Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Gene Regul Mech ; 1864(2): 194629, 2021 02.
Article in English | MEDLINE | ID: mdl-32890768

ABSTRACT

Histone post-translational modifications are essential for the regulation of gene expression in eukaryotes. Gcn5 (KAT2A) is a histone acetyltransferase that catalyzes the post-translational modification at multiple positions of histone H3 through the transfer of acetyl groups to the free amino group of lysine residues. Gcn5 catalyzes histone acetylation in the context of a HAT module containing the Ada2, Ada3 and Sgf29 subunits of the parent megadalton SAGA transcriptional coactivator complex. Biochemical and structural studies have elucidated mechanisms for Gcn5's acetyl- and other acyltransferase activities on histone substrates, for histone H3 phosphorylation and histone H3 methylation crosstalks with histone H3 acetylation, and for how Ada2 increases Gcn5's histone acetyltransferase activity. Other studies have identified Ada2 isoforms in SAGA-related complexes and characterized variant Gcn5 HAT modules containing these Ada2 isoforms. In this review, we highlight biochemical and structural studies of Gcn5 and its functional interactions with Ada2, Ada3 and Sgf29.


Subject(s)
Histone Acetyltransferases/metabolism , Multienzyme Complexes/metabolism , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins/metabolism , Acetylation , Cryoelectron Microscopy , Histone Acetyltransferases/ultrastructure , Histones/metabolism , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Methylation , Multienzyme Complexes/ultrastructure , Phosphorylation , Saccharomyces cerevisiae Proteins/ultrastructure , Transcription Factors/metabolism , Transcription Factors/ultrastructure , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/ultrastructure
2.
Biochim Biophys Acta Bioenerg ; 1861(12): 148290, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32828729

ABSTRACT

We hypothesized that NO• is generated in isolated cardiac mitochondria as the source for ONOO- production during oxidative stress. We monitored generation of ONOO- from guinea pig isolated cardiac mitochondria subjected to excess Ca2+ uptake before adding succinate and determined if ONOO- production was dependent on a nitric oxide synthase (NOS) located in cardiac mitochondria (mtNOS). Mitochondria were suspended in experimental buffer at pH 7.15, and treated with CaCl2 and then the complex II substrate Na-succinate, followed by menadione, a quinone redox cycler, to generate O2•-. L-tyrosine was added to the mitochondrial suspension where it is oxidized by ONOO- to form dityrosine (diTyr) in proportion to the ONOO- present. We found that exposing mitochondria to excess CaCl2 before succinate resulted in an increase in diTyr and amplex red fluorescence (H2O2) signals, indicating that mitochondrial oxidant stress, induced by elevated mtCa2+ and succinate, increased mitochondrial ONOO- production via NO• and O2•-. Changes in mitochondrial ONOO- production dependent on NOS were evidenced by using NOS inhibitors L-NAME/L-NNA, TEMPOL, a superoxide dismutase (SOD) mimetic, and PTIO, a potent global NO• scavenger. L-NAME and L-NNA decreased succinate and menadione-mediated ONOO- production, PTIO decreased production of ONOO-, and TEMPOL decreased ONOO- levels by converting more O2•- to H2O2. Electron microscopy showed immuno-gold labeled iNOS and nNOS in mitochondria isolated from cardiomyocytes and heart tissue. Western blots demonstrated iNOS and nNOS bands in total heart tissue, bands for both iNOS and nNOS in ß-tubulin-free non-purified (crude) mitochondrial preparations, and a prominent iNOS band, but no nNOS band, in purified (Golgi and ER-free) mitochondria. Prior treatment of guinea pigs with lipopolysacharride (LPS) enhanced expression of iNOS in liver mitochondria but not in heart mitochondria. Our results indicate that release of ONOO- into the buffer is dependent both on O2•- released from mitochondria and NO• derived from a mtCa2+-inducible nNOS isoform, possibly attached to mitochondria, and a mtNOS isoform like iNOS that is non-inducible.


Subject(s)
Calcium/pharmacology , Mitochondria, Heart/metabolism , Nitric Oxide/biosynthesis , Oxidative Stress/drug effects , Peroxynitrous Acid/metabolism , Succinic Acid/pharmacology , Animals , Electron Transport/drug effects , Free Radical Scavengers/metabolism , Guinea Pigs , Hydrogen Peroxide/metabolism , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/ultrastructure , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase/ultrastructure , Spectrometry, Fluorescence , Stress, Physiological/drug effects , Superoxide Dismutase/metabolism , Time Factors
3.
Elife ; 82019 12 31.
Article in English | MEDLINE | ID: mdl-31889510

ABSTRACT

Direct activation of the human phospholipase C-γ isozymes (PLC-γ1, -γ2) by tyrosine phosphorylation is fundamental to the control of diverse biological processes, including chemotaxis, platelet aggregation, and adaptive immunity. In turn, aberrant activation of PLC-γ1 and PLC-γ2 is implicated in inflammation, autoimmunity, and cancer. Although structures of isolated domains from PLC-γ isozymes are available, these structures are insufficient to define how release of basal autoinhibition is coupled to phosphorylation-dependent enzyme activation. Here, we describe the first high-resolution structure of a full-length PLC-γ isozyme and use it to underpin a detailed model of their membrane-dependent regulation. Notably, an interlinked set of regulatory domains integrates basal autoinhibition, tyrosine kinase engagement, and additional scaffolding functions with the phosphorylation-dependent, allosteric control of phospholipase activation. The model also explains why mutant forms of the PLC-γ isozymes found in several cancers have a wide spectrum of activities, and highlights how these activities are tuned during disease.


Many enzymes are poised to receive signals from the surrounding environment and translate them into responses inside the cell. One such enzyme is phospholipase C-γ1 (PLC-γ1), which controls how cells grow, divide and migrate.When activating signals are absent, PLC-γ1 usually inhibits its own activity, a mechanism called autoinhibition. This prevents the enzyme from binding to its targets, which are fat molecules known as lipids. When activating signals are present, a phosphate group serves as a 'chemical tag' and is added onto PLC-γ1, allowing the enzyme to bind to lipids.Failure in the regulation of PLC-γ1 or other closely related enzymes may lead to conditions such as cancer, arthritis and Alzheimer's disease. However, it remains unclear how autoinhibition suppresses the activity of the enzyme, and how it is stopped by the addition of the phosphate group.Here, Hajicek et al. determine in great detail the three-dimensional structure of the autoinhibited form of the enzyme using a method known as X-ray crystallography. This reveals that PLC-γ1 has two major lobes: one contains the active site that modifies lipids, and the other sits on top of the active site to prevent lipids from reaching it. The findings suggest that when the phosphate group attaches to PLC-γ1, it triggers a large shape change that shifts the second lobe away from the active site to allow lipids to bind.The three-dimensional structure also helps to understand how mutations identified in certain cancers may activate PLC-γ1. In particular, these mutations disrupt the interactions between elements that usually hold the two lobes together, causing the enzyme to activate more easily.The work by Hajicek et al. provides a framework to understand how cells control PLC-γ1. It is a first step toward designing new drugs that alter the activity of this enzyme, which may ultimately be useful to treat cancer and other diseases.


Subject(s)
Enzyme Activation/genetics , Isoenzymes/ultrastructure , Phospholipase C gamma/ultrastructure , Protein Conformation , Crystallography, X-Ray , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Phospholipase C gamma/chemistry , Phospholipase C gamma/genetics , Phosphorylation/genetics , Protein Domains/genetics , Tyrosine/genetics
4.
J Biotechnol ; 235: 24-31, 2016 Oct 10.
Article in English | MEDLINE | ID: mdl-27067080

ABSTRACT

Hydroxynitrile lyases (HNLs) catalyze the asymmetric addition of HCN to aldehydes producing enantiomerically pure cyanohydrins. These enzymes can be heterologously expressed in large quantities making them interesting candidates for industrial applications. The HNLs from Rosaceae evolved from flavin dependent dehydrogenase/oxidase structures. Here we report the high resolution X-ray structure of the highly glycosylated Prunus amygdalus HNL isoenzyme5 (PaHNL5 V317A) expressed in Aspergillus niger and its complex with benzyl alcohol. A comparison with the structure of isoenzyme PaHNL1 indicates a higher accessibility to the active site and a larger cavity for PaHNL5. Additionally, the PaHNL5 complex structure with benzyl alcohol was compared with the structurally related aryl-alcohol oxidase (AAO). Even though both enzymes contain an FAD-cofactor and histidine residues at crucial positions in the active site, PaHNL5 lacks the oxidoreductase activity. The structures indicate that in PaHNLs benzyl alcohol is bound too far away from the FAD cofactor in order to be oxidized.


Subject(s)
Aldehyde-Lyases , Flavins/metabolism , Plant Proteins , Prunus dulcis/enzymology , Aldehyde-Lyases/chemistry , Aldehyde-Lyases/metabolism , Aldehyde-Lyases/ultrastructure , Isoenzymes/chemistry , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Models, Molecular , Oxidation-Reduction , Plant Proteins/chemistry , Plant Proteins/metabolism , Plant Proteins/ultrastructure
5.
Biochem Biophys Res Commun ; 417(4): 1298-303, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22240025

ABSTRACT

Secretory pathway Ca-ATPases are less characterized mammalian calcium pumps than plasma membrane Ca-ATPases and sarco-endoplasmic reticulum Ca-ATPases. Here we report analysis of molecular evolution, alternative splicing, tissue-specific expression and subcellular localization of the second isoform of the secretory pathway Ca-ATPase (SPCA2), the product of the ATP2C2 gene. The primary structure of SPCA2 from rat duodenum deduced from full-length transcript contains 944 amino acid residues, and exhibits 65% sequence identity with known SPCA1. The rat SPCA2 sequence is also highly homologous to putative human protein KIAA0703, however, the latter seems to have an aberrant N-terminus originating from intron 2. The tissue-specificity of SPCA2 expression is different from ubiquitous SPCA1. Rat SPCA2 transcripts were detected predominantly in gastrointestinal tract, lung, trachea, lactating mammary gland, skin and preputial gland. In the newborn pig, the expression profile is very similar with one remarkable exception: porcine bulbourethral gland gave the strongest signal. Upon overexpression in cultured cells, SPCA2 shows an intracellular distribution with remarkable enrichment in Golgi. However, in vivo SPCA2 may be localized in compartments that differ among various tissues: it is intracellular in epidermis, but enriched in plasma membranes of the intestinal epithelium. Analysis of SPCA2 sequences from various vertebrate species argue that ATP2C2 gene radiated from ATP2C1 (encoding SPCA1) during adaptation of tetrapod ancestors to terrestrial habitats.


Subject(s)
Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Evolution, Molecular , Animals , Calcium-Transporting ATPases/ultrastructure , Intracellular Space/enzymology , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Rats , Swine , Tissue Distribution , Transcription, Genetic
6.
Biosci Biotechnol Biochem ; 75(6): 1160-6, 2011.
Article in English | MEDLINE | ID: mdl-21670507

ABSTRACT

Deblocking aminopeptidase (DAP) is an exoprotease that can release N-terminal amino acids from blocked peptides. Three DAP homologous (TkDAP1, TkDAP2, and TkDAP3) are annotated in the genome data base of Thermococcus kodakarensis KOD1. TkDAP2 and TkDAP3 were identified as proteins that are overexpressed in response to heat and oxidative stress by two-dimensional electrophoresis. In this study, the TkDAP1 and TkDAP2 genes were cloned and expressed in Escherichia coli. The two proteins were purified homogeneity and analyzed by gel filtration chromatography and electron microscopy. TkDAP1 showed two oligomers, which were identified as an octodecimer and a dodecamer. TkDAP2 produced three native forms: octodecimer, dodecamer, and trimer. Dodecamer assembly was the main form in the two proteins. Finally, TkDAP1 was found to have higher deblocking aminopeptidase activity on the substrates of Ac-Leu-pNA and Ac-Ala-Ala-Ala, while TkDAP2 had higher aminopeptidase activity on the substrates of Leu-pNA and Ala-Ala-Ala-pNA.


Subject(s)
Aminopeptidases/metabolism , Archaeal Proteins/metabolism , Isoenzymes/metabolism , Recombinant Proteins/metabolism , Thermococcus/enzymology , Amino Acid Sequence , Aminopeptidases/chemistry , Aminopeptidases/genetics , Aminopeptidases/ultrastructure , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/ultrastructure , Chromatography, Gel , Cloning, Molecular , Electrophoresis, Gel, Two-Dimensional , Escherichia coli , Gene Expression , Hot Temperature , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/ultrastructure , Microscopy, Electron , Molecular Sequence Data , Oligopeptides/metabolism , Plasmids , Polymerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/ultrastructure , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity , Thermococcus/genetics , Transformation, Bacterial
7.
BMC Neurosci ; 12: 16, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21272290

ABSTRACT

BACKGROUND: The Na+,K+-ATPase plays an important role for ion homeostasis in virtually all mammalian cells, including neurons. Despite this, there is as yet little known about the isoform specific distribution in neurons. RESULTS: With help of superresolving stimulated emission depletion microscopy the spatial distribution of Na+,K+-ATPase in dendritic spines of cultured striatum neurons have been dissected. The found compartmentalized distribution provides a strong evidence for the confinement of neuronal Na+,K+-ATPase (α3 isoform) in the postsynaptic region of the spine. CONCLUSIONS: A compartmentalized distribution may have implications for the generation of local sodium gradients within the spine and for the structural and functional interaction between the sodium pump and other synaptic proteins. Superresolution microscopy has thus opened up a new perspective to elucidate the nature of the physiological function, regulation and signaling role of Na+,K+-ATPase from its topological distribution in dendritic spines.


Subject(s)
Dendritic Spines/enzymology , Nanotechnology/methods , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dendritic Spines/ultrastructure , Image Interpretation, Computer-Assisted/instrumentation , Image Interpretation, Computer-Assisted/methods , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/ultrastructure
8.
Cell Tissue Res ; 340(2): 381-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20232214

ABSTRACT

The involvement of nitric oxide (NO) as both pro and anti-inflammatory agent in allergic, airway inflammatory, and asthmatic diseases and the active participation of eosinophils in such ailments have been previously suggested. NO modulates eosinophil number, migration and their survival. The microenvironment of NO synthase (NOS) in subcellular organelles determines its rate and efficiency of catalysis, which in turn influences NO generation at distinct intracellular locales. The present study was undertaken to assess the intracellular distribution of NOS isoforms by transmission electron microscopy followed by morphometric analysis in human and rat eosinophils. Rat eosinophils were explored in parallel, and since they are widely used as model systems to mimic human diseases, a comparative study on NOS localization patterns might provide useful information in deciphering NO role in diverse aspects of eosinophil-related inflammatory ailments. The results demonstrated predominance of neuronal NOS (nNOS) in the eosinophilic granules and even distribution of inducible NOS (iNOS) and nNOS in the cytoplasm and nucleus of human eosinophils. In rat eosinophils, however, iNOS was mainly localized in the eosinophilic granules and nucleus, while nNOS was distributed evenly in cytoplasm and nucleus. Distribution of endothelial NOS (eNOS) in eosinophils was scanty. Differences in NOS isoforms and their localization in human and rat cells might have implications in differential mode of catalysis and functional contribution to eosinophil physiology and pathology, warranting detailed investigations. The present study highlights species-specific differences in the relative abundance and distribution pattern of NOS isoforms in rat and human eosinophils, which should be considered cautiously in interpreting the rat data to humans.


Subject(s)
Eosinophils/enzymology , Eosinophils/ultrastructure , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase/ultrastructure , Animals , Humans , Immunohistochemistry , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Male , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type I/ultrastructure , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type II/ultrastructure , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type III/ultrastructure , Protein Transport , Rats , Rats, Sprague-Dawley , Species Specificity
9.
Arch Biochem Biophys ; 466(1): 31-9, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17764654

ABSTRACT

Phthalate dioxygenase (PDO) is a member of a class of bacterial oxygenases that contain both Rieske [2Fe-2S] and Fe(II) mononuclear centers. Recent crystal structures of several Rieske dioxygenases showed that they exist as alpha(3)beta(3) multimers with subunits arranged head-to-tail in alpha and beta stacked planar rings. The structure of PDO, which consists of only alpha-subunits, remains to be solved. Although similar to other Rieske dioxygenases in many aspects, PDO was shown to differ in the mechanism of catalysis. Gel filtration and analytical centrifugation experiments, supplemented with mass spectrometric analysis (both ESI-MS and ESI-GEMMA), in this work showed a hexameric arrangement of subunits in the PDO multimer. Our proposed model for the subunit arrangement in PDO postulates two alpha(3) planar rings one on top the other, similar to the alpha(3)beta(3) arrangement in other Rieske dioxygenases. Unlike other Rieske dioxygenases, this arrangement brings two Rieske and two mononuclear centers, all on separate subunits, into proximity, allowing their cooperation for catalysis. Potential reasons necessitating this unusual structural arrangement are discussed.


Subject(s)
Isoenzymes/chemistry , Isoenzymes/ultrastructure , Models, Chemical , Models, Molecular , Oxygenases/chemistry , Oxygenases/ultrastructure , Amino Acid Sequence , Dimerization , Molecular Sequence Data , Protein Subunits
10.
Biochem Biophys Res Commun ; 362(4): 965-70, 2007 Nov 03.
Article in English | MEDLINE | ID: mdl-17767923

ABSTRACT

We report here the cloning and characterization of the entire cDNA of a papain-like cysteine protease from a tropical flowering plant. The 1098-bp ORF of the cDNA codify a protease precursor having a signal peptide of 19 amino acids, a cathepsin-L like N-terminal proregion of 114 amino acids, a mature enzyme part of 208 amino acids and a C-terminal proregion of 24 amino acids. The derived amino acid sequence of the mature part tallies with the thermostable cysteine protease Ervatamin-C--as was aimed at. The C-terminal proregion of the protease has altogether a different sequence pattern not observed in other members of the family and it contains a negatively charged helical zone. The three-dimensional model of the precursor, based on the homology modeling and X-ray structure, shows that the extended peptide stretch region of the N-terminal propeptide, covering the interdomain cleft, contains protruding side chains of positively charged residues. This study also indicates that the negatively charged zone of C-terminal propeptide may interact with the positively charged zone of the N-terminal propeptide in a cooperative manner in the maturation process of this enzyme.


Subject(s)
Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/ultrastructure , Models, Chemical , Models, Molecular , Tabernaemontana/metabolism , Amino Acid Sequence , Cloning, Molecular , Computer Simulation , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , DNA, Complementary/genetics , Enzyme Stability , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Molecular Sequence Data , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Proteins/ultrastructure , Protein Conformation , Sequence Analysis, Protein , Sequence Homology, Nucleic Acid , Tabernaemontana/genetics , Temperature
11.
Arch Histol Cytol ; 70(1): 63-78, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17558145

ABSTRACT

C-src deficiency is characterized by osteopetrosis due to impaired bone resorption by hypofunctional osteoclasts and the resultant failure of tooth eruption. In preliminary observations, we frequently encountered erupted molars in c-src deficient mice unlike in other osteopetrotic animals. Here we examine the effects of c-src deficiency on the development of molar teeth with an emphasis on the spatial relation of growing teeth with the surrounding bones. In c-src deficient mice, the magnitude of tooth impaction differed considerably among the types of molars; all maxillary 1st molars were totally impacted deep in the alveolar sockets, whereas most mandibular 1st molars fully erupted into oral cavity. Distribution of osteoclasts in the alveolar bone was identical among all types of molars, and electron microscopy revealed signs of bone resorbing activity in these osteoclasts despite the absence of a ruffled border. From early development, the alveolar space was much narrower in the upper molar tooth germs than in the lower ones in both wild type and homozygous animals, and particularly so in the upper 1st molars. Current observations thus indicate a significant contribution of "hypofunctional osteoclasts" in c-src deficient mice in molar tooth development except for the upper 1st molars, which appear to require highly functional osteoclasts to gain sufficient space for them to grow normally. Taken together, these findings on the seemingly tooth-type specific effects of c-src deficiency on the development and eruption of molar teeth in c-src deficient mice can be attributed to the given differential spatial relation of the respective tooth germs with the surrounding bones in the presence of hypofunctional osteoclasts.


Subject(s)
Molar/growth & development , Proto-Oncogene Proteins pp60(c-src)/deficiency , Tooth Eruption/genetics , Tooth Eruption/physiology , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Acid Phosphatase/ultrastructure , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Female , Fluorescent Antibody Technique, Indirect , Heterozygote , Histocytochemistry , Homozygote , Immunohistochemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Male , Mandible/diagnostic imaging , Maxilla/diagnostic imaging , Mice , Mice, Knockout , Molar/diagnostic imaging , Molar/enzymology , Molar/metabolism , Molar/ultrastructure , Osteoblasts/metabolism , Osteoblasts/ultrastructure , Osteoclasts/metabolism , Osteoclasts/ultrastructure , Periodontal Ligament/metabolism , Periodontal Ligament/ultrastructure , Proto-Oncogene Proteins pp60(c-src)/genetics , Radiography , Tartrate-Resistant Acid Phosphatase , Time Factors , Tooth Germ/embryology , Tooth Germ/metabolism , Tooth Germ/ultrastructure
13.
Free Radic Biol Med ; 42(2): 180-90, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17189824

ABSTRACT

Nox organizer 1 (Noxo1), a p47(phox) homolog, is produced as four isoforms with unique N-terminal PX domains derived by alternative mRNA splicing. We compared the subcellular distribution of these isoforms or their isolated PX domains produced as GFP fusion proteins, as well as their ability to support Nox1 activity in several transfected models. Noxo1alpha, beta, gamma, and delta show different subcellular localization patterns, determined by their PX domains. In HEK293 cells, Noxo1beta exhibits prominent plasma membrane binding, Noxo1gamma shows plasma membrane and nuclear associations, and Noxo1alpha and delta localize primarily on intracellular vesicles or cytoplasmic aggregates, but not the plasma membrane. Nox1 activity correlates with Noxo1 plasma membrane binding in HEK293 cells, since Noxo1beta supports the highest activity and Noxo1gamma and Noxo1alpha support moderate or low activities, respectively. In COS-7 cells, where Noxo1alpha localizes on the plasma membrane, the activities supported by the three isoforms (alpha, beta, and gamma) do not differ significantly. The PX domains of beta and gamma bind the same phospholipids, including phosphatidic acid. These results indicate that the variant PX domains are unique determinants of Noxo1 localization and Nox1 function. Finally, the overexpressed Noxo1 isoforms do not affect p22(phox) localization, although Nox1 is needed to transport p22(phox) to the plasma membrane.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Adaptor Proteins, Vesicular Transport/ultrastructure , Subcellular Fractions/enzymology , Subcellular Fractions/ultrastructure , Adaptor Proteins, Signal Transducing , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cell Membrane/enzymology , Enzyme Activation/physiology , Humans , Immunoblotting , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Microscopy, Confocal , Molecular Sequence Data , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NADPH Oxidases/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , Transfection
14.
J Comp Neurol ; 500(6): 1127-35, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17183553

ABSTRACT

Calcium, a ubiquitous intracellular messenger, regulates numerous intracellular signaling pathways. To permit specificity of signal transduction and prevent unwanted cross-talk between pathways, sites of calcium entry in neurons are localized to specific membrane domains. To test whether Ca(2+) extrusion pumps might exhibit analogous compartmentalization, we used immunohistochemistry to determine the subcellular localization of the two main plasma membrane Ca(2+)-ATPase (PMCA) isoforms in the cortex of the rat cerebellum. We find that both PMCA2 and PMCA3 are targeted to distinct compartments within the plasma membrane. In the molecular layer, both isoforms were at highest levels within synaptic profiles, but PMCA2 was postsynaptic and PMCA3 was presynaptic. Moreover, inside these compartments, both pumps exhibited nonuniform distributions. These data imply that cerebellar neurons possess remarkably effective mechanisms to target and restrict PMCA2 and -3 to specific membrane domains, raising the possibility that calcium pumps contribute to local Ca(2+) signaling.


Subject(s)
Cerebellar Cortex/enzymology , Plasma Membrane Calcium-Transporting ATPases/metabolism , Presynaptic Terminals/enzymology , Synaptic Membranes/metabolism , Animals , Cerebellar Cortex/ultrastructure , Immunohistochemistry , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Male , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Synaptic Membranes/ultrastructure
15.
FEBS J ; 273(4): 682-94, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16441656

ABSTRACT

Ca2+/calmodulin activated protein kinase II (CaMKII) is an oligomeric protein kinase with a unique holoenyzme architecture. The subunits of CaMKII are bound together into the holoenzyme by the association domain, a C-terminal region of approximately 140 residues in the CaMKII polypeptide. Single particle analyses of electron micrographs have suggested previously that the holoenyzme forms a dodecamer that contains two stacked 6-fold symmetric rings. In contrast, a recent crystal structure of the isolated association domain of mouse CaMKIIalpha has revealed a tetradecameric assembly with two stacked 7-fold symmetric rings. In this study, we have determined the crystal structure of the Caenorhabditis elegans CaMKII association domain and it too forms a tetradecamer. We also show by electron microscopy that in its fully assembled form the CaMKII holoenzyme is a dodecamer but without the kinase domains, either from expression of the isolated association domain in bacteria or following their removal by proteolysis, the association domains form a tetradecamer. We speculate that the holoenzyme is held in its 6-fold symmetric state by the interactions of the N-terminal approximately 1-335 residues and that the removal of this region allows the association domain to convert into a more stable 7-fold symmetric form.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Isoenzymes/chemistry , Protein Conformation , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/ultrastructure , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/ultrastructure , Cryoelectron Microscopy , Crystallography, X-Ray , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Holoenzymes/ultrastructure , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Mice , Models, Molecular , Mutagenesis, Site-Directed , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism
16.
Rev. colomb. biotecnol ; 7(1): 59-65, jul. 2005. mapas, tab, graf
Article in English | LILACS | ID: lil-606122

ABSTRACT

Two hundred and sixty-one accessions of the genus Capsicum were obtained from the Colombian Amazonian germplasm bank at Amazonian Institute of Scientific Research (Sinchi) and were evaluated with five polymorphic enzymatic systems, including esterase (EST), peroxidase (PRX), 6-phosphogluconatedehydrogenase (6-PGDH), aspartate amino transferase (GOT), and malic enzyme (ME). Using a cluster analysis (UPGMA) the genetic variability of these accessions were characterized. Grouping of the species C. baccatum and C. pubescens were observed, while the species C. annuum, C. chinense and C. frutescens did not group independently, a result that has been previously reported in isoenzyme analyses of this genus. Several accessions were deemed of particular interest for future ecological and evolutive studies.


Doscientas sesenta y una accesiones del género Capsicum del banco de germoplasma del Instituto Amazónico de Investigaciones Científicas (Sinchi) se evaluaron a través de cinco sistemas enzimáticos polimórficos: esterasa (EST), peroxidasa (PRX), 6-fosfogluconato deshidrogenasa (6-PGDH), aspartato amino transferasa(GOT) y enzima málica (ME). Se utilizó un análisis de agrupamiento (Upgma) con el fin de determinar la variabilidad genética. Se observó un agrupamiento de las especies C. baccatum y C. pubescens, mientras que las especies C. annuum, C. chinense y C. frutescens no mostraron un agrupamiento independiente, lo cual ya ha sido reportado en estudios por isoenzimas para el género. Varias accesiones mostraron característicasparticulares para estudios ecológicos y evolutivos.


Subject(s)
Capsicum/classification , Capsicum/growth & development , Capsicum/enzymology , Capsicum/microbiology , Enzyme Reactivators , Isoenzymes/analysis , Isoenzymes/classification , Isoenzymes/ultrastructure
17.
J Immunol ; 173(7): 4582-9, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15383592

ABSTRACT

Protein kinase C (PKC) plays a prominent role in immune signaling. To elucidate the signal transduction in a respiratory burst and isoform-specific function of PKC during FcgammaR-mediated phagocytosis, we used live, digital fluorescence imaging of mouse microglial cells expressing GFP-tagged molecules. betaI PKC, epsilonPKC, and diacylglycerol kinase (DGK) beta dynamically and transiently accumulated around IgG-opsonized beads (BIgG). Moreover, the accumulation of p47(phox), an essential cytosolic component of NADPH oxidase and a substrate for betaI PKC, at the phagosomal cup/phagosome was apparent during BIgG ingestion. Superoxide (O(2)(-)) production was profoundly inhibited by Gö6976, a cPKC inhibitor, and dramatically increased by the DGK inhibitor, R59949. Ultrastructural analysis revealed that BIgG induced O(2)(-) production at the phagosome but not at the intracellular granules. We conclude that activation/accumulation of betaI PKC is involved in O(2)(-) production, and that O(2)(-) production is primarily initiated at the phagosomal cup/phagosome. This study also suggests that DGKbeta plays a prominent role in regulation of O(2)(-) production during FcgammaR-mediated phagocytosis.


Subject(s)
Microglia/enzymology , Microglia/immunology , Phagocytosis/immunology , Phagosomes/metabolism , Protein Kinase C/physiology , Receptors, IgG/physiology , Superoxides/metabolism , Animals , Cell Line, Transformed , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Diacylglycerol Kinase/ultrastructure , Green Fluorescent Proteins , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , Isoenzymes/ultrastructure , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Luminescent Proteins/ultrastructure , Mice , Microglia/metabolism , Microglia/ultrastructure , Microspheres , Oxidants/biosynthesis , Phagocytes/enzymology , Phagocytes/immunology , Phagocytes/metabolism , Phagocytes/ultrastructure , Phagocytosis/genetics , Phagosomes/enzymology , Phagosomes/genetics , Phagosomes/ultrastructure , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C/ultrastructure , Protein Kinase C beta , Protein Kinase C-epsilon
18.
Biochem J ; 383(Pt. 3): 439-46, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15285718

ABSTRACT

IDE (insulin-degrading enzyme) is a widely expressed zinc-metallopeptidase that has been shown to regulate both cerebral amyloid beta-peptide and plasma insulin levels in vivo. Genetic linkage and allelic association have been reported between the IDE gene locus and both late-onset Alzheimer's disease and Type II diabetes mellitus, suggesting that altered IDE function may contribute to some cases of these highly prevalent disorders. Despite the potentially great importance of this peptidase to health and disease, many fundamental aspects of IDE biology remain unresolved. Here we identify a previously undescribed mitochondrial isoform of IDE generated by translation at an in-frame initiation codon 123 nucleotides upstream of the canonical translation start site, which results in the addition of a 41-amino-acid N-terminal mitochondrial targeting sequence. Fusion of this sequence to the N-terminus of green fluorescent protein directed this normally cytosolic protein to mitochondria, and full-length IDE constructs containing this sequence were also directed to mitochondria, as revealed by immuno-electron microscopy. Endogenous IDE protein was detected in purified mitochondria, where it was protected from digestion by trypsin and migrated at a size consistent with the predicted removal of the N-terminal targeting sequence upon transport into the mitochondrion. Functionally, we provide evidence that IDE can degrade cleaved mitochondrial targeting sequences. Our results identify new mechanisms regulating the subcellular localization of IDE and suggest previously unrecognized roles for IDE within mitochondria.


Subject(s)
Codon, Initiator/genetics , Insulysin/genetics , Amino Acid Sequence/genetics , Animals , CHO Cells/chemistry , Cell Line , Conserved Sequence/genetics , Cricetinae , Cricetulus , Humans , Immunohistochemistry/methods , Insulysin/physiology , Insulysin/ultrastructure , Isoenzymes/genetics , Isoenzymes/physiology , Isoenzymes/ultrastructure , Kidney/chemistry , Kidney/cytology , Kidney/embryology , Methionine/genetics , Mice , Microscopy, Electron/methods , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Mitochondrial Proteins/ultrastructure , Molecular Sequence Data , Rats , Sequence Alignment/methods , Sequence Homology, Nucleic Acid , Submitochondrial Particles/ultrastructure
19.
Biochemistry ; 43(6): 1541-51, 2004 Feb 17.
Article in English | MEDLINE | ID: mdl-14769030

ABSTRACT

The Staphylococcus aureus sortase transpeptidase SrtA isoform is responsible for the covalent attachment of virulence and colonization-associated proteins to the bacterial peptidoglycan. SrtA utilizes two substrates, undecaprenol-pyrophosphoryl-MurNAc(GlcNAc)-Ala-D-isoGlu-Lys(epsilon-Gly(5))-D-Ala-D-Ala (branched Lipid II) and secreted proteins containing a highly conserved C-terminal LPXTG sequence. SrtA simultaneously cleaves the Thr-Gly bond of the LPXTG-containing protein and forms a new amide bond with the nucleophilic amino group of the Gly(5) portion of branched Lipid II, anchoring the protein to this key intermediate that is subsequently polymerized into peptidoglycan. Here we describe the development of a general in vitro method for elucidating the substrate specificity of sortase enzymes. In addition, using immunofluorescence, cell adhesion assays, and transmission electron microscopy, we establish links between in vitro substrate specificity and in vivo function of the S. aureus sortase isoforms. Results from these studies provide strong supporting evidence of a primary role of the SrtA isoform in S. aureus adhesion and host colonization, illustrate a lack of specificity cross talk between SrtA and SrtB isoforms, and highlight the potential of SrtA as a target for the development of antivirulence chemotherapeutics against Gram-positive bacterial pathogens.


Subject(s)
Aminoacyltransferases/chemistry , Staphylococcus aureus/enzymology , Amino Acid Motifs , Aminoacyltransferases/deficiency , Aminoacyltransferases/genetics , Aminoacyltransferases/ultrastructure , Bacterial Adhesion , Bacterial Proteins , Cell Wall/chemistry , Cell Wall/genetics , Cell Wall/ultrastructure , Cloning, Molecular , Conserved Sequence , Cysteine Endopeptidases , Enzyme Activation/genetics , Histidine/chemistry , Isoenzymes/chemistry , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/ultrastructure , Mutation , Peptide Library , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/ultrastructure , Substrate Specificity , Virulence
20.
Biochemistry ; 42(51): 15132-8, 2003 Dec 30.
Article in English | MEDLINE | ID: mdl-14690423

ABSTRACT

Membrane-bound H/K-ATPase was solubilized by octaethylene glycol dodecyl ether (C(12)E(8)) or n-octyl glucoside (nOG). H/K-ATPase activity and the distribution of protomeric and oligomeric components were evaluated by high-performance gel chromatography (HPGC) and by single-molecule detection using total internal reflection fluorescence microscopy (TIRFM). As evidenced by HPGC of the C(12)E(8)-solubilized enzyme, the distribution of oligomers was 12% higher oligomeric, 44% diprotomeric, and 44% protomeric species, although solubilization by C(12)E(8) reduced the H/K-ATPase activity to 1.8% of that of the membrane-bound enzyme. The electron microscopic images of the C(12)E(8)-solubilized enzyme showed the presence of protomers and a combination of two and more protomers. While the nOG-solubilized H/K-ATPase retained the same turnover number and 71% of the specific activity as that of the membrane-bound enzyme, 56% higher oligomeric, 34% diprotomeric, and 10% protomeric species were detected. TIRFM analysis of solubilized fluorescein 5'-isothiocyanate (FITC)-modified H/K-ATPase at Lys-518 of the alpha-chain showed a quantized photobleaching of the FITC fluorescence intensity. For the C(12)E(8)-solubilized FITC-enzyme, the fraction of each of the initial relative fluorescence intensity units of 4, 2, and 1 was, respectively, 5%, 44% and 51%. In the case of the nOG-solubilized FITC-enzyme, each fraction of 4 and 2 units was, respectively, 54% and 46% with no detectable 1 unit fraction. This represents the first direct observation of H/K-ATPase in aqueous solution. The correlation between the enzymatic activities and distribution of oligomeric forms of H/K-ATPase by HPGC and the observation of a single molecule of H/K-ATPase and others suggests that the tetraprotomeric form of H/K-ATPase molecules represents the functional species in the membrane.


Subject(s)
H(+)-K(+)-Exchanging ATPase/chemistry , H(+)-K(+)-Exchanging ATPase/metabolism , Animals , Chemical Fractionation , Chromatography, Gel , Detergents/chemistry , Enzyme Activation , Gastric Mucosa/enzymology , Glucosides/chemistry , H(+)-K(+)-Exchanging ATPase/ultrastructure , Isoenzymes/chemistry , Isoenzymes/metabolism , Isoenzymes/ultrastructure , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Microscopy, Electron , Microscopy, Fluorescence , Molecular Weight , Solubility , Solutions , Structure-Activity Relationship , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...