Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters











Publication year range
1.
J Clin Pharm Ther ; 40(5): 489-495, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26096723

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: It is known that mismanagement of intravenous (IV) fluid therapy may cause serious complications. The 2013 NICE guideline on intravenous fluid therapy in hospitalized adults also emphasizes the importance of appropriate prescribing of IV fluid. So far, no systematic review of the incidence and types of inappropriate prescribing of IV fluid has been conducted. Therefore, this study was undertaken to review the research literature on inappropriate prescribing of IV fluid in adult patients and develop corresponding strategies for improving practice. METHODS: A comprehensive literature search was performed. Critical appraisals were conducted on the articles drawn from the search, and an analysis was performed on the results. RESULTS AND DISCUSSION: Incorrect volumes and types of IV fluids prescribed, classified as misprescribing, was the most common type of inappropriate prescribing. Commonly, patients on IV fluid therapy were prescribed a greater volume of fluid and amount of sodium in excess of normal requirements. Doctors did not always check the body weight, serum electrolyte level and serum creatinine before prescribing IV fluid for patients. The other common type of inappropriate prescribing was incomplete/incorrect prescription writing. These common inappropriate prescribing of IV fluid could be caused by insufficient knowledge and training of the prescribers. In addition, the ignorance of the importance of IV fluid prescribing also contributed to this behaviour. WHAT IS NEW AND CONCLUSION: There is an urgent need to make doctors aware of these problems and enhance appropriate training on IV fluid prescribing, especially on the appropriate volume and amount of electrolytes. Pharmacists could exert a role in reviewing the fluid prescription chart for improving clinical practice.

2.
Genes Brain Behav ; 11(7): 794-805, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22809330

ABSTRACT

Neurogranin (Ng), a brain-specific calmodulin-binding protein, is expressed highly in hippocampus, and is important for cognitive function. Deletion of the Ng gene from mice caused attenuation of signal reaction cascade in hippocampus, impairments in learning and memory and high frequency stimulation-induced long-term potentiation (LTP). Environmental enrichment alone failed to improve cognitive function. In this study, behavioral testing revealed that Ng knockout (NgKO) mice were both hyperactive and socially withdrawn. Methylphenidate (MPH) was given to mice while they were also kept under an enrichment condition. MPH treatment reduced the hyperactivity of NgKO mice tested in both the open field and forced swim chamber. MPH improved their social abilities such that mice recognized and interacted better with novel subjects. The cognitive memories of MPH-treated mutants were improved in both water maze and contextual fear conditioning tests. High frequency stimulation-induced LTP of NgKO mice was also improved by MPH. The present treatment regimen, however, did not fully reverse the deficits of the mutant mice. In contrast, MPH exerted only a minimal effect on the wild type mice. At the cellular level, MPH increased the number of glial fibrillary acidic protein-positive cells in hippocampus, particularly within the dentate gyrus of NgKO mice. Therefore it will be of interest to determine the nature of MPH-mediated astrocyte activation and how it may modulate behavior in future studies. Taken together these NgKO mice may be useful for the development of better drug treatment to improve cognitive and behavioral impairments.


Subject(s)
Cognition Disorders/drug therapy , Mental Disorders/drug therapy , Methylphenidate/therapeutic use , Neurogranin/genetics , Animals , Cognition Disorders/genetics , Female , Gene Expression , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Interpersonal Relations , Maze Learning , Mental Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Swimming
3.
Neuroscience ; 178: 1-12, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21256930

ABSTRACT

Calmodulin (CaM) and neurogranin (Ng) are two abundant neuronal proteins in the forebrain whose interactions are implicated in the enhancement of synaptic plasticity. To gain further insight into the actions of these two proteins we investigated whether they co-localize in principle neurons and whether they respond to high frequency stimulation in a coordinated fashion. Immunohistochemical staining of CaM and Ng in mouse hippocampal slices revealed that CaM was highly concentrated in the nucleus of CA1 pyramidal neurons, whereas Ng was more broadly localized throughout the soma and dendrites. The asymmetrical localization of CaM in the nucleus of pyramidal neurons was in sharp contrast to the distribution observed in pyramidal cells of the neighboring subiculum, where CaM was uniformly localized throughout the soma and dendrites. The somatic concentrations of CaM and Ng in CA1 pyramidal neurons were approximately 10- and two-fold greater than observed in the dendrites, respectively. High frequency stimulation (HFS) of hippocampal slices promoted mobilization of CaM and Ng from soma to dendrites. These responses were spatially restricted to the area close to the site of stimulation and were inhibited by the N-methyl-D-asparate receptor antagonist 2-amino-5-phosphonopentanoic acid. Furthermore, HFS failed to promote translocation of CaM from soma to dendrites of slices from Ng knockout mice, which also exhibited deficits in HFS-induced long-term potentiation. Translocated CaM and Ng exhibited distinct puncta decorating the apical dendrites of pyramidal neurons and appeared to be concentrated in dendritic spines. These findings suggest that mobilization of CaM and Ng to stimulated dendritic spines may enhance synaptic efficacy by increasing and prolonging the Ca2+ transients and activation of Ca2+/CaM-dependent enzymes.


Subject(s)
CA1 Region, Hippocampal/metabolism , Calmodulin/metabolism , Dendrites/metabolism , Neurogranin/metabolism , Protein Transport/drug effects , Protein Transport/genetics , Pyramidal Cells/metabolism , 2-Amino-5-phosphonovalerate/pharmacology , Animals , CA1 Region, Hippocampal/drug effects , Electric Stimulation/methods , Female , Hippocampus/metabolism , In Vitro Techniques , Long-Term Potentiation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogranin/genetics
4.
J Neurosci Res ; 70(3): 298-308, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391589

ABSTRACT

Reactive oxygen species (ROS) can have deleterious effects for both normal aging and Alzheimer's disease (AD). We examined the hypothesis that synapses undergoing long-term potentiation (LTP) are preferentially at risk for ROS-mediated oxidative stress during aging. We observed age-dependent deficits in LTP induced by a high-frequency stimulation (HFS) protocol in the CA1 region of hippocampus from C57BL/6 mice. There was a significant difference between LTP measured over 60 min in young (1-2 months) and old (23-26 months) mice. In oxidative stress studies, exogenous H(2)O(2) (580 micro M) significantly inhibited LTP in young mice; a similar dose of H(2)O(2) failed to inhibit LTP in slices from adult (2-4 months) or from old mice. The results show that there are significant deficits in LTP in aging mice, but such deficits are insensitive to H(2)O(2). Western immunoblotting studies in young mice show that the relative levels of autophosphorylated alpha-Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) are unchanged in hippocampal CA1 treated with H(2)O(2) relative to untreated controls. However with aging, there is a significant enhancement in the levels of autophosphorylated CaMKII in H(2)O(2)-treated CA1 of older mice. Phosphorylation of RC3/neurogranin (Ng) by protein kinase C (PKC) is decreased in CA1 in response to H(2)O(2) treatment, irrespective of age. We propose that, during aging, enhanced local release of H(2)O(2) from mitochondria may induce a compensatory "ceiling" effect at synapses, so that the levels of autophosphorylated alpha CaMKII are aberrantly saturated, leading to alterations in synaptic plasticity.


Subject(s)
Aging/metabolism , Alzheimer Disease/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Hippocampus/enzymology , Hydrogen Peroxide/metabolism , Long-Term Potentiation/physiology , Oxidative Stress/physiology , Aging/drug effects , Alzheimer Disease/physiopathology , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calmodulin-Binding Proteins/drug effects , Calmodulin-Binding Proteins/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/drug effects , Hippocampus/physiopathology , Hydrogen Peroxide/pharmacology , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Neurogranin , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Synapses/drug effects , Synapses/enzymology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
5.
J Biol Chem ; 276(5): 3098-105, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11060308

ABSTRACT

S-Nitrosoglutathione (GSNO) undergoes spontaneous degradation that generates several nitrogen-containing compounds and oxidized glutathione derivatives. We identified glutathione sulfonic acid, glutathione disulfide S-oxide (GS(O)SG), glutathione disulfide S-dioxide, and GSSG as the major decomposition products of GSNO. Each of these compounds and GSNO were tested for their efficacies to modify rat brain neurogranin/RC3 (Ng) and neuromodulin/GAP-43 (Nm). Among them, GS(O)SG was found to be the most potent in causing glutathiolation of both proteins; four glutathiones were incorporated into the four Cys residues of Ng, and two were incorporated into the two Cys residues of Nm. Ng and Nm are two in vivo substrates of protein kinase C; their phosphorylations by protein kinase C attenuate the binding affinities of both proteins for calmodulin. When compared with their respective unmodified forms, the glutathiolated Ng was a poorer substrate and glutathiolated Nm a better substrate for protein kinase C. Glutathiolation of these two proteins caused no change in their binding affinities for calmodulin. Treatment of [(35)S]cysteine-labeled rat brain slices with xanthine/xanthine oxidase or a combination of xanthine/xanthine oxidase with sodium nitroprusside resulted in an increase in cellular level of GS(O)SG. These treatments, as well as those by other oxidants, all resulted in an increase in thiolation of proteins; among them, thiolation of Ng was positively identified by immunoprecipitation. These results show that GS(O)SG is one of the most potent glutathiolating agents generated upon oxidative stress.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Glutathione/analogs & derivatives , Glutathione/metabolism , Nerve Tissue Proteins/metabolism , Nitroso Compounds/metabolism , Proteins/metabolism , Animals , Brain/metabolism , Calmodulin/metabolism , GAP-43 Protein/metabolism , Glutathione Disulfide/metabolism , In Vitro Techniques , Male , Neurogranin , Oxidants/pharmacology , Oxidation-Reduction , Phosphorylation , Precipitin Tests , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , S-Nitrosoglutathione
6.
Hippocampus ; 11(6): 763-75, 2001.
Article in English | MEDLINE | ID: mdl-11811671

ABSTRACT

Neurogranin/RC3 is a protein that binds calmodulin and serves as a substrate for protein kinase C. Neuronally distributed in the hippocampus and forebrain, neurogranin is highly expressed in dendritic spines of hippocampal pyramidal cells, implicating this protein in long-term potentiation and in learning and memory processes. Null mutation of the neurogranin gene Ng generated viable knockout mice for analysis of the behavioral phenotype resulting from the absence of neurogranin protein. Ng -/- mice were normal on measures of general health, neurological reflexes, sensory abilities, and motor functions, as compared to wild type littermate controls. On the Morris water task, Ng -/- mice failed to reach acquisition criterion on the hidden platform test and did not show selective search on the probe trial. In the Barnes circular maze, another test for spatial navigation learning, Ng -/- mice showed impairments on some components of transfer, but normal performance on time spent around the target hole. Abnormal and idiosyncratic behaviors were detected, that appeared to represent an anxiogenic phenotype in Ng -/- mice, as measured in the light<-->dark exploration test and the open field center time parameter. These findings of apparent deficits in spatial learning and anxiety-like tendencies in Ng -/- support a role for neurogranin in the hippocampally-mediated interaction between stress and performance.


Subject(s)
Anxiety/physiopathology , Calmodulin-Binding Proteins/physiology , Maze Learning/physiology , Nerve Tissue Proteins/physiology , Animals , Behavior, Animal/physiology , Calmodulin-Binding Proteins/genetics , Darkness , Exploratory Behavior/physiology , Female , Light , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nervous System Physiological Phenomena , Neurogranin , Reflex/physiology , Sensation/physiology , Swimming
7.
Proc Natl Acad Sci U S A ; 97(21): 11232-7, 2000 Oct 10.
Article in English | MEDLINE | ID: mdl-11016969

ABSTRACT

Neurogranin/RC3 is a neural-specific Ca(2+)-sensitive calmodulin (CaM)-binding protein whose CaM-binding affinity is modulated by phosphorylation and oxidation. Here we show that deletion of the Ng gene in mice did not result in obvious developmental or neuroanatomical abnormalities but caused an impairment of spatial learning and changes in hippocampal short- and long-term plasticity (paired-pulse depression, synaptic fatigue, long-term potentiation induction). These deficits were accompanied by a decreased basal level of the activated Ca(2+)/CaM-dependent kinase II (CaMKII) ( approximately 60% of wild type). Furthermore, hippocampal slices of the mutant mice displayed a reduced ability to generate activated CaMKII after stimulation of protein phosphorylation and oxidation by treatments with okadaic acid and sodium nitroprusside, respectively. These results indicate a central role of Ng in the regulation of CaMKII activity with decisive influences on synaptic plasticity and spatial learning.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin-Binding Proteins/physiology , Learning/physiology , Nerve Tissue Proteins/physiology , Neuronal Plasticity , Animals , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calmodulin-Binding Proteins/genetics , DNA Primers , Female , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neurogranin , Signal Transduction
8.
Biochemistry ; 39(24): 7291-9, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10852729

ABSTRACT

Neurogranin (NG) binding of calmodulin (CaM) at its IQ domain is sensitive to Ca(2+) concentration and to modifications by protein kinase C (PKC) and oxidants. The PKC phosphorylation site of NG is within the IQ domain whereas the four oxidant-sensitive Cys residues are outside this region. These Cys residues were oxidized forming two pairs of intramolecular disulfides, and could also be glutathiolated by S-nitrosoglutathione resulting in the incorporation of four glutathiones per NG. Circular dichroism (CD) showed that modification of NG by phosphorylation, oxidation forming intramolecular disulfides, or glutathiolation did not affect the alpha-helical content of this protein. Mutation of the four Cys residues [Cys(-)-NG] to Gly and Ser did not affect the alpha-helical content either. Interaction of CaM with the reduced (red)-, glutathiolated (GS)-, or Cys(-)-NG in the Ca(2+)-free solution resulted in an increase in the alpha-helicity determined by their CD spectra, but relatively little change was seen with the oxidized NG (ox-NG) or phosphorylated NG (PO(4)-NG). The binding affinities between the various modified forms of NG and CaM were determined by CD spectrometry and sedimentation equilibrium: their affinities were Cys(-)-NG > red-NG, GS-NG > ox-NG > PO(4)-NG. Unlike Cys(-)-, red-, and GS-NG, neither ox- nor PO(4)-NG bound to a CaM-affinity column. Thus, both oxidation of NG to form intramolecular disulfides and phosphorylation of NG by PKC are effective in modulating the intracellular level of CaM. These results indicate that modification of NG to form intramolecular disulfides outside the IQ domain provides an alternative mechanism for regulation of its binding affinity to CaM.


Subject(s)
Brain/metabolism , Calcium/pharmacology , Calmodulin-Binding Proteins/metabolism , Calmodulin/metabolism , Nerve Tissue Proteins/metabolism , Animals , Binding Sites , Calmodulin-Binding Proteins/chemistry , Calmodulin-Binding Proteins/genetics , Circular Dichroism , Cysteine/genetics , Cysteine/metabolism , Disulfides/chemistry , Glutathione/analogs & derivatives , Glutathione/metabolism , Mass Spectrometry , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurogranin , Nitroso Compounds/metabolism , Oxidation-Reduction , Phosphorylation , Protein Binding , Protein Kinase C/metabolism , Protein Structure, Secondary , Rats , S-Nitrosoglutathione , Ultracentrifugation
9.
J Neurochem ; 74(1): 392-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10617144

ABSTRACT

A 20-kDa DNA-binding protein that binds the AT-rich sequences within the promoters of the brain-specific protein kinase C (PKC) gamma and neurogranin/RC3 genes has been characterized as chromosomal nonhistone high-mobility-group protein (HMG)-I. This protein is a substrate of PKC alpha, beta, gamma, and delta but is poorly phosphorylated by PKC epsilon and zeta. Two major (Ser44 and Ser64) and four minor phosphorylation sites have been identified. The extents of phosphorylation of Ser44 and Ser64 were 1:1, whereas those of the four minor sites all together were <30% of the major one. These PKC phosphorylation sites are distinct from those phosphorylated by cdc2 kinase, which phosphorylates Thr53 and Thr78. Phosphorylation of HMG-I by PKC resulted in a reduction of DNA-binding affinity by 28-fold as compared with 12-fold caused by the phosphorylation with cdc2 kinase. HMG-I could be additively phosphorylated by cdc2 kinase and PKC, and the resulting doubly phosphorylated protein exhibited a >100-fold reduction in binding affinity. The two cdc2 kinase phosphorylation sites of HMG-I are adjacent to the N terminus of two of the three predicted DNA-binding domains. In comparison, one of the major PKC phosphorylation sites, Ser64, is adjacent to the C terminus of the second DNA-binding domain, whereas Ser44 is located within the spanning region between the first and second DNA-binding domains. The current results suggest that phosphorylation of the mammalian HMG-I by PKC alone or in combination with cdc2 kinase provides an effective mechanism for the regulation of HMG-I function.


Subject(s)
Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , High Mobility Group Proteins/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic/physiology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Animals , Binding, Competitive , CDC2 Protein Kinase/physiology , DNA/metabolism , Neurogranin , Phosphorylation , Rats
10.
J Neurochem ; 72(3): 1294-306, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10037503

ABSTRACT

The in vivo state of phosphorylation and the modification of two Cys residues of neuromodulin/ GAP-43 (Nm) were analyzed by electrospray ionization-mass spectrometry (ES-MS). The protein was purified from rat brain with homogenization buffer containing 1% Nonidet P-40, protease inhibitors, protein phosphatase inhibitors, and sulfhydryl reagent, 4-vinylpyridine. Nm was purified by HPLC and ion-exchange chromatography, and the various fractions were identified by ES-MS as unphosphorylated and mono-, di-, tri-, and tetraphosphorylated species. All of these Nm species contained 2 mol of added 4-vinylpyridine per mol of Nm, suggesting that the two Cys residues are in the reduced form in the brain. In vivo, the majority of Nm is in the phosphorylated form (approximately 80%), of which the levels of the mono- and diphospho forms are higher than those of the tri- and tetraphospho species. Four in vivo phosphorylation sites, Ser41, Thr95, Ser142, and Thr172, were identified by amino acid sequencing and tandem ES-MS of the peptides derived from Lys-C endoproteinase digestion. Among these sites, only Ser41 is a known target of PKC, whereas the kinases responsible for the phosphorylation of the other three novel sites are unknown. Hypoxia/ischemia caused a preferential dephosphorylation of Ser41 and Thr172, whereas Thr95 is the least susceptible to dephosphorylation.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , GAP-43 Protein/metabolism , Hypoxia, Brain/metabolism , Amino Acid Sequence , Animals , Brain/pathology , Chromatography, High Pressure Liquid , GAP-43 Protein/chemistry , Hydrolysis , Mass Spectrometry , Molecular Sequence Data , Peptide Mapping , Peptides/analysis , Peptides/isolation & purification , Phosphorylation , Rats , Rats, Sprague-Dawley
11.
J Biol Chem ; 274(3): 1294-300, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9880498

ABSTRACT

Neurogranin/RC3 (Ng), a postsynaptic neuronal protein kinase C (PKC) substrate, binds calmodulin (CaM) at low level of Ca2+. In vitro, rat brain Ng can be oxidized by nitric oxide (NO) donors and by oxidants to form an intramolecular disulfide bond with resulting downward mobility shift on nonreducing SDS-polyacrylamide gel electrophoresis. The oxidized Ng, as compared with the reduced form, is a poorer substrate of PKC but like the PKC-phosphorylated Ng has a lower affinity for CaM than the reduced form. To investigate the physiological relevance of Ng oxidation, we tested the effects of neurotransmitter, N-methyl-D-aspartate (NMDA), NO donors, and other oxidants such as hydrogen peroxide and oxidized glutathione on the oxidation of this protein in rat brain slices. Western blot analysis showed that the NMDA-induced oxidation of Ng was rapid and transient, it reached maximum within 3-5 min and declined to base line in 30 min. The response was dose-dependent (EC50 approximately 100 microM) and could be blocked by NMDA-receptor antagonist 2-amino-5-phosphonovaleric acid and by NO synthase inhibitor NG-nitro-L-arginine methyl ester and NG-monomethyl-L-arginine. Ng was oxidized by NO donors, sodium nitroprusside, S-nitroso-N-acetylpenicillamine, and S-nitrosoglutathione, and H2O2 at concentrations less than 0.5 mM. Oxidation of Ng in brain slices induced by sodium nitroprusside could be reversed by dithiothreitol, ascorbic acid, or reduced glutathione. Reversible oxidation and reduction of Ng were also observed in rat brain extracts, in which oxidation was enhanced by Ca2+ and the oxidized Ng could be reduced by NADPH or reduced glutathione. These results suggest that redox of Ng is involved in the NMDA-mediated signaling pathway and that there are enzymes catalyzing the oxidation and reduction of Ng in the brain. We speculate that the redox state of Ng, similar to the state of phosphorylation of this protein, may regulate the level of CaM, which in turn modulates the activities of CaM-dependent enzymes in the neurons.


Subject(s)
Brain/metabolism , Calmodulin-Binding Proteins/metabolism , Excitatory Amino Acid Agonists/pharmacology , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/metabolism , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Brain/drug effects , Enzyme Inhibitors/pharmacology , Glutathione/analogs & derivatives , Glutathione/metabolism , Glutathione Disulfide/metabolism , In Vitro Techniques , NG-Nitroarginine Methyl Ester/pharmacology , Neurogranin , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Nitroso Compounds/metabolism , Oxidation-Reduction , Penicillamine/analogs & derivatives , Penicillamine/metabolism , Rats , Rats, Sprague-Dawley , S-Nitrosoglutathione , omega-N-Methylarginine/pharmacology
12.
Eur J Biochem ; 245(3): 684-92, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9183006

ABSTRACT

We show that, in vitro, Ca2+-dependent protein kinase C (PKC) phosphorylates recombinant murine p53 protein on several residues contained within a conserved basic region of 25 amino acids, located in the C-terminal part of the protein. Accordingly, synthetic p53-(357-381)-peptide is phosphorylated by PKC at multiple Ser and Thr residues, including Ser360, Thr365, Ser370 and Thr377. We also establish that p53-(357-381)-peptide at micromolar concentrations has the ability to stimulate sequence-specific DNA binding by p53. That stimulation is lost upon phosphorylation by PKC. To further characterise the mechanisms that regulate PKC-dependent phosphorylation of p53-(357-381)-peptide, the phosphorylation of recombinant p53 and p53-(357-381)-peptide by PKC were compared. The results suggest that phosphorylation of full-length p53 on the C-terminal PKC sites is highly dependent on the accessibility of the phosphorylation sites and that a domain on p53 distinct from p53-(357-381)-peptide is involved in binding PKC. Accordingly, we have identified a conserved 27-amino-acid peptide, p53-(320-346)-peptide, within the C-terminal region of p53 and adjacent to residues 357-381 that interacts with PKC in vitro. The interaction between p53-(320-346)-peptide and PKC inhibits PKC autophosphorylation and the phosphorylation of substrates, including p53-(357-381)-peptide, neurogranin and histone H1. Conventional Ca2+-dependent PKC alpha, beta and gamma and the catalytic fragment of PKC (PKM) were nearly equally susceptible to inhibition by p53-(320-346)-peptide. The Ca2+-independent PKC delta was much less sensitive to inhibition. The significance of these findings for understanding the in vivo phosphorylation of p53 by PKC are discussed.


Subject(s)
Protein Kinase C/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Binding Sites , Molecular Sequence Data , Phosphorylation , Recombinant Proteins/metabolism
13.
J Biol Chem ; 271(46): 28798-804, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8910523

ABSTRACT

Neurogranin (Ng) is a neuron-specific protein kinase C-selective substrate, which binds calmodulin (CaM) in the dephosphorylated form at low levels of Ca2+. This protein contains redox active Cys residues that are readily oxidized by several nitric oxide (NO) donors and other oxidants to form intramolecular disulfide. Identification of the Cys residues of rat brain Ng, Cys3, Cys4, Cys9, and Cys51, involved in NO-mediated intramolecular disulfide bridge formation was examined by site-directed mutagenesis. Mutation of all four Cys residues or single mutation of Cys51 blocked the oxidant-mediated intramolecular disulfide formation as monitored by the downward mobility shift under nonreducing SDS-polyacrylamide gel electrophoresis. Single mutation of Cys3, Cys4, or Cys9 or double mutation of any pair of these three Cys residues did not block such intramolecular disulfide formation, although the rates of oxidation of these mutant proteins were different. Thus, Cys51 is an essential pairing partner in NO-mediated intramolecular disulfide formation in Ng. Cys3, Cys4, and Cys9 individually could pair with Cys51, and the order of reactivity was Cys9 > Cys4 > Cys3, suggesting that Cys9 and Cys51 form the preferential disulfide bridge. In all cases tested, the intramolecularly disulfide bridged Ng proteins displayed dramatically attenuated CaM-binding affinity and approximately 2-3-fold weaker protein kinase C substrate phosphorylation activity. The data indicate that the N-terminal Cys3, Cys4, and Cys9 are in close proximity to the C-terminal Cys51 in solution. The disulfide bridge between the N- and C-terminal domains of Ng renders the central CaM-binding and phosphorylation site domain in a fixed conformation unfavorable for binding to CaM and as a substrate of protein kinase C.


Subject(s)
Calmodulin-Binding Proteins/chemistry , Cysteine/analysis , Disulfides/chemistry , Nerve Tissue Proteins/chemistry , Nitric Oxide/chemistry , Animals , Calmodulin-Binding Proteins/genetics , Cell Line , Mutagenesis, Site-Directed , Nerve Tissue Proteins/genetics , Neurogranin , Rats
14.
J Biol Chem ; 271(37): 22407-13, 1996 Sep 13.
Article in English | MEDLINE | ID: mdl-8798403

ABSTRACT

Neurogranin (Ng) is a prominent protein kinase C (PKC) substrate which binds calmodulin (CaM) in the absence of Ca2+. Rat brain Ng contains four cysteine residues that were readily oxidized by nitric oxide (NO) donors, 1,1-diethyl-2-hydroxy-2-nitrosohydrazine (DEANO) and sodium nitroprusside, and by oxidants, H2O2 and o-iodosobenzoic acid. NO oxidation of Ng resulted in a conformational change detectable by increased electrophoretic mobility upon SDS-polyacrylamide gel electrophoresis. The NO-mediated mobility shift was reversed by treatment with dithiothreitol and was blocked by modification of Ng sulfhydryl groups with 4-vinylpyridine. Both the nonphosphorylated and PKC-phosphorylated Ng were susceptible to NO oxidation. Modification of Ng by DEANO was blocked by CaM in the absence of Ca2+; while in the presence of Ca2+, CaM did not protect Ng from oxidation by DEANO. CaM also failed to protect DEANO-mediated oxidation of PKC-phosphorylated Ng with or without Ca2+. Oxidation of Ng by the various oxidants apparently resulted in the formation of intramolecular disulfide bond(s) as judged by a reduction of apparent Mr on SDS-polyacrylamide gel electrophoresis; this oxidized form, unlike the reduced form, did not bind to CaM-affinity column. The oxidized Ng was also a poorer substrate for PKC; both the reduced and oxidized forms had similar Km values, but the Vmax of the oxidized form was about one-fourth of the reduced one. When comparing the rate of DEANO-mediated nitrosation of Ng with other sulfhydryl-containing compounds, it became evident that Ng ranked as one of the best NO acceptors among those tested, including serum albumin, glutathione, and dithiothreitol. Ng present in the rat brain synaptosomal preparations was also oxidized by DEANO in a dose-dependent manner when analyzed by immunoblot with a polyclonal antibody against this protein. These results suggest that Ng is a likely target of NO and other oxidants and that oxidation/reduction may serve as a mechanism for controlling both the PKC phosphorylation and the CaM-binding affinity of this protein.


Subject(s)
Brain/metabolism , Calmodulin-Binding Proteins/metabolism , Calmodulin/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide/metabolism , Protein Kinase C/metabolism , Animals , Chromatography, Affinity , Diethylamines/pharmacology , Dithiothreitol/metabolism , Egtazic Acid/pharmacology , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Kinetics , Neurogranin , Nitrogen Oxides , Oxidation-Reduction , Phosphorylation , Rats , Serum Albumin/metabolism , Synaptosomes/metabolism
15.
Arch Biochem Biophys ; 327(1): 131-41, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8615683

ABSTRACT

HASPP28 (heat- and acid-stable phosphoprotein of 28 kDa) has been purified to near homogeneity from the acid-stable protein fraction of rat brain extract. Based on the N-terminal 40 amino acid sequence, a pair of highly degenerate primers was used to generate a 107-bp probe from rat brain RNA by RT-PCR. From the rat brain lambda gt11 library, this probe identified two positive clones that together provided a cDNA of 837 bp with an open reading frame of 546 bp. This cDNA was extended by 3'RACE to 1.2 kb that included a polyadenylation signal and a poly(A) tail. The 180-amino-acid sequence derived from the open reading frame, which did not correspond to any known protein, was predicted to have phosphorylation sites for protein kinase C, casein kinase II (CKII), and protein kinase A. Indeed, both the purified rat brain HASPP28 and the recombinant HASPP28 (rHASPP28) can be phosphorylated by these kinases. Northern blot analysis indicated that HASPP28 was present in all rat tissues tested, including those from the brain, lung, spleen, kidney, liver, heart, and muscle, in decreasing order of abundance. Phosphopeptide analysis of rHASPP28 phosphorylated in vitro by various kinases showed different tryptic patterns on two-dimensional mapping and isoelectric focusing gels. From [32P]PO4-labeled N1E115 neuroblastoma cells, HASPP28 can be immunoprecipitated with a polyclonal antiserum raised against rHASPP28. The immunoprecipitated protein showed a phosphopeptide pattern similar to that of rHASPP28 phosphorylated by CK II in vitro. Furthermore, the immunoprecipitates from cells treated with phorbol 12-myristate 13-acetate or 8-bromo-cAMP did not show any increased phosphorylation over those of untreated ones, and the phosphopeptide patterns of the immunoprecipitates again were similar to that of CK II phosphorylated protein. These results suggest that HASPP28 is a novel phosphoprotein that can be phosphorylated by several kinases in vitro. In intact cells, CK II seems to be solely responsible for the phosphorylation of HASPP28.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Casein Kinase II , Chromatography, High Pressure Liquid , Cloning, Molecular , DNA Primers , DNA, Complementary , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Gene Library , Immunoblotting , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/isolation & purification , Open Reading Frames , Organ Specificity , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptide Mapping , Phosphoproteins/chemistry , Phosphoproteins/isolation & purification , Phosphorylation , Polymerase Chain Reaction , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Restriction Mapping , Substrate Specificity
16.
Arch Biochem Biophys ; 326(2): 193-201, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8611023

ABSTRACT

The myristoylated aline-rich protein kinase C substrate (MARCKS) is a peripheral membrane protein that undergoes phosphorylation-dependent translocation between membrane and cytosol. MARCKS binds to acidic phospholipids with high affinity (Kd less than 0.5 microM) but binds poorly to neutral phospholipids. Although interaction of MARCKS with acidic phospholipids lacks specificity when determined by binding assay, these phospholipids exert distinctive effects on the phosphorylation of this protein by protein kinase C (PKC). Preincubation of MARCKS with phosphatidylserine (PS) or phosphatidylglycerol enhanced the phosphorylation; whereas with phosphatidic acid, phosphatidylinositol (PI), phosphatidylinositol-4-phosphate, or phosphatidylinositol-4,5-biphosphate inhibited the phosphorylation of this substrate by PKC. Phosphoinositide inhibition of MARCKS phosphorylation was apparently directed at the substrate rather than at the kinase as the phosphorylation of two other phospholipid-binding PKC substrates, neuromodulin and neurogranin, exhibited different responses from those of MARCKS. Furthermore, the inhibition of phosphoinositides on MARCKS phosphorylation was seen with PKC isozymes alpha, beta, gamma, and delta and with the catalytic fragment of PKC, protein kinase M. A 25-amino-acid synthetic peptide corresponding to the phosphorylation site domain (PSD) of MARCKS, but not to the myristoylated N-terminal peptide, competed equally effectively with MARCKS in binding to either PS- or PI-containing vesicles, suggesting that both phospholipids bind to the PSD of MARCKS. Binding of PI to MARCKS inhibited PKC phosphorylation of all three phosphorylation sites. These results suggest that phosphoinositides and PS bind at different residues within the MARCKS PSD, so that the resulting phospholipid/MARCKS complexes are differentially phosphorylated by PKC.


Subject(s)
Intracellular Signaling Peptides and Proteins , Membrane Proteins , Protein Kinase C/metabolism , Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Brain/metabolism , In Vitro Techniques , Isoenzymes/metabolism , Kinetics , Membrane Lipids/metabolism , Molecular Sequence Data , Myristic Acids/metabolism , Myristoylated Alanine-Rich C Kinase Substrate , Peptide Fragments/chemical synthesis , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphatidylinositols/metabolism , Phospholipids/metabolism , Phosphorylation , Protein Binding , Proteins/genetics , Rats , Substrate Specificity
17.
Cell Signal ; 7(7): 679-85, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8519597

ABSTRACT

Diacylglycerols (DG) derived from brain phosphatidylinositol (PI) and phosphatidylcholine (PC) and synthetic 1,2-dioleoylglycerol (diC18:1) and 1,2-dioctanoylglycerol (diC8) were tested for their efficacy in stimulating PKC-catalyzed phosphorylation of three physiological substrates in the brain, namely, MARCKS, neuromodulin (Nm), and neurogranin (Ng). The A0.5 of these DGs for PKC were variable dependent on the protein substrates; the values were lowest with MARCKS and highest with Ng. With Ng as a substrate the A0.5 of these DGs for PKC gamma were PI- and PC-DGs < diC18:1 < diC8. Both PI- and PC-DGs, in spite of their differences in unsaturated fatty acids content, were similarly effective in stimulating PKC. Since the phosphorylation of MARCKS, as compared to those of Nm and Ng, has the lowest A0.5 with the various DGs, it seems that among these three PKC substrates MARCKS is most readily phosphorylated by PKCs following DG formation in vivo.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Diglycerides/pharmacology , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Membrane Proteins , Nerve Tissue Proteins/metabolism , Protein Kinase C/metabolism , Proteins/metabolism , Animals , Brain/drug effects , Brain/metabolism , Enzyme Activation/drug effects , GAP-43 Protein , Myristoylated Alanine-Rich C Kinase Substrate , Neurogranin , Phosphoproteins/metabolism , Phosphorylation , Rats
18.
J Biol Chem ; 270(17): 10314-22, 1995 Apr 28.
Article in English | MEDLINE | ID: mdl-7730337

ABSTRACT

A 13-kilobase pair genomic DNA encoding a 78-amino acid brain-specific calmodulin-binding protein kinase C (PKC) substrate, neurogranin (Ng/RC3; also known as RC3 or p17), has been sequenced. The Ng/RC3 gene is composed of four exons and three introns, with the protein-coding region located in the first and second exons. This gene was found to have multiple transcriptional start sites clustered within 20 base pairs (bp); it lacks the TATA, GC, and CCAAT boxes in the proximal upstream region of the start sites. The promoter activity was characterized by transfection of 293 cells with nested deletion mutants of the 5'-flanking region fused to the luciferase reporter gene. A minimal construct containing bp +11 to +256 was nearly as active as that covering bp -1508 to +256, whereas a shorter one covering bp +40 to +256 had a greatly reduced activity. Between bp +11 and +40 lies a 12-nucleotide sequence (CCCCGCCCACCC) containing overlapping binding sites for AP2 (CCGCCCACCC) and SP1 (CCCGCC); this region may be important for conferring the basal transcriptional activity of the Ng/RC3 gene. The expression of a Ng/RC3-luciferase fusion construct (-1508/+256) in transfected 293 cells was stimulated by phorbol 12-myristate 13-acetate (PMA), but not by cAMP, arachidonic acid, vitamin D, retinoic acid, or thyroxines T3 and T4. PMA caused a 2-4-fold stimulation of all the reporter gene constructs ranging from +11/+256 to -1508/+256. The stimulatory effects of PMA could be magnified by cotransfection with both Ca(2+)-dependent and -independent phorbol ester-binding PKC-alpha, -beta I, -beta II, -gamma, -delta, and -epsilon cDNAs, but not by non-phorbol ester-binding PKC-zeta cDNA. The Ng/RC3 and PKC-gamma genes have a similar expression pattern in the brain during development. These two genes share at least four conserved sequence segments 1.5 kilobase pair upstream from their transcriptional start sites and a gross similarity in that they possess several AT-rich segments within bp -550 to -950. A near homogeneous 20-kDa DNA-binding protein purified from rat brain was able to bind to these AT-rich regions of both Ng/RC3 and PKC-gamma genes with footprints containing ATTA, ATAA, and AATA sequences.


Subject(s)
Calmodulin-Binding Proteins/genetics , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Neurons/enzymology , Protein Kinase C/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Transformed , Cloning, Molecular , DNA , DNA-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Neurogranin , Promoter Regions, Genetic , Protein Kinase C/metabolism , Rats , Sequence Homology, Nucleic Acid , Substrate Specificity , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic , Transfection
19.
Biochemistry ; 34(10): 3446-54, 1995 Mar 14.
Article in English | MEDLINE | ID: mdl-7533539

ABSTRACT

Mixed micelles containing Nonidet P40 (NP-40) (829 microM or 4.8 mM), phosphatidylserine (PS) (14.5 or 8 mol%), and 1,2-diacylglycerol (DG) (0.5 or 1 mol%) when preincubated with protein kinase C (PKC) assay mixture containing cationic substrate and CaCl2 (400 microM) formed aggregates in a time-, temperature-, and substrate concentration-dependent manner with a t1/2 approximately 3-12 min (22 degrees C). Concomitant with the formation of these aggregates there was a substantial loss of substrate phosphorylation catalyzed by the Ca(2+)-dependent PKC alpha, beta, and gamma but not the Ca(2+)-independent PKC, delta and epsilon. All cationic PKC substrates tested, neurogranin peptide analog, neurogranin, and histone III-S, formed aggregates with PS/DG/NP-40/Ca2+ mixed micelles in a time-dependent fashion. The poly(cationic-anionic) PKC substrate protamine sulfate also forms aggregates with the mixed micelles in the presence of Ca2+, but without affecting the substrate phosphorylation by the kinase. Under similar conditions, but at 4 degrees C, neither aggregation nor loss of cationic substrate phosphorylation was observed. Another nonionic detergent, octyl glucoside, behaved similarly to NP-40. Phosphatidylinositol (PI) and phosphatidylglycerol like PS, were effective in forming aggregates with NP-40/cationic polypeptide/DG/Ca2+ as monitored by light scattering, yet without affecting substrate phosphorylation. Phosphorylation of cationic substrates by M-kinase, derived from trypsinized PKC beta, was also greatly diminished by the aggregation. In contrast, [3H]phorbol 12,13-dibutyrate binding to PKC beta was unaffected. Formation of the aggregates that were selectively utilized by the Ca(2+)-independent PKCs was dependent on the ratio of cationic substrate to the number of mixed micelles.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Isoenzymes/metabolism , Peptides/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Calcium , Calmodulin-Binding Proteins/genetics , Cattle , Detergents , Diglycerides , In Vitro Techniques , Micelles , Molecular Sequence Data , Myelin Basic Protein/genetics , Nerve Tissue Proteins/genetics , Neurogranin , Octoxynol , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptides/chemistry , Phosphatidylserines , Phosphorylation , Polyethylene Glycols , Rats
20.
Arch Biochem Biophys ; 316(2): 673-9, 1995 Feb 01.
Article in English | MEDLINE | ID: mdl-7864622

ABSTRACT

Neurogranin, neuromodulin, and MARCKS are among the most prominent substrates of protein kinase C (PKC) in the mammalian brain. These phosphoproteins were dephosphorylated by three isoforms of rat brain calcineurin, also known as calmodulin (CaM)-dependent protein phosphatase (CaMPP). The three CaMPP isozymes dephosphorylate neurogranin, the most favorable substrate among the three tested, with subtle differences in their responses to divalent metal ions, Mn2+ and Ni2+. Dephosphorylation of neurogranin by all three CaMPP isozymes, CaMPP-1, -2, and -3, were stimulated to a higher extent by Mn2+ than by Ni2+ in the presence of CaM and Ca2+. The Km values of neurogranin in the presence of Mn2+ were lower than those in the presence of Ni2+ for CaMPP-1 and -2, but that for CaMPP-3 was comparable with either divalent metal ion. The Vmax values were higher in the presence of Mn2+ than those of Ni2+ for all three isozymes. Neurogranin and neuromodulin, both phosphorylated by PKC at a single site, were dephosphorylated completely by CaMPP; however, MARCKS, phosphorylated by PKC at three sites, was partially dephosphorylated by this phosphatase. A higher extent of dephosphorylation of MARCKS could be achieved by the combination of CaMPP and protein phosphatase 2A and a complete dephosphorylation of this protein was observed with protein phosphatase 1. Protein phosphatase 1 and 2A were also effective in a complete dephosphorylation of neurogranin and neuromodulin. Amino acid sequence analysis of the tryptic phosphopeptides derived from MARCKS dephosphorylated by CaMPP and protein phosphatase 2A revealed that the former preferentially dephosphorylated Ser155 and the latter Ser162 of rat brain MARCKS. Both phosphatases dephosphorylated poorly of Ser151. Because of the high concentration of CaMPP in the brain and the colocalization of this phosphatase with major PKC substrates in the various brain regions, it is likely that CaMPP is a phosphatase with potential to reverse the action of PKC.


Subject(s)
Brain/metabolism , Calmodulin-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Membrane Proteins , Nerve Tissue Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Proteins/metabolism , Amino Acid Sequence , Animals , Brain/enzymology , Calcineurin , Calmodulin/pharmacology , Cations, Divalent/pharmacology , GAP-43 Protein , Isoenzymes/metabolism , Molecular Sequence Data , Myristoylated Alanine-Rich C Kinase Substrate , Neurogranin , Phosphoprotein Phosphatases/drug effects , Protein Kinase C/metabolism , Protein Phosphatase 1 , Protein Phosphatase 2 , Rats , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL