1001
|
Wilson RI, Gödecke A, Brown RE, Schrader J, Haas HL. Mice deficient in endothelial nitric oxide synthase exhibit a selective deficit in hippocampal long-term potentiation. Neuroscience 1999; 90:1157-65. [PMID: 10338286 DOI: 10.1016/s0306-4522(98)00479-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Long-term potentiation, a persistent increase in synaptic efficacy, may require a retrograde signal originating in the postsynaptic cell that induces an increase in presynaptic neurotransmitter release. We have constructed a mouse homozygous for a targeted null mutation in the endothelial isoform of nitric oxide synthase and report that long-term potentiation in the CA1 region of these mice is entirely absent under weak stimulation conditions. Application of a membrane-permeant guanosine-3',5'-cyclic monophosphate analogue during tetanus fails to compensate for this deficit, suggesting that nitric oxide produced by endothelial nitric oxide synthase may affect long-term potentiation through a cascade that does not include guanylyl cyclase. We also report that strong tetanic stimulation can induce robust long-term potentiation in these mice which is not blocked by pharmacological inhibitors of nitric oxide synthase. Furthermore, mice lacking endothelial nitric oxide synthase show no shift in the frequency-response curve for the induction of long-term potentiation. Basal synaptic transmission, paired-pulse facilitation and the electrical properties of CA1 cells in these mice were similar to controls. These results support a selective role for endothelial nitric oxide synthase in long-term potentiation, but also demonstrate that nitric oxide synthase is not involved in this process under all conditions.
Collapse
Affiliation(s)
- R I Wilson
- Institut für Neurophysiologie, Heinrich-Heine-Universität, Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
1002
|
Facchinetti F, Sasaki M, Cutting FB, Zhai P, MacDonald JE, Reif D, Beal MF, Huang PL, Dawson TM, Gurney ME, Dawson VL. Lack of involvement of neuronal nitric oxide synthase in the pathogenesis of a transgenic mouse model of familial amyotrophic lateral sclerosis. Neuroscience 1999; 90:1483-92. [PMID: 10338314 DOI: 10.1016/s0306-4522(98)00492-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A subset of familial cases of amyotrophic lateral sclerosis are linked to missense mutations in copper/zinc superoxide dismutase type 1. Patients with missense mutations in copper/zinc superoxide dismutase type 1 develop a paralytic disease indistinguishable from sporadic amyotrophic lateral sclerosis through an unknown toxic gain of function. Nitric oxide reacts with the superoxide anion to form the strong oxidant, peroxynitrite, which participates in neuronal injury in a variety of model systems. Peroxynitrite is an alternate substrate for copper/zinc superoxide dismutase type 1, causing catalytic nitration of tyrosine residues in other proteins. Mutations in copper/zinc superoxide dismutase type 1 may disrupt the active site of the enzyme and permit greater access of peroxynitrite to copper, leading to increased nitration by peroxynitrite of critical cellular targets. To investigate whether neuronal-derived nitric oxide plays a role in the pathogenesis of familial amyotrophic lateral sclerosis, we examined the effects of three different nitric oxide synthase inhibitors: a non-selective nitric oxide synthase inhibitor, nitro-L-arginine methyl ester; a relatively selective inhibitor of neuronal nitric oxide synthase, 7-nitroindazole; and a novel highly selective neuronal nitric oxide synthase inhibitor, AR-R 17,477, in transgenic mice expressing a familial amyotrophic lateral sclerosis-linked mutant human copper/zinc superoxide dismutase type 1 (Gly-->Ala at position 93; G93A) containing a high transgene copy number and a low transgene copy number. AR-R 17,477, but not nitro-L-arginine methyl ester or 7-nitroindazole, significantly prolonged survival in both the high and low transgene transgenic mice. To determine whether neuronal nitric oxide synthase is involved in the pathogenesis resulting from the familial amyotrophic lateral sclerosis copper/zinc superoxide dismutase type 1 mutation, we produced mice with the copper/zinc superoxide dismutase type 1 mutation which lack the neuronal nitric oxide synthase gene. The transgenic mice expressing a familial amyotrophic lateral sclerosis-linked mutant human copper/zinc superoxide dismutase type 1 on neuronal nitric oxide synthase null background do not live significantly longer than transgenic mice expressing a familial amyotrophic lateral sclerosis-linked mutant human copper/zinc superoxide dismutase type 1. Western blot analysis indicates the presence of two neuronal nitric oxide synthase-like immunoreactive bands in spinal cord homogenates of the neuronal nitric oxide synthase null mice, and residual neuronal nitric oxide synthase catalytic activity ( > 7%) is detected in the spinal cord of the transgenic mice expressing a familial amyotrophic lateral sclerosis-linked mutant human copper/zinc superoxide dismutase type 1 on neuronal nitric oxide synthase null background. This amount of residual activity probably does not account for lack of protection afforded by the disrupted neuronal nitric oxide synthase gene in the familial amyotrophic lateral sclerosis-linked mutant human copper/zinc superoxide dismutase type 1 mice. Immunological nitric oxide synthase is not detected in the copper/zinc superoxide dismutase type 1 mutant mice at several different ages, thus excluding immunological nitric oxide synthase as a contributor to the pathogenesis of familial amyotrophic lateral sclerosis. Levels of neuronal nitric oxide synthase as well as Ca2+-dependent nitric oxide synthase catalytic activity in the copper/zinc superoxide dismutase type 1 mutant mice do not differ from wild type mice. Endothelial nitric oxide synthase levels may be decreased in the copper/zinc superoxide dismutase type 1 mutant mice. Together, these results do not support a significant role for neuronal-derived nitric oxide in the pathogenesis of familial amyotrophic lateral sclerosis transgenic mice.
Collapse
Affiliation(s)
- F Facchinetti
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1003
|
Hobbs AJ, Higgs A, Moncada S. Inhibition of nitric oxide synthase as a potential therapeutic target. Annu Rev Pharmacol Toxicol 1999; 39:191-220. [PMID: 10331082 DOI: 10.1146/annurev.pharmtox.39.1.191] [Citation(s) in RCA: 442] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nitric oxide (NO) regulates numerous physiological processes, including neurotransmission, smooth muscle contractility, platelet reactivity, and the cytotoxic activity of immune cells. Because of the ubiquitous nature of NO, inappropriate release of this mediator has been linked to the pathogenesis of a number of disease states. This provides the rationale for the design of therapies that modulate NO concentrations selectively. A well-characterized family of compounds are the inhibitors of NO synthase, the enzyme responsible for the generation of NO; such agents are potentially beneficial in the treatment of conditions associated with an overproduction of NO, including septic shock, neurodegenerative disorders, and inflammation. This article provides an overview of NO synthase inhibitors, focusing on agents that prevent binding of substrate L-arginine.
Collapse
Affiliation(s)
- A J Hobbs
- Wolfson Institute for Biomedical Research, University College London, Rayne Institute, United Kingdom.
| | | | | |
Collapse
|
1004
|
Eu JP, Xu L, Stamler JS, Meissner G. Regulation of ryanodine receptors by reactive nitrogen species. Biochem Pharmacol 1999; 57:1079-84. [PMID: 11230795 DOI: 10.1016/s0006-2952(98)00360-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The ryanodine receptors (RyRs) are large intracellular calcium release channels that play an important role in the control of the calcium levels in excitable and non-excitable cells. Many endogenous modulators such as Mg2+, ATP, or calmodulin can affect the channel activities of the three known mammalian RyR isoforms. RyRs also are known to be redox-responsive. However, the molecular basis and the physiological relevance of redox modulation of RyRs are unclear. Recent evidence suggests that nitric oxide (NO) and related molecules may be endogenous regulators of the skeletal and cardiac muscle RyRs. The two tissues express nitric oxide synthases (NOSs), and NO or NO-related species have been shown to affect Ca2+ release channel activities directly via covalent modifications of thiol groups. Both an oxidative and a nitrosative modification of RyRs have been described, leading to either a reversible or irreversible alteration of RyR ion channel activity. Additional mechanisms of regulation may include cyclic GMP-dependent signaling pathways and NO modification of RyR regulatory proteins such as the surface membrane L-type Ca2+ channel. Modification of RyRs by NO may influence a variety of physiological functions such as insulin release, vasomotor control, and muscle contraction.
Collapse
Affiliation(s)
- J P Eu
- Howard Hughes Medical Institute, Department of Medicine, Divisions of Pulmonary and Cardiovascular Medicine, and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
1005
|
Wang LH, Kalb RG, Strittmatter SM. A PDZ protein regulates the distribution of the transmembrane semaphorin, M-SemF. J Biol Chem 1999; 274:14137-46. [PMID: 10318831 DOI: 10.1074/jbc.274.20.14137] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
M-SemF is a membrane-associated, neurally enriched member of the semaphorin family of axon guidance signals. We considered whether the cytoplasmic domain of M-SemF might possess a signaling function and/or might control the distribution of M-SemF on the cell surface. We identify a PDZ-containing neural protein as an M-SemF cytoplasmic domain-associated protein (SEMCAP-1). SEMCAP-2 is a closely related nonneuronal protein. SEMCAP-1 has recently also been identified as GIPC, by virtue of its interaction with the RGS protein GAIP in vitro (De Vries, L., Lou, X., Zhao, G., Zheng, B., and Farquhar, M. G. (1998) Proc. Natl. Acad. Sci. U. S. A. 95, 12340-12345). Expression studies support the notion that SEMCAP-1(GIPC) interacts with M-SemF, but not GAIP, in brain. Lung SEMCAP-2 and SEMCAP-1(GIPC) are potential partners for both GAIP and M-SemF. The protein interaction requires the single PDZ domain of SEMCAP-1(GIPC) and the carboxyl-terminal four residues of M-SemF, ESSV. While SEMCAP-1(GIPC) also interacts with SemC, it does not interact with other proteins containing a class I PDZ binding motif, nor does M-SemF interact with other class I PDZ proteins. Co-expression of SEMCAP-1(GIPC) induces the redistribution of dispersed M-SemF into detergent-resistant aggregates in HEK293 cells. Thus, SEMCAP-1(GIPC) appears to regulate the subcellular distribution of M-SemF in brain, and SEMCAPs could link M-SemF to G protein signal transduction pathways.
Collapse
Affiliation(s)
- L H Wang
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | |
Collapse
|
1006
|
Abstract
Platelets are small cells, 1/14th the volume of erythrocytes, and about 1000 billion circulate in human blood as smooth anucleate disks. Their job is to survey the lining of our blood vessels, the endothelium. In acute damage and extravasation, platelets are activated by contact with exposed collagen and aggregate together at the wound sites to initiate clotting and stop bleeding. Forming a physical plug to seal a hemorrhaging vessel is the key role of blood platelets. However, milder injury to the endothelium, perhaps a result of high blood pressure, raised plasma cholesterol, or smoking, also causes platelets to adhere to the internal walls of arteries. Such precipitate adhesion and activation of platelets initiates an inflammatory response of the vessel wall and predisposes to vascular complications, including thrombosis, premature heart disease, myocardial infarcts or strokes, and diabetes. It is essential, therefore, that during normal vascular hemostasis platelet activation is tightly controlled. Indeed, both platelets and endothelial cells produce and secrete chemicals that directly inhibit platelet aggregation. A key agent is the free radical gas nitric oxide (NO). Here, we review how this 30-Da molecular messenger is synthesized by a catalytic cassette 10,000 times larger and how it functions to suppress platelet "stickiness." We also present new evidence that directly links plasma lipoproteins with platelet activation: we describe at the molecular level how apoE, a protein with a prominent role in cholesterol transport, interacts with the platelet surface to stimulate NO production and hence attenuate platelet activation.
Collapse
Affiliation(s)
- D R Riddell
- Department of Medicine, Royal Free and University College Medical School, London, England
| | | |
Collapse
|
1007
|
Abstract
The nitric oxide (NO) synthase family of enzymes generate NO from L-arginine, which acts as a biologic effector molecule in a broad number of settings. This report summarizes some of the current information regarding NO synthase structure-function, reaction mechanism, control of catalysis, and protein interactions.
Collapse
Affiliation(s)
- D J Stuehr
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| |
Collapse
|
1008
|
Abstract
Recent studies have begun to characterize the nitric oxide/cyclic GMP/protein kinase G pathway in the mammalian cochlea by demonstrating the presence of both the enzyme that produces nitric oxide (NO), nitric oxide synthase, and the NO receptor, soluble guanylate cyclase. The present study investigated protein kinase G (cyclic GMP-dependent protein kinase-I, cGK-I), the downstream enzyme of this pathway that frequently mediates its physiological effects. A commercial antibody to a human cGK-I sequence recognized a protein of appropriate molecular weight in Western blots of guinea pig aorta. Immunostaining of guinea pig aorta was consistent with the expected distribution of cGK-I. In lateral wall tissues of the cochlea, pericytes lining the blood vessels of the spiral ligament were strongly immunoreactive. In the organ of Corti, cGK-I was detected in Hensen's, Deiters', and pillar cells, but not in inner and outer hair cells. This distribution coincides with the localization of soluble guanylate cyclase activity and suggests that cGK-I mediates the effects of the NO/cyclic GMP pathway in the cochlea. It reinforces the hypothesis that the NO/cyclic GMP/cGK-I pathway is involved in regulation of cochlear blood flow and supporting cell physiology.
Collapse
Affiliation(s)
- F Tian
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor 48109-0506, USA
| | | | | |
Collapse
|
1009
|
Xiong H, Yamada K, Han D, Nabeshima T, Enikolopov G, Carnahan J, Nawa H. Mutual regulation between the intercellular messengers nitric oxide and brain-derived neurotrophic factor in rodent neocortical neurons. Eur J Neurosci 1999; 11:1567-76. [PMID: 10215909 DOI: 10.1046/j.1460-9568.1999.00567.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The diffusible factors, nitric oxide (NO) and brain-derived neurotrophic factor (BDNF) are both suggested to be intercellular messengers that have similar synaptic activities and developmental influences in the brain. In the present study, we have analysed their mutual regulation with respect to their production in rodent neocortical neurons. Some of the cultured rat neocortical neurons exhibited immunoreactivity for both neuronal NO synthase (NOS) and the BDNF receptor trkB. Neuronal NOS appeared to be activated autonomously and produced NO in culture as monitored by nitrite accumulation. Inhibition of the endogenous NO production in culture by a NOS inhibitor, NG-monomethyl-L-arginine (NMMA), enhanced basal expression of BDNF mRNA and protein. Similarly, cerebroventricular administration of another NOS inhibitor, N-omega-nitro-L-arginine methylester (L-NAME), but not D-NAME or saline, increased BDNF content in the neocortex. In the opposite direction, however, BDNF appeared to function as a positive regulator for NO synthesis. Addition of BDNF upregulated the neuronal NOS expression as well as NO production in neocortical culture. In agreement, BDNF knock-out mice exhibited significant impairment of neuronal NOS expression in the neocortex. Taken together, these observations suggest that the trans-synaptic signalling molecules, NO and BDNF, influence the production of each other and mutually regulate the strength of their intercellular communications.
Collapse
Affiliation(s)
- H Xiong
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | | | | | | | | | | | | |
Collapse
|
1010
|
Hasegawa M, Cuenda A, Spillantini MG, Thomas GM, Buée-Scherrer V, Cohen P, Goedert M. Stress-activated protein kinase-3 interacts with the PDZ domain of alpha1-syntrophin. A mechanism for specific substrate recognition. J Biol Chem 1999; 274:12626-31. [PMID: 10212242 DOI: 10.1074/jbc.274.18.12626] [Citation(s) in RCA: 139] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mechanisms for selective targeting to unique subcellular sites play an important role in determining the substrate specificities of protein kinases. Here we show that stress-activated protein kinase-3 (SAPK3, also called ERK6 and p38gamma), a member of the mitogen-activated protein kinase family that is abundantly expressed in skeletal muscle, binds through its carboxyl-terminal sequence -KETXL to the PDZ domain of alpha1-syntrophin. SAPK3 phosphorylates alpha1-syntrophin at serine residues 193 and 201 in vitro and phosphorylation is dependent on binding to the PDZ domain of alpha1-syntrophin. In skeletal muscle SAPK3 and alpha1-syntrophin co-localize at the neuromuscular junction, and both proteins can be co-immunoprecipitated from transfected COS cell lysates. Phosphorylation of a PDZ domain-containing protein by an associated protein kinase is a novel mechanism for determining both the localization and the substrate specificity of a protein kinase.
Collapse
Affiliation(s)
- M Hasegawa
- Medical Research Council Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
1011
|
Abstract
The synapse is a specialized cellular junction with an elaborate and highly evolved capacity for signal transduction. At excitatory synapses, the neurotransmitter glutamate is released from the presynaptic nerve terminal and stimulates several types of glutamate receptors in the postsynaptic membrane. These include the ionotropic receptors, which are glutamate-gated cation channels, and the metabotropic receptors, which are G protein-coupled seven-transmembrane receptors. The ionotropic glutamate receptors have received special attention because of growing evidence that changes in their synaptic abundance, posttranslational modification, or molecular interactions can provide long-term changes in synaptic strength. This review summarizes new information about the ionotropic glutamate receptors and relates receptor function to the organization of the postsynaptic membrane and the regulation of electrophysiologic and biochemical signaling at the synapse.
Collapse
Affiliation(s)
- E B Ziff
- Howard Hughes Medical Institute, Department of Biochemistry, New York University Medical Center, New York 10016, USA.
| |
Collapse
|
1012
|
Sheng M, Pak DT. Glutamate receptor anchoring proteins and the molecular organization of excitatory synapses. Ann N Y Acad Sci 1999; 868:483-93. [PMID: 10414325 DOI: 10.1111/j.1749-6632.1999.tb11317.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ionotropic glutamate receptors are concentrated at postsynaptic sites in excitatory synapses. The cytoplasmic C-terminal tail of certain glutamate receptor subunits interact with specific PDZ domain-containing proteins. NMDA receptor NR2 subunits bind to the PSD-95 family of proteins, whereas AMPA receptor subunits GluR2/3 bind to GRIP. These interactions may underlie the clustering, targeting, and immobilization of the glutamate receptors at postsynaptic sites. By virtue of their multiple protein-binding domains (e.g., three PDZs in PSD-95 and seven PDZs in GRIP), PSD-95 and GRIP can function as multivalent proteins that organize a specific cytoskeletal and signaling complex associated with each class of glutamate receptor. The network of protein-protein interactions mediated by these abundant PDZ proteins is likely to contribute significantly to the molecular scaffold of the postsynaptic density.
Collapse
Affiliation(s)
- M Sheng
- Howard Hughes Medical Institute, Massachussets General Hospital, Boston 02114, USA.
| | | |
Collapse
|
1013
|
Dong H, Zhang P, Liao D, Huganir RL. Characterization, expression, and distribution of GRIP protein. Ann N Y Acad Sci 1999; 868:535-40. [PMID: 10414331 DOI: 10.1111/j.1749-6632.1999.tb11323.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- H Dong
- Howard Hughes Medical Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| | | | | | | |
Collapse
|
1014
|
Hillier BJ, Christopherson KS, Prehoda KE, Bredt DS, Lim WA. Unexpected Modes of PDZ Domain Scaffolding Revealed by Structure of nNOS-Syntrophin Complex. Science 1999. [DOI: 10.1126/science.284.5415.812] [Citation(s) in RCA: 420] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The PDZ protein interaction domain of neuronal nitric oxide synthase (nNOS) can heterodimerize with the PDZ domains of postsynaptic density protein 95 and syntrophin through interactions that are not mediated by recognition of a typical carboxyl-terminal motif. The nNOS-syntrophin PDZ complex structure revealed that the domains interact in an unusual linear head-to-tail arrangement. The nNOS PDZ domain has two opposite interaction surfaces—one face has the canonical peptide binding groove, whereas the other has a β-hairpin “finger.” This nNOS β finger docks in the syntrophin peptide binding groove, mimicking a peptide ligand, except that a sharp β turn replaces the normally required carboxyl terminus. This structure explains how PDZ domains can participate in diverse interaction modes to assemble protein networks.
Collapse
Affiliation(s)
- Brian J. Hillier
- Department of Cellular and Molecular Pharmacology and Department of Biochemistry and Biophysics,
| | - Karen S. Christopherson
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kenneth E. Prehoda
- Department of Cellular and Molecular Pharmacology and Department of Biochemistry and Biophysics,
| | - David S. Bredt
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Wendell A. Lim
- Department of Cellular and Molecular Pharmacology and Department of Biochemistry and Biophysics,
| |
Collapse
|
1015
|
Yan H, Tsai MD. Nucleoside monophosphate kinases: structure, mechanism, and substrate specificity. ADVANCES IN ENZYMOLOGY AND RELATED AREAS OF MOLECULAR BIOLOGY 1999; 73:103-34, x. [PMID: 10218107 DOI: 10.1002/9780470123195.ch4] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
The catalytic mechanisms of adenylate kinase, guanylate kinase, uridylate kinase, and cytidylate kinase are reviewed in terms of kinetic and structural information that has been obtained in recent years. All four kinases share a highly related tertiary structure, characterized by a central five-stranded parallel beta-sheet with helices on both sides, as well as the three regions designated as the CORE, NMPbind, and LID domains. The catalytic mechanism continues to be refined to higher levels of resolution by iterative structure-function studies, and the strengths and limitations of site-directed mutagenesis are well illustrated in the case of adenylate kinase. The identity and roles of active site residues now appear to be resolved, and this review describes how specific site substitutions with unnatural amino acid side-chains have proven to be a major advance. Likewise, there is mounting evidence that phosphoryl transfer occurs by an associative transition state, based on (a) the stereochemical course of phosphoryl transfer, (b) geometric considerations, (c) examination of likely electronic distributions, (d) the orientation of the phosphoryl acceptor relative to the phosphoryl being transferred, (e) the most likely role of magnesium ion, (f) the lack of restricted access of solvent water, and (g) the results of oxygen-18 kinetic isotope. effect experiments.
Collapse
Affiliation(s)
- H Yan
- Department of Biochemistry, Michigan State University, East Lansing 48824, USA
| | | |
Collapse
|
1016
|
Hashida-Okumura A, Okumura N, Iwamatsu A, Buijs RM, Romijn HJ, Nagai K. Interaction of neuronal nitric-oxide synthase with alpha1-syntrophin in rat brain. J Biol Chem 1999; 274:11736-41. [PMID: 10206989 DOI: 10.1074/jbc.274.17.11736] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuronal nitric-oxide synthase (nNOS) has a PSD-95/Dlg/ZO-1 (PDZ) domain that can interact with multiple proteins. nNOS has been known to interact with PSD-95 and a related protein, PSD-93, in brain and with alpha1-syntrophin in skeletal muscle in mammals. In this study, we have purified an nNOS-interacting protein from bovine brain using an affinity column made of Sepharose conjugated with glutathione S-transferase-rat nNOS fusion protein and identified it as alpha1-syntrophin by microsequencing. Immunostaining of primary cultures of rat embryonic brain neuronal cells with antibodies against these proteins showed that nNOS and alpha1-syntrophin were colocalized in neuronal cell bodies and neurites. Immunohistochemical analysis indicated that the nNOS- and alpha1-syntrophin-like immunoreactive substances were highly expressed in the rat hypothalamic suprachiasmatic nucleus (SCN) and paraventricular nucleus. In the SCN, nNOS- and alpha1-syntrophin-like immunoreactive substances were colocalized in the same neurons as detected by confocal microscopy. These results indicate that nNOS in brain interacts with alpha1-syntrophin in specific neurons of the SCN and paraventricular nucleus and that this interaction might play a physiological role in functions of these neurons.
Collapse
Affiliation(s)
- A Hashida-Okumura
- Division of Protein Metabolism, Institute for Protein Research, Osaka University, 3-2 Yamada-Oka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | |
Collapse
|
1017
|
Yao I, Hata Y, Ide N, Hirao K, Deguchi M, Nishioka H, Mizoguchi A, Takai Y. MAGUIN, a novel neuronal membrane-associated guanylate kinase-interacting protein. J Biol Chem 1999; 274:11889-96. [PMID: 10207009 DOI: 10.1074/jbc.274.17.11889] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Postsynaptic density (PSD)-95/Synapse-associated protein (SAP) 90 and synaptic scaffolding molecule (S-SCAM) are neuronal membrane-associated guanylate kinases. Because PSD-95/SAP90 and S-SCAM function as synaptic scaffolding proteins, identification of ligands for these proteins is important to elucidate the structure of synaptic junctions. Here, we report a novel protein interacting with the PDZ domains of PSD-95/SAP90 and S-SCAM and named it MAGUIN-1 (membrane-associated guanylate kinase-interacting protein-1). MAGUIN-1 has one sterile alpha motif, one PDZ, and one plekstrin homology domain. MAGUIN-1 is localized at the plasma membrane via the plekstrin homology domain and the C-terminal region and interacts with PSD-95/SAP90 and S-SCAM via a C-terminal PDZ domain-binding motif. MAGUIN-1 has a short isoform, MAGUIN-2, which lacks a PDZ domain-binding motif. MAGUINs are expressed in neurons and localized in the cell body and neurites and are coimmunoprecipitated with PSD-95/SAP90 and S-SCAM from rat crude synaptosome. MAGUIN-1 may play an important role with PSD-95/SAP90 and S-SCAM to assemble the components of synaptic junctions.
Collapse
Affiliation(s)
- I Yao
- Takai Biotimer Project, Exploratory Research for Advanced Technology, Japan Science and Technology Corporation, c/o JCR Pharmaceuticals Co. Ltd., 2-2-10 Murotani, Nishi-ku, Kobe 651-2241, Japan
| | | | | | | | | | | | | | | |
Collapse
|
1018
|
Kachinsky AM, Froehner SC, Milgram SL. A PDZ-containing scaffold related to the dystrophin complex at the basolateral membrane of epithelial cells. J Biophys Biochem Cytol 1999; 145:391-402. [PMID: 10209032 PMCID: PMC2133114 DOI: 10.1083/jcb.145.2.391] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Membrane scaffolding complexes are key features of many cell types, serving as specialized links between the extracellular matrix and the actin cytoskeleton. An important scaffold in skeletal muscle is the dystrophin-associated protein complex. One of the proteins bound directly to dystrophin is syntrophin, a modular protein comprised entirely of interaction motifs, including PDZ (protein domain named for PSD-95, discs large, ZO-1) and pleckstrin homology (PH) domains. In skeletal muscle, the syntrophin PDZ domain recruits sodium channels and signaling molecules, such as neuronal nitric oxide synthase, to the dystrophin complex. In epithelia, we identified a variation of the dystrophin complex, in which syntrophin, and the dystrophin homologues, utrophin and dystrobrevin, are restricted to the basolateral membrane. We used exogenously expressed green fluorescent protein (GFP)-tagged fusion proteins to determine which domains of syntrophin are responsible for its polarized localization. GFP-tagged full-length syntrophin targeted to the basolateral membrane, but individual domains remained in the cytoplasm. In contrast, the second PH domain tandemly linked to a highly conserved, COOH-terminal region was sufficient for basolateral membrane targeting and association with utrophin. The results suggest an interaction between syntrophin and utrophin that leaves the PDZ domain of syntrophin available to recruit additional proteins to the epithelial basolateral membrane. The assembly of multiprotein signaling complexes at sites of membrane specialization may be a widespread function of dystrophin-related protein complexes.
Collapse
Affiliation(s)
- A M Kachinsky
- Department of Cell and Molecular Physiology and Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7545, USA
| | | | | |
Collapse
|
1019
|
Scanlan MJ, Williamson B, Jungbluth A, Stockert E, Arden KC, Viars CS, Gure AO, Gordan JD, Chen YT, Old LJ. Isoforms of the human PDZ-73 protein exhibit differential tissue expression. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1445:39-52. [PMID: 10209257 DOI: 10.1016/s0167-4781(99)00033-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Patients with renal and colon cancer frequently develop IgG autoantibodies toward the NY-CO-38/PDZ-73 antigen, a protein of 652 amino acids (73 kDa) which contains three copies of the PDZ protein-protein interaction domain. The gene encoding PDZ-73 mapped to chromosome 11p15.4-p15.1. Additional tissue-specific isoforms were identified: PDZ-45, which lacks the third PDZ domain and the putative PEST protein degradation motif, is expressed in kidney, colon, small intestine, brain and testis; PDZ-54 and PDZ-59, which also lack the third PDZ domains, have unique carboxyl terminal amino acids and are expressed in brain, kidney, bladder, colon cancer and renal cancer; and a putative PDZ-37 isoform, containing only the third PDZ domain, that is expressed in the central nervous system. Immunohistochemical staining with anti-PDZ 73 monoclonal antibodies showed strong cytoplasmic reactivity in epithelial cells of the small intestine, colon and kidney tubules, with a prominent apical staining pattern in cells of the small intestine. The reactivity pattern of the antibodies with various tissues correlated with the mRNA expression pattern of the PDZ-45 isoform. The existence of multiple PDZ-73 isoforms with variations in tissue distribution, PDZ domains, protein degradation sequences and carboxyl terminal structure indicate that these isoforms have distinct tissue-specific functions.
Collapse
Affiliation(s)
- M J Scanlan
- Ludwig Institute for Cancer Research, New York Branch at Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1020
|
Firestein BL, Bredt DS. Interaction of neuronal nitric-oxide synthase and phosphofructokinase-M. J Biol Chem 1999; 274:10545-50. [PMID: 10187848 DOI: 10.1074/jbc.274.15.10545] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Neurons that express neuronal nitric-oxide synthase (nNOS) are resistant to NO-induced neurotoxicity; however, the mechanism by which these neurons are protected is not clear. To identify proteins possibly involved in this process, we performed affinity chromatography with the nNOS PDZ domain, a N-terminal motif that mediates protein interactions. Using this method to fractionate soluble tissue extracts, we identified the muscle isoform of phosphofructokinase (PFK-M) as a protein that binds to nNOS both in brain and skeletal muscle. PFK-M interacts with the PDZ domain of nNOS, and nNOS-PFK-M binding can be competed by peptides that bind to the PDZ domain of nNOS. We found that nNOS is significantly associated with PFK-M in skeletal muscle because nNOS can be immunodepleted from cytosolic skeletal muscle extracts using an antibody directed against PFK-M. In brain, nNOS and PFK-M are both enriched in synaptosomes, and specifically, in the synaptic vesicle fraction, where they can interact. At the cellular level, PFK-M is enriched in neurons that express nNOS protein. As fructose-1, 6-bisphosphate, the product of PFK activity, is neuroprotective, the interaction of nNOS and PFK may contribute to neuroprotection of nNOS positive cells.
Collapse
Affiliation(s)
- B L Firestein
- Department of Physiology, UCSF, San Francisco, California 94143-0444, USA.
| | | |
Collapse
|
1021
|
Boissel JP, Schwarz PM, Förstermann U. Neuronal-type NO synthase: transcript diversity and expressional regulation. Nitric Oxide 1999; 2:337-49. [PMID: 10100489 DOI: 10.1006/niox.1998.0189] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Of the three established isoforms of NO synthase, the gene for the neuronal-type enzyme (NOS I) is by far the largest and most complicated one. The genomic locus of the human NOS I gene is located on chromosome 12 and distributed over a region greater than 200 kb. The nucleotide sequence corresponding to the major neuronal mRNA transcript is encoded by 29 exons. The full-length open reading frame codes for a protein of 1434 amino acids with a predicted molecular weight of 160.8 kDa. However, both in rodents and in humans, multiple, tissue-specific or developmentally regulated NOS I mRNA transcripts have been reported. They arise from the initiation by different transcriptional units containing alternative promoters (at least eight in the human gene), cassette exon deletions or insertions, and/or the usage of alternate polyadenylation signals. Depending on the insertions and deletions, translation results in functional or nonfunctional proteins. The use of alternative promoters can influence gene expression by various means. Indeed, NOS I is not a static, constitutively expressed enzyme, but subject to expressional regulation by various compounds and conditions. The molecular mechanisms underlying these regulations are currently being studied in several laboratories including our own.
Collapse
Affiliation(s)
- J P Boissel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| | | | | |
Collapse
|
1022
|
Abstract
The organization and regulation of synaptic connections in the mammalian nervous system entail complicated and co-ordinated molecular and cellular processes. The unveiling of various protein-protein interactions and their functional consequences at synapses have led to a greater understanding of the process of synapse formation and the modulation of synaptic transmission. Recent studies indicate that the major excitatory neurotransmitter receptors in the brain, the glutamate receptors, are associated with many different molecules that are involved in the formation of elaborate synaptic cytoskeletal networks and signal transduction cascades. These complex protein networks may play critical roles in the regulation of neurotransmitter receptor function and the efficacy of synaptic transmission.
Collapse
Affiliation(s)
- J H Kim
- Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|
1023
|
Tojo A, Bredt DS, Wilcox CS. Distribution of postsynaptic density proteins in rat kidney: relationship to neuronal nitric oxide synthase. Kidney Int 1999; 55:1384-94. [PMID: 10201003 DOI: 10.1046/j.1523-1755.1999.00392.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Neuronal nitric oxide synthase (nNOS) is expressed in skeletal muscle beneath the sarcolemma associated with dystrophin complex. In brain, nNOS is anchored to synaptic membranes by specific postsynaptic density proteins (PSD)-95 and PSD-93. We have investigated the cellular and subcellular localization of these PSD proteins in the kidney and their relationship to nNOS and the cell membrane. METHODS Kidneys from male Sprague-Dawley rats were processed for light and electron microscopic immunohistochemistry with polyclonal antibodies against PSD and nNOS proteins. RESULTS Western blot analysis of rat kidney revealed a specific band for PSD-93 at the molecular weight of 103 kDa. Immunostaining for PSD-93 was located in the thick ascending limb of the loop of Henle, macula densa cells, distal convoluted tubules, cortical collecting ducts, outer and inner medullary collecting duct, glomerular epithelium, and Bowman's capsule. A pre-embedding electron microscopic immunoperoxidase procedure localized PSD-93 to the basolateral membrane of these tubular cells. Using different sized immunogold particles, a portion of nNOS in the macula densa colocalized with PSD-93 adjacent to cytoplasmic vesicles and the basolateral membrane. In contrast, PSD-95 protein was detected only weakly in the cortex by Western blot. Immunostaining for PSD-95 was located only faintly in the apical membrane of the thick ascending limb, macula densa, distal convoluted tubule and cortical collecting duct cells. CONCLUSION PSD-93 is the predominant PSD expressed in the rat kidney. It is located primarily in the basolateral membranes of distal nephron and colocalizes with a pool of nNOS in cytoplasmic vesicles and basolateral membranes of macula densa cells.
Collapse
Affiliation(s)
- A Tojo
- Division of Nephrology and Endocrinology, University of Tokyo, Japan
| | | | | |
Collapse
|
1024
|
Loihl AK, Whalen S, Campbell IL, Mudgett JS, Murphy S. Transcriptional activation following cerebral ischemia in mice of a promoter-deleted nitric oxide synthase-2 gene. J Biol Chem 1999; 274:8844-9. [PMID: 10085127 DOI: 10.1074/jbc.274.13.8844] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nitric oxide synthase (NOS)-2 is transcriptionally activated in a wide variety of injurious conditions, including cerebral ischemia, and the resulting nitric oxide is implicated both in tissue damage and recovery. Studies in vitro suggest that the proximal region of the NOS-2 promoter is obligatory for gene activation by proinflammatory cytokines. However, following cerebral ischemia in a NOS-2 gene-deficient mouse in which this region and exons 1-4 have been deleted, we find temporal and spatial expression, identical to wild-type, from a previously unidentified promoter region. The resulting protein is predicted to lack the first 113 amino acids and is NOS-2-incompetent. Fortuitously, this gene-deficient mouse presents a unique opportunity to determine more about the mechanisms of NOS-2 gene regulation in vivo.
Collapse
Affiliation(s)
- A K Loihl
- Department of Pharmacology and the Neuroscience Program, University of Iowa College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|
1025
|
Adams DR, Brochwicz-Lewinski M, Butler AR. Nitric oxide: physiological roles, biosynthesis and medical uses. FORTSCHRITTE DER CHEMIE ORGANISCHER NATURSTOFFE = PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS. PROGRES DANS LA CHIMIE DES SUBSTANCES ORGANIQUES NATURELLES 1999; 76:1-211. [PMID: 10091554 DOI: 10.1007/978-3-7091-6351-1_1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- D R Adams
- Department of Chemistry, Heriot Watt University, Edinburgh, Scotland
| | | | | |
Collapse
|
1026
|
Fanning AS, Anderson JM. PDZ domains: fundamental building blocks in the organization of protein complexes at the plasma membrane. J Clin Invest 1999; 103:767-72. [PMID: 10079096 PMCID: PMC408156 DOI: 10.1172/jci6509] [Citation(s) in RCA: 365] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- A S Fanning
- Departments of Internal Medicine and Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520-8019, USA.
| | | |
Collapse
|
1027
|
Tutdibi O, Brinkmeier H, Rüdel R, Föhr KJ. Increased calcium entry into dystrophin-deficient muscle fibres of MDX and ADR-MDX mice is reduced by ion channel blockers. J Physiol 1999; 515 ( Pt 3):859-68. [PMID: 10066910 PMCID: PMC2269189 DOI: 10.1111/j.1469-7793.1999.859ab.x] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
1. Single fibres were enzymatically isolated from interosseus muscles of dystrophic MDX mice, myotonic-dystrophic double mutant ADR-MDX mice and C57BL/10 controls. The fibres were kept in cell culture for up to 2 weeks for the study of Ca2+ homeostasis and sarcolemmal Ca2+ permeability. 2. Resting levels of intracellular free Ca2+, determined with the fluorescent Ca2+ indicator fura-2, were slightly higher in MDX (63 +/- 20 nM; means +/- s.d.; n = 454 analysed fibres) and ADR-MDX (65 +/- 12 nM; n = 87) fibres than in controls (51 +/- 20 nM; n = 265). 3. The amplitudes of electrically induced Ca2+ transients did not differ between MDX fibres and controls. Decay time constants of Ca2+ transients ranged between 10 and 55 ms in both genotypes. In 50 % of MDX fibres (n = 68), but in only 20 % of controls (n = 54), the decay time constants were > 35 ms. 4. Bath application of Mn2+ resulted in a progressive quench of fura-2 fluorescence emitted from the fibres. The quench rate was about 2 times higher in MDX fibres (3.98 +/- 1.9 % min-1; n = 275) than in controls (2.03 +/- 1.4 % min-1; n = 204). The quench rate in ADR-MDX fibres (2.49 +/- 1.4 % min-1; n = 87) was closer to that of controls. 5. The Mn2+ influx into MDX fibres was reduced to 10 % by Gd3+, to 19 % by La3+ and to 47 % by Ni2+ (all at 50 microM). Bath application of 50 microM amiloride inhibited the Mn2+ influx to 37 %. 6. We conclude that in isolated, resting MDX muscle fibres the membrane permeability for divalent cations is increased. The presumed additional influx of Ca2+ occurs through ion channels, but is well compensated for by effective cellular Ca2+ transport systems. The milder dystrophic phenotype of ADR-MDX mice is correlated with a smaller increase of their sarcolemmal Ca2+ permeability.
Collapse
Affiliation(s)
- O Tutdibi
- Department of General Physiology, University of Ulm, D-89069 Ulm, Germany
| | | | | | | |
Collapse
|
1028
|
Yamada Y, Chochi Y, Takamiya K, Sobue K, Inui M. Modulation of the channel activity of the epsilon2/zeta1-subtype N-methyl D-aspartate receptor by PSD-95. J Biol Chem 1999; 274:6647-52. [PMID: 10037761 DOI: 10.1074/jbc.274.10.6647] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A channel-associated protein PSD-95 has been shown to induce clustering of N-methyl D-aspartate (NMDA) receptors, interacting with the COOH terminus of the epsilon subunit of the receptors. The effects of PSD-95 on the channel activity of the epsilon2/zeta1 heteromeric NMDA receptor were examined by injection of PSD-95 cRNA into Xenopus oocytes expressing the NMDA receptors. Expression of PSD-95 decreased the sensitivity of the NMDA receptor channels to L-glutamate. Mutational studies showed that the interaction between the COOH terminus of the epsilon2 subunit of the NMDA receptor and the second PSD-95/Dlg/Z0-1 domain of PSD-95 is critical for the decrease in glutamate sensitivity. It is known that protein kinase C markedly potentiates the channel activity of the NMDA receptor expressed in oocytes. PSD-95 inhibited the protein kinase C-mediated potentiation of the channels. Thus, we demonstrated that PSD-95 functionally modulates the channel activity of the epsilon2/zeta1 NMDA receptor. PSD-95 makes signal transmission more efficient by clustering the channels at postsynaptic sites. In addition to this, our results suggest that PSD-95 plays a protective role against neuronal excitotoxicity by decreasing the glutamate sensitivity of the channels and by inhibiting the protein kinase C-mediated potentiation of the channels.
Collapse
Affiliation(s)
- Y Yamada
- Department of Pharmacology, Yamaguchi University School of Medicine, 1144 Kogushi, Ube, Yamaguchi 755-8505, Japan
| | | | | | | | | |
Collapse
|
1029
|
Keshet GI, Ovadia H, Taraboulos A, Gabizon R. Scrapie-infected mice and PrP knockout mice share abnormal localization and activity of neuronal nitric oxide synthase. J Neurochem 1999; 72:1224-31. [PMID: 10037495 DOI: 10.1046/j.1471-4159.1999.0721224.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PrP(Sc), the only identified component of the scrapie prion, is a conformational isoform of PrPc. The physiological role of PrPc, a glycolipid-anchored glycoprotein, is still unknown. We have shown previously that neuronal nitric oxide synthase (nNOS) activity is impaired in the brains of mice sick with experimental scrapie as well as in scrapie-infected neuroblastoma cells. In this work we investigated the cell localization of nNOS in brains of wild-type and scrapie-infected mice as well as in mice in which the PrP gene was ablated. We now report that whereas in wild-type mice, nNOS, like PrPc, is associated with detergent-insoluble cholesterol-rich membranous microdomains (rafts), this is not the case in brains of scrapie-infected or in those of adult PrP(0/0) mice. Also, adult PrP(0/0), like scrapie-infected mice, show reduced nNOS activity. We suggest that PrPc may play a role in the targeting of nNOS to its proper subcellular localization. The similarities of nNOS properties in PrP(0/0) as compared with scrapie-infected mice suggest that at least this role of PrPc may be impaired in scrapie-infected brains.
Collapse
Affiliation(s)
- G I Keshet
- Department of Neurology, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | | | | | | |
Collapse
|
1030
|
Albers DS, Weiss SW, Iadarola MJ, Standaert DG. Immunohistochemical localization of N-methyl-D-aspartate and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptor subunits in the substantia nigra pars compacta of the rat. Neuroscience 1999; 89:209-20. [PMID: 10051230 DOI: 10.1016/s0306-4522(98)00328-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Ionotropic glutamate receptors in the substantia nigra pars compacta regulate the activity of dopamine neurons. We have used dual-label immunofluoresence and confocal laser microscopy to study the localization of subunits of two types of ionotropic receptors within the substantia nigra pars compacta of the rat. Immunostaining for N-methyl-D-aspartate receptor 1 and glutamate receptor 2/3 was prominent in the soma and proximal dendrites of all tyrosine hydroxylase-immunopositive cells, while only low amounts of N-methyl-D-aspartate receptor 2A and N-methyl-D-aspartate receptor 2B were present. Selective antibodies were used to determine the isoforms of N-methyl-D-aspartate receptor 1 present. Immunostaining for the N1, C1 and C2 variably spliced segments of N-methyl-D-aspartate receptor 1 were scarce in the substantia nigra pars compacta, while immunoreactivity for the alternative C2' terminus of N-methyl-D-aspartate receptor 1 was quite abundant. Staining for glutamate receptor 1 was heterogeneous; about half of the tyrosine hydroxylase immunopositive cells stained intensely, while the other half were immunonegative. The glutamate receptor 1-stained cells were concentrated in the ventral tier of the substantia nigra pars compacta. Glutamate receptor 4 was not found in tyrosine hydroxylase-immunopositive cells within the substantia nigra pars compacta. Together, these data demonstrate that dopaminergic neurons in the substantia nigra pars compacta express primarily glutamate receptor 1, glutamate receptor 2/3 and N-methyl-D-aspartate receptor 1 isoforms containing the alternative C2' terminus.
Collapse
Affiliation(s)
- D S Albers
- Neurology Service, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | | | | | | |
Collapse
|
1031
|
Sato K, Nakagawa H, Kuriyama H, Altschuler RA. Differential distribution of N-methyl-D-aspartate receptor-2 subunit messenger RNA in the rat superior olivary complex. Neuroscience 1999; 89:839-53. [PMID: 10199617 DOI: 10.1016/s0306-4522(98)00350-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The distribution of N-methyl-D-aspartate receptor subunit messenger RNA was examined in the superior olivary complex of the rat using in situ hybridization methods. Non-radioactive methods showed N-methyl-D-aspartate receptor-1 and N-methyl-D-aspartate receptor-2A, -2B and -2C subunit expression to be abundant in principal cells of all major superior olivary complex regions, with N-methyl-D-aspartate receptor-1 expression higher than N-methyl-D-aspartate receptor-2. After auto-radiographic in situ hybridization, counting silver grains over cells was used to quantitate and compare levels of expression of the N-methyl-D-aspartate receptor-2 subunits. N-Methyl-D-aspartate receptor-2A, -2B and -2C expression was detected over cells in the lateral and medial superior olive, the medial, lateral and ventral nucleus of the trapezoid body, and the superior paraolivary nucleus. N-Methyl-D-aspartate receptor-2D expression was only observed in the lateral superior olive, lateral nucleus of the trapezoid body and ventral nucleus of the trapezoid body. Three distinct patterns of labeling were observed. The lateral superior olive and ventral nucleus of the trapezoid body showed higher expression of N-methyl-D-aspartate receptor-2A and -2C than other subunits; the medial nucleus of the trapezoid body showed higher N-methyl-D-aspartate receptor-2B expression than other subunits; and the medial superior olive, superior paraolivary nucleus and ventral nucleus of the trapezoid body showed relatively equivalent expression of N-methyl-D-aspartate receptor-2A, -2B and -2C subunits. N-Methyl-D-aspartate receptor-2D had the lowest expression, with levels greater than background in only the lateral superior olive, ventral nucleus of the trapezoid body and lateral nucleus of the trapezoid body. Immunocytochemistry using antibodies to N-methyl-D-aspartate receptor-2A, -2B and -2C showed immunolabeling consistent with in situ hybridization results. These results show diversity in expression of N-methyl-D-aspartate receptor-2 subunits in different superior olivary complex regions and would predict pharmacological differences.
Collapse
Affiliation(s)
- K Sato
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor 48109-0506, USA
| | | | | | | |
Collapse
|
1032
|
van Rossum D, Kuhse J, Betz H. Dynamic interaction between soluble tubulin and C-terminal domains of N-methyl-D-aspartate receptor subunits. J Neurochem 1999; 72:962-73. [PMID: 10037467 DOI: 10.1046/j.1471-4159.1999.0720962.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The cytoplasmic C-terminal domains (CTs) of the NR1 and NR2 subunits of the NMDA receptor have been implicated in its anchoring to the subsynaptic cytoskeleton. Here, we used affinity chromatography with glutathione S-transferase-NR1-CT and -NR2B-CT fusion proteins to identify novel binding partner(s) of these NMDA receptor subunits. Upon incubation with rat brain cytosolic protein fraction, both NR1-CT and NR2B-CT, but not glutathione S-transferase, specifically bound tubulin. The respective fusion proteins also bound tubulin purified from brain, suggesting a direct interaction between the two binding partners. In tubulin polymerization assays, NR1-CT and NR2B-CT significantly decreased the rate of microtubule formation without destabilizing preformed microtubules. Moreover, only minor fractions of either fusion protein coprecipitated with the newly formed microtubules. Consistent with these findings, ultrastructural analysis of the newly formed microtubules revealed a limited association only with the CTs of the NR1 and NR2B. These data suggest a direct interaction of the NMDA receptor channel subunit CTs and tubulin dimers or soluble forms of tubulin. The efficient modulation of microtubule dynamics by the NR1 and NR2 cytoplasmic domains suggests a functional interaction of the receptor and the subsynaptic cytoskeletal network that may play a role during morphological adaptations, as observed during synaptogenesis and in adult CNS plasticity.
Collapse
Affiliation(s)
- D van Rossum
- Department of Neurochemistry, Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | | | | |
Collapse
|
1033
|
Craven SE, El-Husseini AE, Bredt DS. Synaptic targeting of the postsynaptic density protein PSD-95 mediated by lipid and protein motifs. Neuron 1999; 22:497-509. [PMID: 10197530 DOI: 10.1016/s0896-6273(00)80705-9] [Citation(s) in RCA: 271] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
During synaptic development, proteins aggregate at specialized pre- and postsynaptic structures. Mechanisms that mediate protein clustering at these sites remain unknown. To investigate this process, we analyzed synaptic targeting of a postsynaptic density protein, PSD-95, by expressing green fluorescent protein- (GFP-) tagged PSD-95 in cultured hippocampal neurons. We find that postsynaptic clustering relies on three elements of PSD-95: N-terminal palmitoylation, the first two PDZ domains, and a C-terminal targeting motif. In contrast, disruptions of PDZ3, SH3, or guanylate kinase (GK) domains do not affect synaptic targeting. Palmitoylation is sufficient to target the diffusely expressed SAP-97 to synapses, and palmitoylation cannot be replaced with alternative membrane association motifs, suggesting that a specialized synaptic lipid environment mediates postsynaptic clustering. The requirements for PDZ domains and a C-terminal domain of PSD-95 indicate that protein-protein interactions cooperate with lipid interactions in synaptic targeting.
Collapse
Affiliation(s)
- S E Craven
- Department of Physiology and Program in Neuroscience, University of California, San Francisco 94143-0444, USA
| | | | | |
Collapse
|
1034
|
Kriegsfeld LJ, Eliasson MJ, Demas GE, Blackshaw S, Dawson TM, Nelson RJ, Snyder SH. Nocturnal motor coordination deficits in neuronal nitric oxide synthase knock-out mice. Neuroscience 1999; 89:311-5. [PMID: 10077313 DOI: 10.1016/s0306-4522(98)00614-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Nitric oxide is formed in the brain primarily by neurons containing neuronal nitric oxide synthase (nNOS), though some neurons may express endothelial NOS (eNOS), and inducible NOS (iNOS) only occurs in neurons following toxic stimuli. Mice with targeted disruption of nNOS (nNOS-) display distended stomachs with hypertrophied pyloric sphincters reflecting loss of nNOS in myenteric plexus neurons. nNOS- animals resist brain damage following middle cerebral artery occlusions consistent with evidence that excess release of nitric oxide mediates neurotoxicity in ischemic stroke. Neuronal NOS- mice have no grossly evident defects in locomotor activity, breeding long-term depression in the cerebellum, long-term potentiation in the hippocampus, and overall sensorimotor function. However, nNOS- animals display excessive, inappropriate sexual behavior and dramatic increases in aggression. Because the cerebellum possesses the greatest levels of nNOS neurons in the brain, it was surprising that presumed cerebellar functions such as balance and coordination were grossly normal in nNOS- mice. These previous studies were all conducted during the day (between 1400 and 1600, lights on at 0700). We now report striking, discrete abnormalities in balance and motor coordination in nNOS-mice reflected selectively at night.
Collapse
Affiliation(s)
- L J Kriegsfeld
- Department of Psychology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | | | | | | | | | |
Collapse
|
1035
|
Itoh M, Morita K, Tsukita S. Characterization of ZO-2 as a MAGUK family member associated with tight as well as adherens junctions with a binding affinity to occludin and alpha catenin. J Biol Chem 1999; 274:5981-6. [PMID: 10026224 DOI: 10.1074/jbc.274.9.5981] [Citation(s) in RCA: 223] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ZO-2, a member of the MAGUK family, was thought to be specific for tight junctions (TJs) in contrast to ZO-1, another MAGUK family member, which is localized at TJs and adherens junctions (AJs) in epithelial and nonepithelial cells, respectively. Mouse ZO-2 cDNA was isolated, and a specific polyclonal antibody was generated using corresponding synthetic peptides as antigens. Immunofluorescence microscopy with this polyclonal antibody revealed that, similarly to ZO-1, in addition to TJs in epithelial cells, ZO-2 was also concentrated at AJs in nonepithelial cells such as fibroblasts and cardiac muscle cells lacking TJs. When NH2-terminal dlg-like and COOH-terminal non-dlg-like domains of ZO-2 (N-ZO-2 and C-ZO-2, respectively) were separately introduced into cultured cells, N-ZO-2 was colocalized with endogenous ZO-1/ZO-2, i.e. at TJs in epithelial cells and at AJs in non-epithelial cells, whereas C-ZO-2 was distributed along actin filaments. Consistently, occludin as well as alpha catenin directly bound to N-ZO-2 as well as the NH2-terminal dlg-like portion of ZO-1 (N-ZO-1) in vitro. Furthermore, immunoprecipitation experiments revealed that the second PDZ domain of ZO-2 was directly associated with N-ZO-1. These findings indicated that ZO-2 forms a complex with ZO-1/occludin or ZO-1/alpha catenin to establish TJ or AJ domains, respectively.
Collapse
Affiliation(s)
- M Itoh
- Department of Cell Biology, Faculty of Medicine, Kyoto University, Sakyo-ku, Kyoto 606, Japan
| | | | | |
Collapse
|
1036
|
Masuko N, Makino K, Kuwahara H, Fukunaga K, Sudo T, Araki N, Yamamoto H, Yamada Y, Miyamoto E, Saya H. Interaction of NE-dlg/SAP102, a neuronal and endocrine tissue-specific membrane-associated guanylate kinase protein, with calmodulin and PSD-95/SAP90. A possible regulatory role in molecular clustering at synaptic sites. J Biol Chem 1999; 274:5782-90. [PMID: 10026200 DOI: 10.1074/jbc.274.9.5782] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NE-dlg/SAP102, a neuronal and endocrine tissue-specific membrane-associated guanylate kinase family protein, is known to bind to C-terminal ends of N-methyl-D-aspartate receptor 2B (NR2B) through its PDZ (PSD-95/Dlg/ZO-1) domains. NE-dlg/SAP102 and NR2B colocalize at synaptic sites in cultured rat hippocampal neurons, and their expressions increase in parallel with the onset of synaptogenesis. We have identified that NE-dlg/SAP102 interacts with calmodulin in a Ca2+-dependent manner. The binding site for calmodulin has been determined to lie at the putative basic alpha-helix region located around the src homology 3 (SH3) domain of NE-dlg/SAP102. Using a surface plasmon resonance measurement system, we detected specific binding of recombinant NE-dlg/SAP102 to the immobilized calmodulin with a Kd value of 44 nM. However, the binding of Ca2+/calmodulin to NE-dlg/SAP102 did not modulate the interaction between PDZ domains of NE-dlg/SAP102 and the C-terminal end of rat NR2B. We have also identified that the region near the calmodulin binding site of NE-dlg/SAP102 interacts with the GUK-like domain of PSD-95/SAP90 by two-hybrid screening. Pull down assay revealed that NE-dlg/SAP102 can interact with PSD-95/SAP90 in the presence of both Ca2+ and calmodulin. These findings suggest that the Ca2+/calmodulin modulates interaction of neuronal membrane-associated guanylate kinase proteins and regulates clustering of neurotransmitter receptors at central synapses.
Collapse
Affiliation(s)
- N Masuko
- Department of Tumor Genetics and Biology, Kumamoto University School of Medicine, 2-2-1, Honjo, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1037
|
Abstract
Nitric oxide (NO) has been implicated in the establishment of precise synaptic connectivity throughout the neuroaxis in several species. To determine the contribution of NO to NMDA receptor-dependent dendritic growth in motor neurons, we administered the NMDA antagonist MK-801 to wild-type mice and neuronal nitric oxide synthase (nNOS) knock-out mice between postnatal days 7 and 14. Compared to saline-treated wild-type animals the number of dendritic bifurcations was significantly reduced in nNOS knock-out animals and MK-801-treated wild-type animals. There was no significant difference in dendritic bifurcation between MK-801-treated wild-type, MK-801-treated nNOS knock-out, and saline-treated nNOS knock-out animals, suggesting that nNOS knock-out and NMDA receptor block had similar effects. The path of the longest dendrite and the number of primary dendrites was the same in all treatment groups, indicating an effect specific to bifurcation. Sholl analysis revealed that differences in bifurcation numbers occurred between 160 and 320 micrometers from the cell body, the distance at which second, third, and fourth order dendrites are most prevalent. Dendrite order analyses confirmed a significant reduction in numbers, but not lengths, of third and fourth order dendrites in nNOS knock-out and drug-treatment groups. Finally, immunohistochemical examination of the developing spinal cord indicated that NMDA receptors and nNOS are colocalized within interneurons surrounding the motor neuron pool. These results support the view that at least part of NMDA receptor-dependent arborization of motor neuron dendrites is mediated by the local production of NO within the developing spinal cord.
Collapse
|
1038
|
Abstract
The highly reactive free radical gas, nitric oxide, serves a variety of biomodulatory functions and has been implicated in a growing array of physiological and pathophysiological states. The striking differences between this labile substance and other, more conventional, signaling molecules highlight the tight degree of nitric oxide regulation that is required in order to maintain appropriate cellular homeostasis. The generation of nitric oxide represents a common component of the signal transduction pathways of a number of chemical signaling molecules that act via binding to G protein-coupled receptors. This review focuses on the relationship between this receptor superfamily, the generation of nitric oxide via the actions of the nitric oxide synthases and some of the inter- and intracellular roles of nitric oxide.
Collapse
Affiliation(s)
- A Christopoulos
- Neuroscience Research in Psychiatry, University of Minnesota Medical School, Minneapolis 55455, USA
| | | |
Collapse
|
1039
|
Affiliation(s)
- Y P Hsueh
- Howard Hughes Medical Institute, Massachusetts General Hospital, Boston 02114, USA
| | | |
Collapse
|
1040
|
Abstract
Proteins of the membrane-associated guanylate kinase family play an important role in the anchoring and clustering of neurotransmitter receptors in the postsynaptic density (PSD) at many central synapses. However, relatively little is known about how these multifunctional scaffold proteins might provide a privileged site for activity- and cell type-dependent specification of the postsynaptic signaling machinery. Rho signaling pathway has classically been implicated in mechanisms of axonal outgrowth, dendrogenesis, and cell migration during neural development, but its contribution remains unclear at the synapses in the mature CNS. Here, we present evidence that Citron, a Rho-effector in the brain, is enriched in the PSD fraction and interacts with PSD-95/synapse-associated protein (SAP)-90 both in vivo and in vitro. Citron colocalization with PSD-95 occurred, not exclusively but certainly, at glutamatergic synapses in a limited set of neurons, such as the thalamic excitatory neurons; Citron expression, however, could not be detected in the principal neurons of the hippocampus and the cerebellum in the adult mouse brain. In a heterologous system, Citron was shown to form a heteromeric complex not only with PSD-95 but also with NMDA receptors. Thus, Citron-PSD-95/SAP-90 interaction may provide a region- and cell type-specific link between the Rho signaling cascade and the synaptic NMDA receptor complex.
Collapse
|
1041
|
Lipton SA, Rayudu PV, Choi YB, Sucher NJ, Chen HS. Redox modulation of the NMDA receptor by NO-related species. PROGRESS IN BRAIN RESEARCH 1999; 118:73-82. [PMID: 9932435 DOI: 10.1016/s0079-6123(08)63201-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The chemical reactions of NO are largely dictated by its redox state. Increasing evidence suggests that the various redox states of the NO group exist endogenously in biological tissues. In the case of NO+ equivalents, the mechanism of reaction often involves S-nitrosylation (transfer of the NO group to a cysteine sulfhydryl to form an RS-NO); further oxidation of critical thiols can possibly form disulfide bonds. We have physiological and chemical evidence that NMDA receptor activity can be modulated by S-nitrosylation, resulting in a decrease in channel opening. Recent data suggest that NO-, probably in the singlet (or high-energy) state, can also react with critical sulfhydryl group(s) of the NMDA receptor to down-regulate its activity; in the triplet (lower-energy) state NO- may oxidize these NMDA receptor sulfhydryl groups by formation of an intermediate such as peroxynitrite. It has also been reported that NO can react with thiol but only under specific circumstances, e.g., if an electron acceptor such as O2 is present, as well at catalytic amounts of metals like copper, and if the conditions do not favor the kinetically preferred reaction with O2.- to yield peroxynitrite. Mounting evidence in many fields suggests that S-nitrosylation can regulate the biological activity of a great variety of proteins, perhaps analogous to phosphorylation. Thus, this chemical reaction is gaining acceptance as a newly-recognized molecular switch to control protein function via reactive thiol groups such as those encountered on the NMDA receptor.
Collapse
Affiliation(s)
- S A Lipton
- CNS Research Institute, Brigham and Women's Hospital, and Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
1042
|
Hawkins RD, Son H, Arancio O. Nitric oxide as a retrograde messenger during long-term potentiation in hippocampus. PROGRESS IN BRAIN RESEARCH 1999; 118:155-72. [PMID: 9932440 DOI: 10.1016/s0079-6123(08)63206-9] [Citation(s) in RCA: 215] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nitric oxide (NO) is widespread in the nervous system and is thought to play a role in a variety of different neuronal functions, including learning and memory (see other chapters, this volume). A number of behavioral studies have indicated that NO is involved in several types of learning such as motor learning (Yanagihara and Kondo, 1996), avoidance learning (Barati and Kopf, 1996; Myslivecek et al., 1996), olfactory learning (Okere et. al., 1996; Kendrick et al., 1997), and spatial learning (Holscher et al., 1995; Yamada et al., 1996) (for review of earlier papers see Hawkins, 1996). Moreover, NO is thought to be involved in neuronal plasticity contributing to these different types of learning in different brain areas including the cerebellum (chapter by R. Tsien, this volume) and hippocampus. In this chapter we review evidence on the role of NO in long-term potentiation (LTP), a type of synaptic plasticity in hippocampus that is believed to contribute to declarative forms of learning such as spatial learning.
Collapse
Affiliation(s)
- R D Hawkins
- Center for Neurobiology and Behavior, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| | | | | |
Collapse
|
1043
|
Lin D, Gish GD, Songyang Z, Pawson T. The carboxyl terminus of B class ephrins constitutes a PDZ domain binding motif. J Biol Chem 1999; 274:3726-33. [PMID: 9920925 DOI: 10.1074/jbc.274.6.3726] [Citation(s) in RCA: 201] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ephrin B proteins function as ligands for B class Eph receptor tyrosine kinases and are postulated to possess an intrinsic signaling function. The sequence at the carboxyl terminus of B-type ephrins contains a putative PDZ binding site, providing a possible mechanism through which transmembrane ephrins might interact with cytoplasmic proteins. To test this notion, a day 10.5 mouse embryonic expression library was screened with a biotinylated peptide corresponding to the carboxyl terminus of ephrin B3. Three of the positive cDNAs encoded polypeptides with multiple PDZ domains, representing fragments of the molecule GRIP, the protein syntenin, and PHIP, a novel PDZ domain-containing protein related to Caenorhabditis elegans PAR-3. In addition, the binding specificities of PDZ domains previously predicted by an oriented library approach (Songyang, Z., Fanning, A. S., Fu, C., Xu, J., Marfatia, S. M., Chishti, A. H., Crompton, A., Chan, A. C., Anderson, J. M., and Cantley, L. C. (1997) Science 275, 73-77) identified the tyrosine phosphatase FAP-1 as a potential binding partner for B ephrins. In vitro studies demonstrated that the fifth PDZ domain of FAP-1 and full-length syntenin bound ephrin B1 via the carboxyl-terminal motif. Lastly, syntenin and ephrin B1 could be co-immunoprecipitated from transfected COS-1 cells, suggesting that PDZ domain binding of B ephrins can occur in cells. These results indicate that the carboxyl-terminal motif of B ephrins provides a binding site for specific PDZ domain-containing proteins, which might localize the transmembrane ligands for interactions with Eph receptors or participate in signaling within ephrin B-expressing cells.
Collapse
Affiliation(s)
- D Lin
- Programme in Molecular Biology and Cancer, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada
| | | | | | | |
Collapse
|
1044
|
Cramer KS, Leamey CA, Sur M. Nitric oxide as a signaling molecule in visual system development. PROGRESS IN BRAIN RESEARCH 1999; 118:101-14. [PMID: 9932437 DOI: 10.1016/s0079-6123(08)63203-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
The lateral geniculate nucleus (LGN) of the ferret is characterized by the readily discernible anatomical patterning of afferent terminations from the retina into both eye-specific layers and On/Off sublaminae. The eye-specific layers form during the first post-natal week, and On/Off sublaminae become apparent during the third to fourth post-natal weeks. The post-natal appearance of these patterns thus provides an advantageous model for the study of the mechanisms of activity-dependent development. The second phase of pattern formation, the appearance of On/Off sublaminae, involves the elaboration of appropriately placed axonal terminals and the restriction (or retraction) of inappropriately placed terminals. Previous work has demonstrated that this process is dependent on the activation of NMDA-receptors. Other studies have provided strong evidence that nitric oxide, a diffusible gas which is produced downstream of NMDA-receptor activation, acts as a retrograde messenger molecule to induce changes in pre-synaptic structures. In this article we review the evidence that nitric oxide plays a role in activity-dependent synaptic plasticity in the developing retinogeniculate pathway. The role of nitric oxide in other aspects of visual system development is also discussed.
Collapse
Affiliation(s)
- K S Cramer
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge 02139, USA.
| | | | | |
Collapse
|
1045
|
Affiliation(s)
- T Dalkara
- Department of Neurology, Hacettepe University Hospitals, Ankara, Turkey
| | | | | |
Collapse
|
1046
|
Huang PL, Lo EH. Genetic analysis of NOS isoforms using nNOS and eNOS knockout animals. PROGRESS IN BRAIN RESEARCH 1999; 118:13-25. [PMID: 9932431 DOI: 10.1016/s0079-6123(08)63197-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
All three major isoforms of nitric oxide synthase (NOS) are expressed in the brain. Because of complex and overlapping expression patterns (Marletta, 1994; Nathan and Xie, 1994), the particular NOS isoform involved in many processes is not clear. In fact, NO generated by separate isoforms may have different roles and potentially opposing effects (Iadecola et al., 1994). We have taken a genetic approach, to disrupt or knockout the genes for NOS isoforms to circumvent some of the limitations of pharmacologic agents. This approach allows the study of each individual NOS isoform in physiologic processes in the context of intact animals. It gives insights into possible developmental roles for NO and parallel processes that may compensate for the absence of each NOS isoform. We have made nNOS and eNOS knockout mice, as well as double knockout mice that lack both nNOS and eNOS isoforms (Huang et al., 1993; Huang et al., 1995; Son et al., 1996). In this chapter, we review some of the physiologic roles for NO that have been elucidated making use of these mice, including regulation of cerebral blood flow, response to cerebral ischemia, regulation of neurotransmitter release in the brain, and development of synaptic plasticity. Other chapters will discuss results using NOS knockout animals in studies of long term potentiation (see Hawkins, this volume), neuronal development (see Mize, this volume), and potential mechanisms for protection in nNOS knockout mice (Moskowitz, M.A.; Dawson, V.L, this volume).
Collapse
Affiliation(s)
- P L Huang
- Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Boston 02114, USA
| | | |
Collapse
|
1047
|
Kara P, Friedlander MJ. Dynamic modulation of cerebral cortex synaptic function by nitric oxide. PROGRESS IN BRAIN RESEARCH 1999; 118:183-98. [PMID: 9932442 DOI: 10.1016/s0079-6123(08)63208-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our experiments demonstrate that NO exerts several actions in the cerebral cortex (see Fig. 4). Its production is mediated by neuronal activity through at least two pathways, NMDA receptors and AMPA receptors. By virtue of its diffusion in extracellular space, NO can interact with synapses that are near the production site but not necessarily anatomically connected to the NO source by a conventional synaptic linkage. NO's primary action is amplification of the release of the excitatory neurotransmitter, L-glutamate, thus effectively creating a positive feed-forward gain system. However, a number of effective brakes, presumably activated under physiological conditions, serve to limit the cascade. These include NO's ability to inhibit NMDA receptors, its negative feedback on the rate limiting enzyme, NOS (Rengasamy and Johns, 1993; Park et al., 1994; Ravichandran et al., 1995) and other inhibitory actions (Figs. 3H and L). Under conditions of extremely strong activation or curtailment of the inhibitory feedback mechanisms, as might occur with a change in the local redox milieu (see Lipton, this volume), the amplification cascade may proceed unchecked leading to neurotoxicity (see Dawson, this volume). NO's ability to modulate synaptic function is indicated by both its positive and negative modulatory role in a form of activity-dependent synaptic plasticity, covariance-induced synaptic potentiation. These opposing effects may be due to NO's ability to amplify glutamate release and inhibit NMDA receptors, respectively. The actions of endogenous NO in vivo are primarily facilitatory in visual cortex (Fig. 4). However, inhibitory actions also occur in vivo. The targets for NO in vivo, are potentially more diverse including the neurotransmitter release process, NMDA receptors, other receptors and ion channels and the cerebral vasculature. However, regardless of the signaling pathways, the net result of endogenous NO production in the intact visual cortex is a potent modulation of cells' responses to visual stimulation. Thus, it is likely that this signal plays an important role in ongoing information processing in the mature cerebral cortex, dynamically altering the effective strength of cortical networks.
Collapse
Affiliation(s)
- P Kara
- Department of Neurobiology, University of Alabama at Birmingham, USA
| | | |
Collapse
|
1048
|
Fessenden JD, Altschuler RA, Seasholtz AF, Schacht J. Nitric oxide/cyclic guanosine monophosphate pathway in the peripheral and central auditory system of the rat. J Comp Neurol 1999. [DOI: 10.1002/(sici)1096-9861(19990201)404:1<52::aid-cne4>3.0.co;2-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
1049
|
Schuller AG, King MA, Zhang J, Bolan E, Pan YX, Morgan DJ, Chang A, Czick ME, Unterwald EM, Pasternak GW, Pintar JE. Retention of heroin and morphine-6 beta-glucuronide analgesia in a new line of mice lacking exon 1 of MOR-1. Nat Neurosci 1999; 2:151-6. [PMID: 10195199 DOI: 10.1038/5706] [Citation(s) in RCA: 245] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Morphine produces analgesia by activating mu opioid receptors encoded by the MOR-1 gene. Although morphine-6 beta-glucuronide (M6G), heroin and 6-acetylmorphine also are considered mu opioids, recent evidence suggests that they act through a distinct receptor mechanism. We examined this question in knockout mice containing disruptions of either the first or second coding exon of MOR-1. Mice homozygous for either MOR-1 mutation were insensitive to morphine. Heroin, 6-acetylmorphine and M6G still elicited analgesia in the exon-1 MOR-1 mutant, which also showed specific M6G binding, whereas M6G and 6-acetylmorphine were inactive in the exon-2 MOR-1 mutant. These results provide genetic evidence for a unique receptor site for M6G and heroin analgesia.
Collapse
Affiliation(s)
- A G Schuller
- Dept. of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscätaway 08854, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1050
|
Guo Y, Greenwood MT, Petrof BJ, Hussain SN. Expression and regulation of protein inhibitor of neuronal nitric oxide synthase in ventilatory muscles. Am J Respir Cell Mol Biol 1999; 20:319-26. [PMID: 9922224 DOI: 10.1165/ajrcmb.20.2.3437] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In skeletal muscle fibers, nitric oxide (NO) is synthesized by neuronal NO synthase (nNOS) and regulates excitation-contraction coupling, glucose uptake, and mitochondrial respiration. Recently, a novel 89-amino acid protein, designated protein inhibitor of nNOS (PIN), has been shown to interact with and specifically inhibit nNOS activity. In this study, we investigated the distribution, localization, and regulation of PIN expression in ventilatory and limb muscles of various species. Amplified PIN cDNA from the rat diaphragm revealed an open reading frame identical to that of human PIN. Among muscles of adult rats, PIN mRNA was strongly expressed in muscles rich in type I fibers, whereas much weaker expression was evident in muscles rich in type II fibers. By comparison, PIN protein expression was not related to fiber-type distribution. Similarly, PIN protein was equally expressed among rat, mouse, and human diaphragms. Both PIN mRNA and PIN protein were expressed at much higher levels in the embryonic rat diaphragm than in adult muscle. Immunohistochemistry revealed that PIN protein was localized in close proximity to the sarcolemma and nuclei. PIN protein was also abundant in muscle spindles and axons of nerves supplying skeletal muscle fibers. We conclude that PIN is expressed in various skeletal muscle fibers and that its expression is regulated during muscle development. The localization of PIN in muscle regions containing abundant nNOS protein suggests that it plays a role in the regulation of NO synthesis in skeletal muscle fibers.
Collapse
Affiliation(s)
- Y Guo
- Critical Care and Respiratory Divisions, Royal Victoria Hospital; and Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|