic MK-801 behavioural deficits in rats: Partial reversal by the novel nitrate GT 1061. Pharmacol Biochem Behav 2009; 91:495-502. [DOI: 10.1016/j.pbb.2008.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Revised: 08/29/2008] [Accepted: 09/03/2008] [Indexed: 01/03/2023]
|
19
|
Abstract
The dorsal lateral geniculate nucleus (dLGN) not only serves as the obligatory pathway for visual information transfer from the retina to neocortex but can also generate intrathalamic rhythmic activities associated with different arousal states and certain pathological conditions. The gating activity of thalamocortical circuits is under neuromodulatory control by various brainstem nuclei as well as intrinsic thalamic neurons (e.g. thalamic reticular nucleus (TRN) neurons and dLGN interneurons). In this study, we examined the effect of the putative neuromodulator nitric oxide (NO) on thalamic neuron excitability. There are multiple potential sources of NO in thalamus: cholinergic terminals originating from brainstem nuclei, GABAergic TRN neurons, and local GABAergic interneurons. Using whole cell recording techniques in in vitro thalamic slices, we found that the NO donor SNAP produced a robust, long-lasting depolarization in TRN neurons, a weaker depolarization in thalamocortical relay neurons, and no effect in local interneurons. SNAP preferentially depolarized stereotypical TRN neurons that could produced strong burst discharge. In contrast, SNAP had little effect on atypical burst and non-burst TRN cells. The NO donor SIN-1 and the endogenous NO precursor, L-arginine, mimicked the SNAP-mediated actions. The NO-mediated depolarizations were blocked by the guanylyl cyclase inhibitor ODQ indicating involvement of the cGMP pathway. In addition, the phosphodiesterase (PDE) inhibitor zaprinast depolarized and occluded the NO-mediated depolarization in TRN neurons. At the circuit level, NO activation significantly attenuated intrathalamic rhythmic activities likely resulting from the shifting of the firing mode of thalamic neurons, perhaps both TRN and thalamocortical neurons, from burst- to tonic-discharge mode. These alterations in thalamic neuron excitability not only change rhythmic circuit activity, but could also influence sensory information processing through thalamocortical circuits.
Collapse
Affiliation(s)
- Sunggu Yang
- Department of Molecular and Integrative Physiology, University of Illinois, 2357 Beckman Institute, 405 North Mathews, Urbana, IL 61801, USA
| | | |
Collapse
|
20
|
Puzzo D, Sapienza S, Arancio O, Palmeri A. Role of phosphodiesterase 5 in synaptic plasticity and memory. Neuropsychiatr Dis Treat 2008; 4:371-87. [PMID: 18728748 PMCID: PMC2518390 DOI: 10.2147/ndt.s2447] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Phosphodiesterases (PDEs) are enzymes that break down the phosphodiesteric bond of the cyclic nucleotides, cAMP and cGMP, second messengers that regulate many biological processes. PDEs participate in the regulation of signal transduction by means of a fine regulation of cyclic nucleotides so that the response to cell stimuli is both specific and activates the correct third messengers. Several PDE inhibitors have been developed and used as therapeutic agents because they increase cyclic nucleotide levels by blocking the PDE function. In particular, sildenafil, an inhibitor of PDE5, has been mainly used in the treatment of erectile dysfunction but is now also utilized against pulmonary hypertension. This review examines the physiological role of PDE5 in synaptic plasticity and memory and the use of PDE5 inhibitors as possible therapeutic agents against disorders of the central nervous system (CNS).
Collapse
Affiliation(s)
- Daniela Puzzo
- Dept of Physiological Sciences, University of Catania Catania, Italy.
| | | | | | | |
Collapse
|
21
|
Clasadonte J, Poulain P, Beauvillain JC, Prevot V. Activation of neuronal nitric oxide release inhibits spontaneous firing in adult gonadotropin-releasing hormone neurons: a possible local synchronizing signal. Endocrinology 2008; 149:587-96. [PMID: 18006627 DOI: 10.1210/en.2007-1260] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The activation of nitric oxide (NO) signaling pathways in hypothalamic neurons plays a key role in the control of GnRH secretion that is central to reproductive function. It is unknown whether NO directly modulates the firing behavior of GnRH neurons in the preoptic region of the mature brain. Using patch-clamp recordings from GnRH neurons expressing green fluorescent protein in adult mice brain slices, we demonstrate that the NO precursor, L-arginine (Arg), or the NO donor, diethylamine/NO, induced a robust and reversible reduction in the spontaneous firing activity of GnRH neurons, including bursting activity. The effects of L-Arg were prevented by the NO synthase inhibitor N omega-nitro-L-Arg methyl ester hydrochloride. Histochemical studies revealing a close anatomical relationship between neurons producing NO and GnRH perikarya, together with the loss of the L-Arg-mediated inhibition of GnRH neuronal activity via the selective blockade of neuronal NO synthase, suggested that the primary source of local NO production in the mouse preoptic region was neuronal. Synaptic transmission uncoupling did not alter the effect of NO, suggesting that NO affects the firing pattern of GnRH neurons by acting at a postsynaptic site. We also show that the NO-mediated changes in membrane properties in the GnRH neurons require soluble guanylyl cyclase activity and may involve potassium conductance. By revealing that NO is a direct modulator of GnRH neuronal activity, our results introduce the intriguing possibility that this gaseous neurotransmitter may be used by the sexual brain to modulate burst firing patterns. It may set into phase the bursting activity of GnRH neurons at key stages of reproductive physiology.
Collapse
Affiliation(s)
- Jérôme Clasadonte
- Inserm, Jean-Pierre Aubert Research Center, Unité 837, Development and Plasticity of the Postnatal Brain, Place de Verdun, 59045, Lille Cedex, France
| | | | | | | |
Collapse
|
22
|
Serulle Y, Zhang S, Ninan I, Puzzo D, McCarthy M, Khatri L, Arancio O, Ziff EB. A GluR1-cGKII interaction regulates AMPA receptor trafficking. Neuron 2008; 56:670-88. [PMID: 18031684 DOI: 10.1016/j.neuron.2007.09.016] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 11/13/2006] [Accepted: 09/14/2007] [Indexed: 10/24/2022]
Abstract
Trafficking of AMPA receptors (AMPARs) is regulated by specific interactions of the subunit intracellular C-terminal domains (CTDs) with other proteins, but the mechanisms involved in this process are still unclear. We have found that the GluR1 CTD binds to cGMP-dependent protein kinase II (cGKII) adjacent to the kinase catalytic site. Binding of GluR1 is increased when cGKII is activated by cGMP. cGKII and GluR1 form a complex in the brain, and cGKII in this complex phosphorylates GluR1 at S845, a site also phosphorylated by PKA. Activation of cGKII by cGMP increases the surface expression of AMPARs at extrasynaptic sites. Inhibition of cGKII activity blocks the surface increase of GluR1 during chemLTP and reduces LTP in the hippocampal slice. This work identifies a pathway, downstream from the NMDA receptor (NMDAR) and nitric oxide (NO), which stimulates GluR1 accumulation in the plasma membrane and plays an important role in synaptic plasticity.
Collapse
Affiliation(s)
- Yafell Serulle
- Program in Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Liu S, Fa M, Ninan I, Trinchese F, Dauer W, Arancio O. Alpha-synuclein involvement in hippocampal synaptic plasticity: role of NO, cGMP, cGK and CaMKII. Eur J Neurosci 2007; 25:3583-96. [PMID: 17610578 DOI: 10.1111/j.1460-9568.2007.05569.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Synaptic plasticity involves a series of coordinate changes occurring both pre- and postsynaptically, of which alpha-synuclein is an integral part. We have investigated on mouse primary hippocampal neurons in culture whether redistribution of alpha-synuclein during plasticity involves retrograde signaling activation through nitric oxide (NO), cGMP, cGMP-dependent protein kinase (cGK) and calmodulin-dependent protein kinase II. We have found that deletion of the alpha-synuclein gene blocks both the long-lasting enhancement of evoked and miniature transmitter release and the increase in the number of functional presynaptic boutons evoked through the NO donor, DEA/NO, and the cGMP analog, 8-Br-cGMP. In agreement with these findings both DEA/NO and 8-Br-cGMP were capable of producing a long-lasting increase in number of clusters for alpha-synuclein through activation of soluble guanylyl cyclase, cGK and calcium/calmodulin-dependent protein kinase IIalpha. Thus, our results suggest that NO, cGMP, GMP-dependent protein kinase and calmodulin-dependent protein kinase II play a key role in the redistribution of alpha-synuclein during plasticity.
Collapse
Affiliation(s)
- Shumin Liu
- Department of Pathology, Taub Institute, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
24
|
Haghikia A, Mergia E, Friebe A, Eysel UT, Koesling D, Mittmann T. Long-term potentiation in the visual cortex requires both nitric oxide receptor guanylyl cyclases. J Neurosci 2007; 27:818-23. [PMID: 17251421 PMCID: PMC6672906 DOI: 10.1523/jneurosci.4706-06.2007] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The role of nitric oxide (NO)/cGMP signaling in long-term potentiation (LTP) has been a lingering matter of debate. Within the cascade, the NO receptor guanylyl cyclase (GC), the cGMP-forming enzyme that is stimulated by NO, plays a key role. Two isoforms of GC (alpha2-GC, alpha1-GC) exist. To evaluate their contribution to synaptic plasticity, we analyzed knock-out mice lacking either one of the GC isoforms. We found that LTP induced in the visual cortex is NO dependent in the wild-type mice, absent in either of the GC isoform-deficient mice, and restored with application of a cGMP analog in both strains. The requirement of both NO receptor GCs for LTP indicates the existence of two distinct NO/cGMP-mediated pathways, which have to work in concert for expression of LTP.
Collapse
Affiliation(s)
- Arash Haghikia
- Faculty of Medicine, Institute of Physiology, Department of Neurophysiology, and
| | - Evanthia Mergia
- Faculty of Medicine, Institute of Pharmacology and Toxicology, Ruhr-University Bochum, D-44780 Bochum, Germany
| | - Andreas Friebe
- Faculty of Medicine, Institute of Pharmacology and Toxicology, Ruhr-University Bochum, D-44780 Bochum, Germany
| | - Ulf T. Eysel
- Faculty of Medicine, Institute of Physiology, Department of Neurophysiology, and
| | - Doris Koesling
- Faculty of Medicine, Institute of Pharmacology and Toxicology, Ruhr-University Bochum, D-44780 Bochum, Germany
| | - Thomas Mittmann
- Faculty of Medicine, Institute of Physiology, Department of Neurophysiology, and
| |
Collapse
|
25
|
Puzzo D, Palmeri A, Arancio O. Involvement of the nitric oxide pathway in synaptic dysfunction following amyloid elevation in Alzheimer's disease. Rev Neurosci 2007; 17:497-523. [PMID: 17180876 DOI: 10.1515/revneuro.2006.17.5.497] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Amyloid-beta (Abeta), a peptide thought to play a crucial role in Alzheimer's disease (AD), has attracted scientific interest with the aim of characterizing the mechanisms by which it is involved in AD pathogenesis. Abeta has been found to markedly impair hippocampal long-term potentiation (LTP), a widely studied cellular model of synaptic plasticity that is thought to underlie learning and memory. The overall purpose of this review is to define the role of the nitric oxide (NO)/cGMP/cAMP-regulatory element binding (CREB) pathway in beta-amyloid-induced changes of basal neurotransmission and synaptic plasticity in the hippocampus, a structure within the temporal lobe of the brain critical for memory storage.
Collapse
Affiliation(s)
- Daniela Puzzo
- Department of Pathology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| | | | | |
Collapse
|
26
|
Arrigoni E, Rosenberg PA. Nitric oxide-induced adenosine inhibition of hippocampal synaptic transmission depends on adenosine kinase inhibition and is cyclic GMP independent. Eur J Neurosci 2007; 24:2471-80. [PMID: 17100836 DOI: 10.1111/j.1460-9568.2006.05124.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Adenosine is an important inhibitory neuromodulator that regulates neuronal excitability. Several studies have shown that nitric oxide induces release of adenosine. Here we investigated the mechanism of this release. We studied the effects of nitric oxide on evoked field excitatory postsynaptic potentials (fEPSPs) recorded in the CA1 area of rat hippocampal slices. The nitric oxide donor 1,1-diethyl-2-hydroxy-2-nitroso-hydrazine sodium (DEA/NO; 100 microm) depressed the fEPSP by 77.6 +/- 4.1%. This effect was abolished by the adenosine A1 antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX; 400 nm), indicating that the nitric oxide effect was mediated by adenosine accumulation. The DEA/NO effect was unaltered by the 5'-ectonucleotidase inhibitor alpha,beta-methylene-adenosine 5'-diphosphate (AMP-CP; 100 microm), indicating that extracellular adenosine did not derive from ATP or cAMP release. The guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazole[4,3-a]quinoxaline-1-one (ODQ; 5 microm) did not affect nitric oxide depression of the fEPSPs, indicating that nitric oxide-mediated adenosine release was not mediated through a cGMP signaling cascade. This conclusion was confirmed by the observation that 8-(4-chlorophenylthio)-guanosine-3',5'-cyclic monophosphate (8-pCPT-cGMP; 1 mm) reversibly depressed the fEPSP by 24.9 +/- 4.5%, but this effect was not blocked by adenosine antagonists. Adenosine kinase inhibitor 5-iodotubercidin (ITU; 7 microm) occluded the nitric oxide effects by 74%, suggesting that inhibition of adenosine kinase activity contributes to adenosine release. In conclusion, exogenous nitric oxide evokes adenosine release by a cGMP-independent pathway. Intracellular cGMP elevation partially inhibits the fEPSP but not through adenosine release. Although a direct block of adenosine kinase by nitric oxide can not be excluded, the depression of adenosine kinase activity may be due to inhibition by its own substrate adenosine.
Collapse
Affiliation(s)
- Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | |
Collapse
|
27
|
Tornieri K, Rehder V. Nitric oxide release from a single cell affects filopodial motility on growth cones of neighboring neurons. Dev Neurobiol 2007; 67:1932-43. [PMID: 17874460 DOI: 10.1002/dneu.20572] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nitric oxide (NO), a gaseous messenger, has been reported to be involved in a variety of functions in the nervous system, ranging from neuronal pathfinding to learning and memory. We have shown previously that the application of NO via NO donors to growth cones of identified Helisoma buccal neurons B5 in vitro induces an increase in filopodial length, a decrease in filopodial number, and a slowing in neurite advance. It is unclear, however, whether NO released from a physiological source would affect growth cone dynamics. Here we used cell bodies of identified neurons known to express the NO synthesizing enzyme nitric oxide synthase (NOS) as a source of constitutive NO production and tested their effect on growth cones of other cells in a sender-receiver paradigm. We showed that B5 cell bodies induced a rapid increase in filopodial length in NO-responsive growth cones, and that this effect was blocked by the NOS inhibitor 7-NI, suggesting that the effect was mediated by NO. Inhibition of soluble guanylyl cyclase (sGC) with ODQ blocked filopodial elongation induced by B5 somata, confirming that NO acted via sGC. We also demonstrate that the effect of NO was reversible and that a cell releasing NO can affect growth cones over a distance of at least 100 microm. Our results suggest that NO released from a physiological source can affect the motility of nearby growth cones and thus should be considered a signaling molecule with the potential to affect the outcome of neuronal pathfinding in vivo.
Collapse
Affiliation(s)
- Karine Tornieri
- Department of Biology, Georgia State University, Atlanta, GA 30302-4010, USA
| | | |
Collapse
|
28
|
Zhang XL, Zhou ZY, Winterer J, Müller W, Stanton PK. NMDA-dependent, but not group I metabotropic glutamate receptor-dependent, long-term depression at Schaffer collateral-CA1 synapses is associated with long-term reduction of release from the rapidly recycling presynaptic vesicle pool. J Neurosci 2006; 26:10270-80. [PMID: 17021182 PMCID: PMC6674623 DOI: 10.1523/jneurosci.3091-06.2006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Postsynaptic alterations have been suggested to account for NMDA receptor (NMDAR)-dependent long-term depression (LTD) and long-term potentiation of synaptic strength, although there is substantial evidence supporting changes in presynaptic release. Direct chemical activation of either NMDA or group I metabotropic glutamate receptor (mGluR1) elicits LTD of similar magnitudes, but it is unknown whether they share common expression mechanisms. Using dual-photon laser-scanning microscopy of FM1-43 [N-(3-triethylammoniumpropyl)-4-(4-(dibutylamino)styryl)pyridinium dibromide] to directly visualize presynaptic vesicular release from the rapidly recycling vesicle pool (RRP) at Schaffer collateral terminals in field CA1 of rat hippocampal slices, we found that a persistent reduction in vesicular release from the RRP is induced by NMDA-LTD but not by mGluR1-LTD. Variance-mean analyses of Schaffer collateral release probability (P(r)) at varying extracellular calcium concentrations confirmed that NMDA-LTD was associated with reduced P(r), whereas mGluR1-LTD was not. Pharmacological isolation of NMDAR-dependent and mGluR-dependent forms of stimulus-evoked LTD revealed that both are composed of a combination of presynaptic and postsynaptic alterations. However, when group I mGluR-dependent LTD was isolated by combining an NMDAR blocker with a group II mGluR antagonist, this form of LTD was purely postsynaptic. The nitric oxide synthase inhibitor N omega-nitro-L-arginine blocked the induction of NMDA-LTD but did not alter mGluR-LTD, consistent with a selective role for nitric oxide as a retrograde messenger mediating NMDA-LTD. These data demonstrate that single synapses can express multiple forms of LTD with different sites of expression, that NMDA-LTD is a combination of presynaptic and postsynaptic alterations, but that group I mGluR-LTD appears to be expressed entirely postsynaptically.
Collapse
Affiliation(s)
| | | | - Jochen Winterer
- Neuroscience Research Institute and
- Department of Psychiatry, Charité, Humboldt University, D-10117 Berlin, Germany, and
| | - Wolfgang Müller
- Departments of Neurosurgery, Neurology, and Neuroscience, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131
| | - Patric K. Stanton
- Departments of Cell Biology and Anatomy and
- Neurology, New York Medical College, Valhalla, New York 10595
| |
Collapse
|
29
|
Rintoul GL, Bennett VJ, Papaconstandinou NA, Reynolds IJ. Nitric oxide inhibits mitochondrial movement in forebrain neurons associated with disruption of mitochondrial membrane potential. J Neurochem 2006; 97:800-6. [PMID: 16573650 DOI: 10.1111/j.1471-4159.2006.03788.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Nitric oxide (NO) has a number of physiological and pathophysiological effects in the nervous system. One target of NO is the mitochondrion, where it inhibits respiration and ATP synthesis, which may contribute to NO-mediated neuronal injury. Our recent studies suggested that impaired mitochondrial function impairs mitochondrial trafficking, which could also contribute to neuronal injury. Here, we studied the effects of NO on mitochondrial movement and morphology in primary cultures of forebrain neurons using a mitochondrially targeted enhanced yellow fluorescent protein. NO produced by two NO donors, papa non-oate and diethylamine/NO complex, caused a rapid cessation of mitochondrial movement but did not alter morphology. Movement recovered after removal of NO. The effects of NO on movement were associated with dissipation of the mitochondrial membrane potential. Increasing cGMP levels using 8-bromoguanosine 3',5'-cyclic monophosphate, did not mimic the effects on mitochondrial movement. Furthermore, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), an inhibitor of NO-induced activation of soluble guanylate cyclase, did not block the effects of NO. Thus, neither increasing nor decreasing cGMP levels had an effect on mitochondrial movement. Based on these data, we conclude that NO is a novel modulator of mitochondrial trafficking in neurons, which may act through the inhibition of mitochondrial function.
Collapse
Affiliation(s)
- Gordon L Rintoul
- Department of Pharmacology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | |
Collapse
|
30
|
Philippides A, Ott SR, Husbands P, Lovick TA, O'Shea M. Modeling cooperative volume signaling in a plexus of nitric-oxide-synthase-expressing neurons. J Neurosci 2006; 25:6520-32. [PMID: 16014713 PMCID: PMC6725429 DOI: 10.1523/jneurosci.1264-05.2005] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In vertebrate and invertebrate brains, nitric oxide (NO) synthase (NOS) is frequently expressed in extensive meshworks (plexuses) of exceedingly fine fibers. In this paper, we investigate the functional implications of this morphology by modeling NO diffusion in fiber systems of varying fineness and dispersal. Because size severely limits the signaling ability of an NO-producing fiber, the predominance of fine fibers seems paradoxical. Our modeling reveals, however, that cooperation between many fibers of low individual efficacy can generate an extensive and strong volume signal. Importantly, the signal produced by such a system of cooperating dispersed fibers is significantly more homogeneous in both space and time than that produced by fewer larger sources. Signals generated by plexuses of fine fibers are also better centered on the active region and less dependent on their particular branching morphology. We conclude that an ultrafine plexus is configured to target a volume of the brain with a homogeneous volume signal. Moreover, by translating only persistent regional activity into an effective NO volume signal, dispersed sources integrate neural activity over both space and time. In the mammalian cerebral cortex, for example, the NOS plexus would preferentially translate persistent regional increases in neural activity into a signal that targets blood vessels residing in the same region of the cortex, resulting in an increased regional blood flow. We propose that the fineness-dependent properties of volume signals may in part account for the presence of similar NOS plexus morphologies in distantly related animals.
Collapse
Affiliation(s)
- Andrew Philippides
- Sussex Centre for Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, United Kingdom
| | | | | | | | | |
Collapse
|
31
|
Marcoli M, Cervetto C, Paluzzi P, Guarnieri S, Raiteri M, Maura G. Nitric oxide-evoked glutamate release and cGMP production in cerebellar slices: control by presynaptic 5-HT1D receptors. Neurochem Int 2006; 49:12-9. [PMID: 16469416 DOI: 10.1016/j.neuint.2005.12.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2005] [Revised: 12/05/2005] [Accepted: 12/19/2005] [Indexed: 11/22/2022]
Abstract
We previously reported that pre- and postsynaptic 5-hydroxytryptamine (5-HT) receptors effectively control glutamatergic transmission in adult rat cerebellum. To investigate where 5-HT acts in the glutamate ionotropic receptors/nitric oxide/guanosine 3',5'-cyclic monophosphate (cGMP) pathway, in the present study 5-HT modulation of the cGMP response to the nitric oxide donor S-nitroso-penicillamine (SNAP) was studied in adult rat cerebellar slices. While cGMP elevation produced by high-micromolar SNAP was insensitive to 5-HT, 1 microM SNAP, expected to release nitric oxide in the low-nanomolar concentration range, elicited cGMP production and endogenous glutamate release both of which could be prevented by activating presynaptic 5-HT1D receptors. Released nitric oxide appeared responsible for cGMP production and glutamate release evoked by 1 microM SNAP, as both the effects were mimicked by the structurally unrelated nitric oxide donor 2-(N,N-diethylamino)-diazenolate-2-oxide (0.1 microM). Dependency of the 1 microM SNAP-evoked release of glutamate on external Ca2+, sensitivity to presynaptic release-regulating receptors and dependency on ionotropic glutamate receptor functioning, suggest that nitric oxide stimulates exocytotic-like, activity-dependent glutamate release. Activation of ionotropic glutamate receptors/nitric oxide synthase/guanylyl cyclase pathway by endogenously released glutamate was involved in the cGMP response to 1 microM SNAP, as blockade of NMDA/non-NMDA receptors, nitric oxide synthase or guanylyl cyclase, abolished the cGMP response. To conclude, in adult rat cerebellar slices low-nanomolar exogenous nitric oxide could facilitate glutamate exocytotic-like release possibly from parallel fibers that subsequently activated the glutamate ionotropic receptors/nitric oxide/cGMP pathway. Presynaptic 5-HT1D receptors could regulate the nitric oxide-evoked release of glutamate and subsequent cGMP production.
Collapse
Affiliation(s)
- Manuela Marcoli
- Department of Experimental Medicine, Pharmacology and Toxicology Section, University of Genoa, Viale Cembrano 4, Genoa, Italy
| | | | | | | | | | | |
Collapse
|
32
|
Stanton PK, Winterer J, Zhang XL, Müller W. Imaging LTP of presynaptic release of FM1-43 from the rapidly recycling vesicle pool of Schaffer collateral-CA1 synapses in rat hippocampal slices. Eur J Neurosci 2006; 22:2451-61. [PMID: 16307588 DOI: 10.1111/j.1460-9568.2005.04437.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Recent studies using the styryl dye FM1-43 and two-photon microscopy to directly visualize transmitter release at CA3-CA1 excitatory synapses in the hippocampus have demonstrated that activity-dependent long-term potentiation (LTP) and long-term depression are associated with alterations in vesicular release. It is not known whether particular vesicle pools preferentially express these alterations or what second messenger cascades are involved. To address these questions, we selectively loaded FM1-43 into the rapidly recycling pool (RRP) of vesicles by use of a brief hypertonic shock to release and load the RRP. We demonstrate here that the induction of LTP can lead to a selective long-lasting enhancement in presynaptic release from the RRP, while reserve pool kinetics remain unchanged. LTP of RRP release was N-methyl-d-aspartate receptor-dependent and also required production of the intercellular messenger NO and activation of receptor tyrosine kinase. Measurement of FM1-43 stimulus-evoked uptake rates following induction of LTP confirmed that LTP produces more rapid recycling of vesicles released by electrical stimulation, consistent with an enhanced release probability from the RRP.
Collapse
Affiliation(s)
- Patric K Stanton
- Departments of Cell Biology & Anatomy and Neurology, New York Medical College, Valhalla, NY 10595, USA.
| | | | | | | |
Collapse
|
33
|
Shahraki A, Stone TW. Blockade of presynaptic adenosine A1 receptor responses by nitric oxide and superoxide in rat hippocampus. Eur J Neurosci 2004; 20:719-28. [PMID: 15255982 DOI: 10.1111/j.1460-9568.2004.03502.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Activation of N-methyl-D-aspartate (NMDA) receptors prevents the neuronal responses to adenosine in hippocampal slices. As NMDA receptor activation leads to the generation of nitric oxide (NO) and superoxide, we have examined whether these can modify neuronal responses to adenosine and mediate the actions of NMDA. Field excitatory postsynaptic potentials were recorded in the CA1 region of rat hippocampal slices. Paired-pulse interactions were studied to localize the observed interactions to presynaptic terminals. The NO donors S-nitroso-N-acetylpenicillamine and diethylamine NONOate induced a long-lasting potentiation (NO-induced potentiation) of field excitatory postsynaptic potential slope and significantly prevented the presynaptic inhibitory effect of adenosine or the A1 receptor agonist N6-cyclopentyladenosine selectively with no effect on responses to baclofen. The superoxide-generating system of xanthine/xanthine oxidase also prevented presynaptic responses to adenosine and this effect was prevented by superoxide dismutase (SOD). The guanylate cyclase inhibitor 1H-[1,2,4]-oxadiazolo[4,3a]quinoxalin-1-one (10 microM) prevented NO-induced potentiation and the inhibitory effects of S-nitroso-N-acetylpenicillamine and xanthine/xanthine oxidase on adenosine responses. The inhibitory effect of NMDA on adenosine responses was unchanged by 1H-[1,2,4]-oxadiazolo[4,3a]quinoxalin-1-one, indicating that guanosine-3',5-cyclic monophosphate does not mediate this interaction, although it was partially reduced by SOD, suggesting that superoxide might contribute. The reduction of adenosine responses by electrically-induced long-term potentiation was prevented by NO synthase inhibition or SOD. The results indicate that the presynaptic effects of adenosine at presynaptic sites can be prevented by NO or superoxide but that neither of these individually can fully account for the prevention of adenosine responses by NMDA.
Collapse
Affiliation(s)
- Ali Shahraki
- Institute of Biomedical and Life Sciences, West Medical Building, University of Glasgow, Glasgow G12 8QQ, UK
| | | |
Collapse
|
34
|
Abstract
Nitric oxide (NO) is generated in central synapses on activation of N-methyl-D-aspartate (NMDA) receptors and exerts physiological effects by changing cGMP levels. NO has frequently also been claimed to engage a different mechanism, namely the covalent modification of thiol residues (S-nitrosation), and thereby exert a negative feedback on NMDA receptors. Tests of this hypothesis were conducted by recording NMDA receptor-mediated synaptic potentials in the CA1 area of rat hippocampal slices. Manipulations designed to increase or decrease endogenous NO levels had no effect. Addition of exogenous NO using a NONOate donor in concentrations up to 30-fold higher than those needed to evoke maximal cGMP accumulation also had no effect. Nevertheless, in agreement with previous findings, photolysis of a caged NO derivative with UV light led to an enduring block of synaptic NMDA receptors. To address these contradictory results, NMDA receptor-mediated currents were recorded from HEK-293 cells transfected with NR1 and NR2A subunits. As found in slices, photolysis of caged NO inhibited the currents whereas perfusion of NO (up to 5 microM) was ineffective. However, when NO was supplied at a concentration found to be effective when released photolytically (5 microM) and the cells simultaneously exposed to the UV light used for photolysis, NMDA receptor-mediated currents were inhibited. This effect was not observed at more physiological NO concentrations (10 nM range). The results indicate that neither endogenous NO nor exogenous NO in supra-physiological concentration inhibits synaptic NMDA receptors; the combination of high NO concentration and UV light can give an artifactual result.
Collapse
Affiliation(s)
- Rachel Hopper
- Wolfson Institute for Biomedical Research, University College London, Cruciform Building, Gower Street, London WC1E 6BT, United Kingdom
| | | | | |
Collapse
|
35
|
Bradshaw WT. The use of nitric oxide in neonatal care. Crit Care Nurs Clin North Am 2004; 16:249-55. [PMID: 15145370 DOI: 10.1016/j.ccell.2004.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Knowledge of NO and its role in the human body currently is limited. Further scientific research involving this unique molecule will expand its clinical usefulness. It is an exciting era in research,involving numerous body processes and systems. The initial work on pulmonary vascular response in newborns who have PPHN has opened the door to seemingly endless possibilities involving many aspects of health.
Collapse
Affiliation(s)
- Wanda T Bradshaw
- Neonatal Nurse Practitioner Progam, School of Nursing, Duke University, Durham, NC, USA.
| |
Collapse
|
36
|
Keynes RG, Duport S, Garthwaite J. Hippocampal neurons in organotypic slice culture are highly resistant to damage by endogenous and exogenous nitric oxide. Eur J Neurosci 2004; 19:1163-73. [PMID: 15016075 DOI: 10.1111/j.1460-9568.2004.03217.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Nitric oxide (NO) has been proposed to mediate neurodegeneration arising from NMDA receptor activity, but the issue remains controversial. The hypothesis was re-examined using organotypic slice cultures of rat hippocampus, with steps being taken to avoid known artefacts. The NO-cGMP signalling pathway was well preserved in such cultures. Brief exposure to NMDA resulted in a concentration-dependent delayed neuronal death that could be nullified by administration of the NMDA antagonist MK801 (10 microm) given postexposure. Two inhibitors of NO synthesis failed to protect the slices, despite fully blocking NMDA-induced cGMP accumulation. By comparing NMDA-induced cGMP accumulation with that produced by an NO donor, toxic NMDA concentrations were estimated to produce only physiological NO concentrations (2 nm). In studies of the vulnerability of the slices to exogenous NO, it was found that continuous exposure to up to 4.5 microm NO failed to affect ATP levels (measured after 6 h) or cause damage during 24 h, whereas treatment with the respiratory inhibitors myxothiazol or cyanide caused ATP depletion and complete cell death within 24 h. An NO concentration of 10 microm was required for ATP depletion and cell death, presumably through respiratory inhibition. It is concluded that sustained activity of neuronal NO synthase in intact hippocampal tissue can generate only low nanomolar NO concentrations, which are unlikely to be toxic. At the same time, the tissue is remarkably resistant to exogenous NO at up to 1000-fold higher concentrations. Together, the results seriously question the proposed role of NO in NMDA receptor-mediated excitotoxicity.
Collapse
Affiliation(s)
- Robert G Keynes
- Wolfson Institute for Biomedical Research, University College London, Cruciform Building, Gower Street, London WC1E 6BT, UK
| | | | | |
Collapse
|
37
|
Chan JYH, Chan SHH, Chang AYW. Contribution of cGMP but not peroxynitrite to negative feedback regulation of penile erection elicited by nitric oxide in the hippocampal formation of the rat. Neuropharmacology 2004; 46:126-32. [PMID: 14654104 DOI: 10.1016/s0028-3908(03)00303-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We established previously that nitric oxide (NO) in the hippocampal formation (HF) participates actively in negative feedback regulation of penile erection. This study further evaluated whether this process engaged soluble guanylyl cyclase (sGC)/cGMP cascade or peroxynitrite in the HF. Intracavernous pressure (ICP) recorded from the penis in adult, male Sprague-Dawley rats anesthetized with chloral hydrate was employed as our experimental index for penile erection. Microinjection bilaterally of a NO-independent sGC activator, YC-1 (0.1 or 1 nmol) or a cGMP analog, 8-Bromo-cGMP (0.1 or 1 nmol), into the HF elicited a significant reduction in baseline ICP. Bilateral application into the HF of equimolar doses (0.5 or 1 nmol) of a sGC inhibitor, LY83583 or a NO-sensitive sGC inhibitor, ODQ significantly antagonized the decrease in baseline ICP induced by co-administration of the NO precursor, L-arginine (5 nmol), along with significant enhancement of the magnitude of papaverine-induced elevation in ICP. In contrast, a peroxynitrite scavenger, L-cysteine (50 or 100 pmol), or an active peroxynitrite decomposition catalyst, 5,10,15,20-tetrakis-(N-methyl-4'-pyridyl)-porphyrinato iron (III) (10 or 50 pmol), was ineffective in both events. These results suggest that NO may participate in negative feedback regulation of penile erection by activating the sGC/cGMP cascade in the HF selectively.
Collapse
Affiliation(s)
- J Y H Chan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, ROC
| | | | | |
Collapse
|
38
|
Solenski NJ, Kostecki VK, Dovey S, Periasamy A. Nitric-oxide-induced depolarization of neuronal mitochondria: implications for neuronal cell death. Mol Cell Neurosci 2003; 24:1151-69. [PMID: 14697675 DOI: 10.1016/j.mcn.2003.08.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Nitric oxide (NO(*)) has known toxic effects on central nervous system neurons. This study characterized the effect of NO(*) on mitochondrial membrane changes by exploring the relationship among NO(*), excitatory receptor activation, and the induction of peroxynitrite, a highly toxic NO(*) reactant, to neuronal injury. Cultured rat cortical neurons were exposed to the NO(*) generator, diethylenetriamine/nitric oxide adduct, and were examined for signs of cell death, mitochondrial membrane potential changes (Deltapsi(m)), and the induction of a mitochondrial permeability transition (MPT). Neurons were also examined for nitrotyrosine (NT) immunoreactivity, a marker of reactive nitrogen species (RNS) formation. Neurons exposed to NO(*) or to N-methyl-D-aspartate (NMDA) exhibited similar rapid depolarization of mitochondria, which was prevented by an NMDA receptor antagonist. Electrophysiological studies demonstrated NO(*) potentiation of NMDA-induced NMDA receptor currents. NO(*) and NMDA-treated neurons had evidence of mitochondrial-specific NT immunoreactivity that was prevented by a SOD/catalase mimetic (EUK-134). EUK-134 treatment reduced both NO(*) and NMDA-induced NT formation and neuronal cell death. EUK-134 did not prevent NO-induced Deltapsi(m) but partially prevented NMDA-induced Deltapsi(m) loss. Although NO(*) and NMDA both induced MPT and MPT inhibitors prevented NO-induced Deltapsi(m), they did not result in significant neuroprotection, in contrast to treatment designed to decrease peroxynitrite formation. These data suggest that NO-induced NMDA receptor activation is closely linked to intramitochondrial NO-peroxynitrite/RNS formation and thereby acts as a major mediator of neuronal cell death.
Collapse
Affiliation(s)
- Nina J Solenski
- Department of Neurology, University of Virginia Health Sciences System, Charlottesville, VA 22908, USA.
| | | | | | | |
Collapse
|
39
|
Abstract
Induction of long-term potentiation and application of short periods of anoxia/hypoglycemia result in the growth of dendritic filopodia and formation of new spines. Here we investigated whether these conditions also affected the morphology of presynaptic structures. Using confocal imaging of DiI-labeled axons, electron microscopy, and stereological analyses, we show that short anoxia/hypoglycemia and theta burst stimulation induced rapid, calcium-dependent growth of presynaptic filopodia-like protrusions and remodeling of presynaptic varicosities. Three-dimensional reconstruction of axonal outgrowths revealed that, within 30 min, they made contacts and triggered the formation of a postsynaptic density on the target cell. Interestingly, these axonal filopodia first established synapses with the dendritic shaft and later mostly with spines. They also contributed to the formation of multi-innervated spines. Because these presynaptic growth mechanisms depended on NMDA receptor activation, we investigated whether a diffusing messenger could be involved. We found that blockade of nitric oxide synthase prevented these changes, and conversely, a nitric oxide donor could reproduce them. A model is presented that proposes that activation of NMDA receptors and subsequent release of nitric oxide could trigger the growth of presynaptic filopodia, which, in turn, play an active role in synaptogenesis and spine formation.
Collapse
|
40
|
Long-term depression of presynaptic release from the readily releasable vesicle pool induced by NMDA receptor-dependent retrograde nitric oxide. J Neurosci 2003. [PMID: 12843298 DOI: 10.1523/jneurosci.23-13-05936.2003] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Postsynaptic alterations are currently believed to be able to fully account for NMDA-receptor-dependent long-term depression (LTD) and long-term potentiation of synaptic strength, although there is also evidence supporting changes in presynaptic release. Using dualphoton laser scan microscopy of N-(3-triethylammoniumpropyl)-4-(4-(dibutylamino)styryl) pyridinium dibromide (FM1-43) to directly visualize presynaptic vesicular release at Schaffer collateral-CA1 excitatory synapses in hippocampal slices, we demonstrate reduced vesicular release associated with LTD. Selective loading, by hypertonic shock, of the readily releasable vesicle pool (RRP) showed that LTD of release is a selective modification of release from the RRP. Presynaptic LTD of RRP release required activation of NMDA receptors, production and extracellular diffusion of the intercellular messenger NO, and activation of cGMP-dependent protein kinase.
Collapse
|
41
|
Huang CC, Chan SHH, Hsu KS. cGMP/protein kinase G-dependent potentiation of glutamatergic transmission induced by nitric oxide in immature rat rostral ventrolateral medulla neurons in vitro. Mol Pharmacol 2003; 64:521-32. [PMID: 12869658 DOI: 10.1124/mol.64.2.521] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although both nitric oxide (NO) and glutamate within the rostral ventrolateral medulla (RVLM) are important mediators of the central cardiovascular regulation, little is known about the functional interactions between these two mediators. Herein, we investigated the possible role of NO on the glutamatergic transmission of RVLM neurons. Whole-cell patch-clamp recordings were performed on visualized RVLM neurons in the brainstem slice preparation of rats. We found that bath application of l-arginine, the substrate for NO production, significantly increased the amplitude of excitatory postsynaptic currents (EPSCs). This enhancement was completely abolished by coadministration of the NO synthase inhibitor 7-nitroindazole and mimicked by the NO donors 3-morpholinylsydnoneimine and spermine NONOate. Bath application of a NO-sensitive guanylyl cyclase inhibitor, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or a protein kinase G (PKG) inhibitor, Rp-8-bromo-guanosine 3',5'-cyclic monophosphorothioate, fully prevented the l-arginine-, 3-morpholinylsydnoneimine-, and N-[4-[1-(3-aminopropyl)-2-hydroxy-2-nitrosohydrazino]-butyl]-1,3-propanediamin (spermine NONOate)-induced synaptic potentiation. Direct activation of PKG with 8-(4-chlorophenylthio)-cGMP mimicked the action of NO donors. Furthermore, the augmentation by spermine NONOate of EPSC was accompanied by a reduction of the paired-pulse facilitation and synaptic failure rate of EPSCs. Spermine NONOate also significantly increased the frequency of both spontaneous and miniature EPSCs without altering their amplitude distribution. Pretreatment with the N-type Ca2+ channel blocker omega-conotoxin GVIA selectively blocked the spermine NONOate-induced synaptic potentiation. These results suggest that NO acts presynaptically to elicit a synaptic potentiation on the RVLM neurons through an enhancement of presynaptic N-type Ca2+ channel activity leading to facilitating glutamate release. The presynaptic action of NO is mediated by a cGMP/PKG-coupled signaling pathway.
Collapse
Affiliation(s)
- Chiung-Chun Huang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, 1, Ta-Hsiue Rd., Tainan 701, Taiwan
| | | | | |
Collapse
|
42
|
Islam ATMS, Kuraoka A, Kawabuchi M. Morphological basis of nitric oxide production and its correlation with the polysialylated precursor cells in the dentate gyrus of the adult guinea pig hippocampus. Anat Sci Int 2003; 78:98-103. [PMID: 12828422 DOI: 10.1046/j.0022-7722.2003.00045.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neurogenesis in the hippocampus persist throughout life and precursors of neurons reside in the granule cell layer of the dentate gyrus. Until now, the role of nitric oxide (NO) in the phenomenon has been unclear. By using specific antibodies and a confocal laser scanning microscope, the localization of NO synthase (NOS) was examined in the dentate gyrus of the adult guinea pig in relation with the neuronal precursor marker highly polysialylated neural cell adhesion molecule (PSA-N-CAM). Observation of single immunolabeled sections has revealed that both the PSA-N-CAM- and most NOS-positive cells were localized in the granule cell layer of the dentate gyrus. The former were small in size and showed a punctate, clustered immunoreaction with an irregular cellular margin, whereas the latter showed somewhat diverse cellular profiles. Some NOS-positive neurons had elliptical-like morphology with elongated dendrites, whereas others were small, irregularly shaped and mostly lacking dendritic spines. Double immunolabeling has revealed that NOS-immunoreactivity intermingled, as well as colocalized, with that of PSA-N-CAM, particulary in the granule cell layer. The doubly stained cells were morphologically indistinguishable from PSA-N-CAM single positive cells. These results not only suggest the role of NO production in adult hippocampal neurogenesis, but also indicate that some PSA-N-CAM-expressing neuronal precursors produce NO.
Collapse
Affiliation(s)
- A T M Shariful Islam
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University Fukuoka, Japan
| | | | | |
Collapse
|
43
|
Abstract
Nitric oxide (NO) functions in several types of synaptic plasticity, including hippocampal long-term potentiation (LTP), in which it may serve as a retrograde messenger after postsynaptic NMDA receptor activation. In accordance with a prediction of this hypothesis, and with previous findings using guinea pig tissue, exogenous NO, when paired with a short tetanus (ST) to afferent fibers, generated a stable NMDA receptor-independent potentiation of rat CA1 hippocampal synaptic transmission that occluded LTP. Contrary to predictions, however, the pairing-induced potentiation was abolished in the presence of NO synthase inhibitors, indicating that endogenous NO is required for exogenous NO to facilitate LTP. Periodic application of NO while endogenous NO synthesis was blocked indicated that a tonic low level is necessary on both sides of the NO-ST pairing for the plasticity to occur. A similar dependence on tonic NO seems to extend to LTP, because application of an NO synthase inhibitor 5 min after tetanic stimulation blocked LTP as effectively as adding it beforehand. The posttetanus time window during which NO operated was restricted to <15 min. Inhibition of the guanylyl cyclase-coupled NO receptor indicated that the potentiation resulting from NO-ST pairing and the NO signal transduction pathway during early LTP are both through cGMP. We conclude that NO does not function simply as an acute signaling molecule in LTP induction but has an equally important role outside this phase. The results resonate with observations concerning the role of the hippocampal NO-cGMP pathway in certain types of learning behavior.
Collapse
|
44
|
Bon CLM, Garthwaite J. Adenosine acting on A1 receptors protects NO-triggered rebound potentiation and LTP in rat hippocampal slices. J Neurophysiol 2002; 87:1781-9. [PMID: 11929899 DOI: 10.1152/jn.00630.2001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exposure of hippocampal slices to nitric oxide (NO) results in a depression of CA1 synaptic transmission. Under 0.2-Hz stimulation, washout of NO leads to a persistent potentiation that depends on N-methyl-D-aspartate (NMDA) receptors and endogenous NO formation and that occludes tetanus-induced long-term potentiation (LTP). The experiments were initially aimed at determining the relationship between the NO-induced synaptic depression and rebound potentiation. The adenosine A1 antagonist, 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) partially inhibited the depression produced by the NO donor diethylamine NONOate (300 microM). It also led to a complete block of both the rebound potentiation and the subsequent tetanus-induced LTP. LTP was preserved in the presence of DPCPX if the stimulation frequency was reduced to 0.033 Hz or if the NO application was omitted. The NO-triggered rebound potentiation was restored if the experiment (DPCPX followed by exogenous NO) was conducted in the presence of an NMDA antagonist. The restored potentiation was completely blocked by the NO synthase inhibitor, L-nitroarginine. It is concluded that the NO-induced depression is partially mediated by increased release of endogenous adenosine acting on A1 receptors. Moreover, tonic A1 receptor activation by adenosine protects LTP and the rebound potentiation from being disabled by untimely NMDA receptor activity. Hence, the NO-induced depression and rebound potentiation are linked in the sense that the depression helps to preserve the capacity of the synapses to undergo potentiation. Finally, the results give the first example of exogenous NO eliciting an enduring potentiation of hippocampal synaptic transmission that is dependent on endogenous NO formation, but not on NMDA receptors.
Collapse
Affiliation(s)
- Christelle L M Bon
- The Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, United Kingdom
| | | |
Collapse
|