Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 5897, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33214556

ABSTRACT

Since the fluorescent reagent N-(1-pyrene)iodoacetamide was first used to label skeletal muscle actin in 1981, the pyrene-labeled actin has become the most widely employed tool to measure the kinetics of actin polymerization and the interaction between actin and actin-binding proteins. Here we report high-resolution cryo-electron microscopy structures of actin filaments with N-1-pyrene conjugated to cysteine 374 and either ADP (3.2 Å) or ADP-phosphate (3.0 Å) in the active site. Polymerization buries pyrene in a hydrophobic cavity between subunits along the long-pitch helix with only minor differences in conformation compared with native actin filaments. These structures explain how polymerization increases the fluorescence 20-fold, how myosin and cofilin binding to filaments reduces the fluorescence, and how profilin binding to actin monomers increases the fluorescence.


Subject(s)
Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Adenosine Diphosphate/metabolism , Phosphates/metabolism , Pyrenes/chemistry , Actins/chemistry , Actins/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Catalytic Domain , Cryoelectron Microscopy , Fluorescence , Hydrophobic and Hydrophilic Interactions , Iodoacetamide/analogs & derivatives , Iodoacetamide/chemistry , Kinetics , Microfilament Proteins/metabolism , Polymerization , Protein Binding
2.
Drug Metab Dispos ; 48(6): 508-514, 2020 06.
Article in English | MEDLINE | ID: mdl-32193357

ABSTRACT

Experiments designed to identify the mechanism of cytochrome P450 inactivation are critical to drug discovery. Small molecules irreversibly inhibit P450 enzymatic activity via two primary mechanisms: apoprotein adduct formation or heme modification. Understanding the interplay between chemical structures of reactive electrophiles and the impact on CYP3A4 structure and function can ultimately provide insights into drug design to minimize P450 inactivation. In a previous study, raloxifene and N-(1-pyrene) iodoacetamide (PIA) alkylated CYP3A4 in vitro; however, only raloxifene influenced enzyme activity. Here, two alkylating agents with cysteine selectivity, PIA and pyrene maleimide (PM), were used to investigate this apparent compound-dependent disconnect between CYP3A4 protein alkylation and activity loss. The compound's effect on 1) enzymatic activity, 2) carbon monoxide (CO) binding capacity, 3) intact heme content, and 4) protein conformation were measured. Results showed that PM had a large time-dependent loss of enzyme activity, whereas PIA did not. The differential effect on enzymatic activity between PM and PIA was mirrored in the CO binding data. Despite disruption of CO binding, neither compound affected the heme concentrations, inferring there was no destruction or alkylation of the heme. Lastly, differential scanning fluorescence showed PM-treated CYP3A4 caused a shift in the onset temperature required to induce protein aggregation, which was not observed for CYP3A4 treated with PIA. In conclusion, alkylation of CYP3A4 apoprotein can have a variable impact on catalytic activity, CO binding, and protein conformation that may be compound-dependent. These results highlight the need for careful interpretation of experimental results aimed at characterizing the nature of P450 enzyme inactivation. SIGNIFICANCE STATEMENT: Understanding the mechanism of CYP3A4 time-dependent inhibition is critical to drug discovery. In this study, we use two cysteine-targeting electrophiles to probe how subtle variation in inhibitor structure may impact the mechanism of CYP3A4 time-dependent inhibition and confound interpretation of traditional diagnostic experiments. Ultimately, this simplified system was used to reveal insights into CYP3A4 biochemical behavior. The insights may have implications that aid in understanding the susceptibility of CYP enzymes to the effects of electrophilic intermediates generated via bioactivation.


Subject(s)
Apoproteins/metabolism , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A/metabolism , Alkylation/drug effects , Apoproteins/antagonists & inhibitors , Apoproteins/chemistry , Carbon Monoxide/metabolism , Cysteine/chemistry , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 CYP3A Inhibitors/chemistry , Enzyme Assays , Iodoacetamide/analogs & derivatives , Iodoacetamide/chemistry , Iodoacetamide/pharmacology , Maleimides/chemistry , Maleimides/pharmacology , Oxidation-Reduction/drug effects , Protein Conformation/drug effects , Recombinant Proteins/metabolism
3.
Anal Bioanal Chem ; 406(26): 6723-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25168110

ABSTRACT

A new long-wavelength fluorescent probe, 1,7-dimethyl-3,5-distyryl-8-phenyl-(4'-iodoacetamido)difluoroboradiaza-s-indacene (DMDSPAB-I), was designed and synthesized for thiol labeling in high-performance liquid chromatography (HPLC). The excitation and emission wavelengths of DMDSPAB-I are 620 and 630 nm, respectively, with a high fluorescence quantum yield of 0.557, which is advantageous in preventing interference of intrinsic fluorescence from complex biological matrices and enabling high sensitivity HPLC. Based on DMDSPAB-I, a reversed-phase HPLC method was developed for measuring low-molecular-weight thiols including glutathione, cysteine, homocysteine, N-acetylcysteine, cysteinylglycine, and penicillamine. After the specific reaction of DMDSPAB-I with thiols, baseline separation of all six stable derivatives was achieved through isocratic elution on a C18 column within 25 min, with the limits of detection (signal-to-noise ratio = 3) from 0.24 nmol L(-1) for glutathione to 0.72 nmol L(-1) for penicillamine. The proposed method was validated in part by measuring thiols in blood samples from mice, with recoveries of 95.3-104.3%.


Subject(s)
Boron Compounds/chemistry , Chromatography, High Pressure Liquid/methods , Chromatography, Reverse-Phase/methods , Fluorescent Dyes/chemistry , Iodoacetamide/analogs & derivatives , Sulfhydryl Compounds/blood , Acetylcysteine/blood , Acetylcysteine/isolation & purification , Animals , Cysteine/blood , Cysteine/isolation & purification , Dipeptides/blood , Dipeptides/isolation & purification , Glutathione/blood , Glutathione/isolation & purification , Homocysteine/blood , Homocysteine/isolation & purification , Iodoacetamide/chemistry , Limit of Detection , Male , Mice , Penicillamine/blood , Penicillamine/isolation & purification , Sulfhydryl Compounds/isolation & purification
4.
J Proteomics ; 80: 160-70, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23352896

ABSTRACT

Microwave-assisted proteolytic digestion methods have evolved into a highly effective approach and serve as an alternative to conventional overnight digestion. This approach typically exploits the unique microwave properties to facilitate the digestion of proteins into their peptides within minutes. Conventional digestion is carried out at 37°C while microwave-assisted digestion requires much higher and sometimes inconsistent temperatures. Thus, this study aims to investigate whether the faster reaction rate is due to the microwave quantum effect or the thermal effect. Quantitative mass spectrometry was used to conduct kinetic analysis of tryptic digestion for several proteins by microwave and conventional heating. The percentages of digestion products relative to internal standards showed no significant difference between microwave and conventional heating conditions at the same digestion temperature. The optimum temperature for tryptic digestion was determined to be 50°C. Furthermore, this study compares the digestion completeness indicators of several proteins under microwave and conventional heating. Again, the values obtained from microwave and conventional heating were similar given identical temperatures. The overall results prove that a nonthermal effect does not exist in microwave-assisted tryptic digestion. Therefore, conventional heating at high temperatures (50°C) can be also used to accelerate digestion reactions.


Subject(s)
Microwaves , Proteolysis , Animals , Cattle , Chickens , Cytochromes/chemistry , Enzymes/chemistry , Hot Temperature , Humans , Iodoacetamide/analogs & derivatives , Iodoacetamide/chemistry , Mass Spectrometry , Peptides/chemistry , Proteins/chemistry , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Temperature , Trypsin/chemistry
5.
Proc Natl Acad Sci U S A ; 109(45): 18425-30, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23091026

ABSTRACT

Cooperative activation of actin-myosin interaction by tropomyosin (Tm) is central to regulation of contraction in muscle cells and cellular and intracellular movements in nonmuscle cells. The steric blocking model of muscle regulation proposed 40 y ago has been substantiated at both the kinetic and structural levels. Even with atomic resolution structures of the major players, how Tm binds and is designed for regulatory function has remained a mystery. Here we show that a set of periodically distributed evolutionarily conserved surface residues of Tm is required for cooperative regulation of actomyosin. Based on our results, we propose a model of Tm on a structure of actin-Tm-myosin in the "open" (on) state showing potential electrostatic interactions of the residues with both actin and myosin. The sites alternate with a second set of conserved surface residues that are important for actin binding in the inhibitory state in the absence of myosin. The transition from the closed to open states requires the sites identified here, even when troponin + Ca(2+) is present. The evolutionarily conserved residues are important for actomyosin regulation, a universal function of Tm that has a common structural basis and mechanism.


Subject(s)
Actins/metabolism , Conserved Sequence , Myosins/metabolism , Tropomyosin/metabolism , Actin Cytoskeleton/metabolism , Adenosine Diphosphate/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calcium/metabolism , Evolution, Molecular , Fluorescence , Iodoacetamide/analogs & derivatives , Iodoacetamide/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Phosphates/metabolism , Protein Binding , Protein Transport , Rats , Scattering, Radiation , Tropomyosin/chemistry , Tropomyosin/genetics , Troponin/metabolism
6.
Methods Mol Biol ; 889: 325-46, 2012.
Article in English | MEDLINE | ID: mdl-22669675

ABSTRACT

Oxidative stress has been recognized as a contributing factor in the toxicity of a large number of developmental toxicants. Traditional definitions of oxidative stress state that a shift in the balance between reduced and oxidized biomolecules within cells, in favor of the latter, result in changes that are deleterious to vital cell functions and can culminate in malformations and death. The glutathione (GSH)/glutathione disulfide (GSSG) redox couple has been the traditional marker of choice for characterization of oxidative stress because of its high concentrations and direct roles as antioxidant and cellular protectant. Steady state depletion of GSH through conjugation, oxidation, or export has often been reported as the sole criteria for invoking oxidative stress and a myriad of associated deleterious consequences. Numerous other, mostly qualitative, observations have also been reported to suggest oxidative stress has occurred but it is not always clear how well they reflect the state of a cell or its functions. Our emerging understanding of redox signaling and the roles of reactive oxygen species (ROS), thiols, oxidant molecules, and cellular antioxidants, all acting as second messengers, has prompted a redefinition of oxidative stress based on changes in the real posttranslational protein thiol modifications that are central to redox regulation and control. Thiol-based redox couples such as GSH/GSSG, cysteine/cystine (cys/cySS), thioredoxin-reduced/thioredoxin-oxidized (TRX(red)/TRX(ox)) form independent signaling nodes that selectively regulate developmental events and are closely linked to changes in intracellular redox potentials. Accurate assessment of the consequences of increased free radicals in developing conceptuses should best be made using a battery of measurements including the quantitative assessment of intracellular redox potential, ROS, redox status of biomolecules, and induced changes in specific redox signaling nodes. Methods are presented for a determination of ROS production, soluble thiol oxidation, redox potential, and a proteomic approach to evaluate the thiol oxidation state of specific proteins.


Subject(s)
Oxidative Stress , Sulfhydryl Compounds/metabolism , Algorithms , Animals , Blotting, Western/methods , Cell Culture Techniques , Cells, Cultured , Chromatography, High Pressure Liquid/methods , Chromatography, High Pressure Liquid/standards , Electrophoresis, Polyacrylamide Gel/methods , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Iodoacetamide/analogs & derivatives , Iodoacetamide/chemistry , Limb Buds/cytology , Mice , Oxidation-Reduction , Proteins/chemistry , Proteins/isolation & purification , Proteins/metabolism , Rats , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism , Reference Standards , Signal Transduction , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/isolation & purification
7.
Gen Physiol Biophys ; 28(2): 160-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19592712

ABSTRACT

The present study deals with the determination of hydrodynamic size of DNA/cationic gemini surfactant complex in sodium bromide solution using the dynamic light scattering method. Cationic gemini surfactants with polymethylene spacer of variable length were used for the interaction with DNA. The scattering experiments were performed at constant DNA and sodium bromide concentrations and variable surfactant concentration in the premicellar and micellar regions as a function of surfactant spacer length. It was found that the DNA conformation strongly depends on the polymethylene spacer length as well as on the surfactant concentration relative to the surfactant critical micelle concentration. Gemini surfactant molecules with 4 methylene groups in the spacer were found to be the least efficient DNA compacting agent in the region above the surfactant cmc. Gemini molecules with the shortest spacer length (2 methylene groups) and the longest spacer length (8 methylene groups) investigated showed the most efficient DNA compaction ability.


Subject(s)
Benzalkonium Compounds/chemistry , DNA/chemistry , Surface-Active Agents/chemistry , Animals , Bromides/chemistry , Fluoresceins , Iodoacetamide/analogs & derivatives , Light , Micelles , Nucleic Acid Conformation , Particle Size , Salmon , Scattering, Radiation , Sodium Compounds/chemistry
8.
J Sep Sci ; 31(3): 538-47, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18210377

ABSTRACT

The use of iodoacetamide (IAA) and N-methyliodoacetamide (MIAA) as labeling agents for the relative measurements of proteins using MALDI-MS is described herein. These reagents, which alkylate the thiol groups of cysteine residues in proteins, were introduced during the alkylation step of a common protein denaturation and digestion process. This approach is simpler and cheaper than those involving isotope labeling agents. The labeling agents described herein displayed good dynamic ranges and correlation coefficients for protein quantification analyses when the proteins were treated through either in-solution or in-gel digestion. The best dynamic ranges (in the molar ratio) for proteins lysozyme, transferrin, and BSA (in-solution digestion) are 0.1-10, 0.1-8, and 0.1-8, respectively. The corresponding correlation coefficients are greater than 0.99. The IAA/MIAA labeling is a useful method for the relative quantification of peptides and digested proteins when the chromatographic isotope effect is not a major concern.


Subject(s)
Iodoacetamide/analogs & derivatives , Iodoacetamide/chemistry , Peptide Fragments/analysis , Proteins/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Amino Acid Sequence , Animals , Indicators and Reagents/chemistry , Muramidase/analysis , Muramidase/chemistry , Proteins/chemistry , Reproducibility of Results , Sensitivity and Specificity , Serum Albumin, Bovine/analysis , Serum Albumin, Bovine/chemistry , Transferrin/analysis , Transferrin/chemistry
10.
Anal Biochem ; 375(2): 187-95, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18162165

ABSTRACT

Cysteine reactivity in enzymes is imparted to a large extent by the stabilization of the deprotonated form of the reduced cysteine (i.e., the thiolate) within the active site. Although this is likely to be an important chemical attribute of many thiol-based enzymes, including cysteine-dependent peroxidases (peroxiredoxins) and proteases, only relatively few pK(a) values have been determined experimentally. Presented here is a new technique for determining the pK(a) value of cysteine residues through quantitative mass spectrometry following chemical modification with an iodoacetamide-based reagent over a range of pH buffers. This isotope-coded reagent, N-phenyl iodoacetamide (iodoacetanilide), is readily prepared in deuterated (d(5)) and protiated (d(0)) versions and is more reactive toward free cysteine than is iodoacetamide. Using this approach, the pK(a) values for the two cysteine residues in Escherichia coli thioredoxin were determined to be 6.5 and greater than 10.0, in good agreement with previous reports using chemical modification approaches. This technique allows the pK(a) of specific cysteine residues to be determined in a clear, fast, and simple manner and, because cysteine residues on separate tryptic peptides are measured separately, is not complicated by the presence of multiple cysteines within the protein of interest.


Subject(s)
Cysteine/chemistry , Iodoacetamide/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Acetanilides/chemistry , Deuterium/chemistry , Hydrogen-Ion Concentration , Indicators and Reagents/chemical synthesis , Indicators and Reagents/chemistry , Iodoacetamide/analogs & derivatives , Iodoacetamide/chemical synthesis , Isotopes , Kinetics , Oxidation-Reduction , Peptides/chemistry , Tandem Mass Spectrometry , Thioredoxins/chemistry , Titrimetry
11.
Chem Res Toxicol ; 20(6): 954-64, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17497897

ABSTRACT

Time-dependent inactivation of cytochrome P450s is typically a result of substrate bioactivation to form reactive species that subsequently alkylate the heme group, apoprotein, or both. The chemical identity of many reactive intermediates is generally proposed based on the products of trapping reactions with nucleophilic agents as only a few P450-drug adducts have been directly characterized. We describe the use of mass spectrometry to show that a single equivalent of raloxifene is bound to the intact P450 apoprotein. Furthermore, mass analysis of peptides following digestion with proteinase K revealed that the covalently bound drug is localized to residue Cys239. A mass shift of 471 Da to the intact protein and peptide, relative to control samples, indicated that time-dependent inactivation of P450 3A4 occurred through the raloxifene diquinone methide intermediately prior to nucleophilic attack of the sulfur of Cys239. Association between raloxifene adduction to P450 3A4 apoprotein and the observed time-dependent inactivation was further investigated with the use of cysteine-specific modifying reagents. When P450 3A4 was treated with iodoacetamide or N-(1-pyrene)iodoacetamide, which alkylated residue Cys239 exclusively, time-dependent inactivation of P450 3A4 by raloxifene was prevented. The change in protein mass of 471 Da combined with the protection from inactivation that occurred through pre-alkylation of Cys239 provided conclusive evidence that raloxifene-mediated P450 3A4 inactivation occurred through the bioactivation of raloxifene to the diquinone methide and subsequent alkylation of Cys239.


Subject(s)
Apoproteins/metabolism , Cysteine/metabolism , Cytochrome P-450 Enzyme System/metabolism , Raloxifene Hydrochloride/pharmacology , Alkylating Agents/chemistry , Alkylating Agents/pharmacology , Alkylation/drug effects , Apoproteins/chemistry , Binding Sites , Cysteine/chemistry , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Endopeptidase K/chemistry , Endopeptidase K/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Glutathione/chemistry , Glutathione/metabolism , Iodoacetamide/analogs & derivatives , Iodoacetamide/chemistry , Iodoacetamide/pharmacology , Kinetics , Midazolam/chemistry , Midazolam/pharmacology , Molecular Structure , Quinolines/chemistry , Quinolines/metabolism , Raloxifene Hydrochloride/chemistry , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Ultraviolet , Time Factors
12.
Bioorg Khim ; 33(6): 593-7, 2007.
Article in Russian | MEDLINE | ID: mdl-18173121

ABSTRACT

5-Bromo-2[(2-iodoacetyl)amino]benzenesulfonic acid (AIBSA), a reagent for modification of free of cysteine thiol groups in proteins and peptides, was synthesized. Rate constants of its interaction with thiol groups were determined. The presence of a bromine atom allows an easy identification of the AIBSA-labeled peptides in mass spectra due to the characteristic isotope distribution. The compound is stable in solution and under exposure to light.


Subject(s)
Benzenesulfonates/chemistry , Bromine/chemistry , Cysteine/chemistry , Iodoacetamide/analogs & derivatives , Proteins/chemistry , Benzenesulfonates/chemical synthesis , Benzenesulfonates/radiation effects , Iodoacetamide/chemical synthesis , Iodoacetamide/chemistry , Iodoacetamide/radiation effects
13.
J Biol Chem ; 281(31): 22152-22160, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16740630

ABSTRACT

Peptide:N-glycanase (PNGase) is the deglycosylating enzyme, which releases N-linked glycan chains from N-linked glycopeptides and glycoproteins. Recent studies have revealed that the cytoplasmic PNGase is involved in the degradation of misfolded/unassembled glycoproteins. This enzyme has a Cys, His, and Asp catalytic triad, which is required for its enzymatic activity and can be inhibited by "free" N-linked glycans. These observations prompted us to investigate the possible use of haloacetamidyl derivatives of N-glycans as potent inhibitors and labeling reagents of this enzyme. Using a cytoplasmic PNGase from budding yeast (Png1), Man9GlcNAc2-iodoacetoamide was shown to be a strong inhibitor of this enzyme. The inhibition was found to be through covalent binding of the carbohydrate to a single Cys residue on Png1, and the binding was highly selective. The mutant enzyme in which Cys191 of the catalytic triad was changed to Ala did not bind to the carbohydrate probe, suggesting that the catalytic Cys is the binding site for this compound. Precise determination of the carbohydrate attachment site by mass spectrometry clearly identified Cys191 as the site of covalent attachment. Molecular modeling of N,N'-diacetylchitobiose (chitobiose) binding to the protein suggests that the carbohydrate binding site is distinct from but adjacent to that of Z-VAD-fmk, a peptide-based inhibitor of this enzyme. These results suggest that cytoplasmic PNGase has a separate binding site for chitobiose and other carbohydrates, and haloacetamide derivatives can irreversibly inhibit that catalytic Cys in a highly specific manner.


Subject(s)
Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/chemistry , Amino Acid Substitution , Binding Sites , Carbohydrates/chemistry , Cytoplasm/enzymology , Disaccharides/chemistry , Disaccharides/metabolism , Fungal Proteins/chemistry , Iodoacetamide/analogs & derivatives , Iodoacetamide/chemistry , Iodoacetamide/metabolism , Models, Molecular , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/antagonists & inhibitors , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/genetics
14.
Ukr Biokhim Zh (1999) ; 75(2): 72-7, 2003.
Article in Ukrainian | MEDLINE | ID: mdl-14577174

ABSTRACT

Actin polymerization was investigated using fluorescence probe N-(1-pyrenyl)iodoacetamide, which was bound covalently to reactive sulfhydryl group, Cys-373. Labeled actin in the bulk was 0.5 to 1% of total actin concentration. Actin polymerization at concentration 12 mM was started by addition of 20 mM KCl and 2 mM MgCl2. The label fluorescence was excited at 365 nm and registered at 386 nm. Under actin polymerization the label fluorescence increased almost 10 times. Two main phases may be distinguished in the process of actin polymerization: 1) monomer activation and nucleus (trimer) formation, 2) growth of actin filaments on the nuclei. In our experimental conditions, both for pure actin and for that with added annexin VI, the 1st phase continued for about 3 min and after that the 2nd phase was perfectly approximated by exponential dependence. An analysis of the exponential curves showed that actin monomer lifetime increased from 327 s, at annexin absence, to about 373 s at 0.7 microM annexin and more. Calculation of rate constants at two ends of growing actin filament suggests that annexin VI binds with pointed ("slow") end so that at sufficient annexin concentration the filament grows only on barbed ("fast") end. Our results, together with data of other researchers showing that annexin VI binds with the inner membrane surface of smooth muscle cell through Ca2+, may indicate that, at Ca2+ entering the cell, this annexin binds actin filament pointed ends to cell surface making it ready for the act of contraction.


Subject(s)
Actins/metabolism , Annexin A6/metabolism , Iodoacetamide/analogs & derivatives , Animals , Fluorescent Dyes/pharmacology , Iodoacetamide/pharmacology , Rabbits
15.
Anal Biochem ; 316(2): 154-61, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12711335

ABSTRACT

The amount of endogenous melatonin in the individual pineal glands of inbred mice has been determined using reversed-phase micro-high-performance liquid chromatography after precolumn oxidation of melatonin to a compound having strong fluorescence. The fluorescent compound was identified as N-[(6-methoxy-4-oxo-1,4-dihydroquinolin-3-yl)methyl]acetamide. The excitation and emission wavelengths of this compound are 245 and 380 nm, respectively, and the fluorescence intensity is 6.8 times greater than that of melatonin. Molar absorptivity and fluorescence quantum yield of this compound are 46,300[L mol(-1)cm(-1)] and 0.31 (245 nm), respectively. The lower quantification limit of melatonin in biological samples using this precolumn oxidation method is 200 amol, and the calibration curve of spiked melatonin is linear from 200 amol to 50 fmol (r>0.999). The sensitivity of the present method is almost 10 times higher than that of the previous method. The values of endogenous melatonin obtained for ICR, C57BL, BALB/c, and AKR mice are 4.7, 6.1, 7.4, and 18.8 fmol/pineal gland, respectively. The amounts of endogenous pineal melatonin of these strains had not been clearly reported due to the poor enzymatic activities for melatonin biosynthesis; this is the first report that clearly demonstrates the existence of endogenous melatonin in these inbred mice.


Subject(s)
Iodoacetamide/analogs & derivatives , Melatonin/analysis , Pineal Gland/metabolism , Animals , Chromatography, High Pressure Liquid/methods , Fluorescence , Iodoacetamide/standards , Magnetic Resonance Spectroscopy , Melatonin/analogs & derivatives , Mice , Mice, Inbred Strains , Microchemistry/methods , Molecular Structure , Oxidation-Reduction , Pineal Gland/chemistry , Reference Standards
17.
Cancer Res ; 62(21): 6080-8, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12414632

ABSTRACT

3-Iodoacetamido benzoyl ethyl ester (3-IAABE) is a new compound synthesized in our laboratory. The primary action of 3-IAABE is to inhibit microtubule assembly by interacting with -SH groups on tubulin. In contrast to other known microtubule disrupters, 3-IAABE caused a double blockade in the cell cycle at G(1)-S transition and in M phase. The blockade was determined by cell cycle analysis and chromosome distribution. Kinase activities of cyclin E and cyclin-dependent kinase 2 responsible for the G(1)-S transition were increased, as were the activities of mitotic cyclin B and cdc2. 3-IAABE treatment also increased p53 expression and dephosphorylated (or activated) retinoblastoma protein. Investigation of the signal transduction pathway showed that 3-IAABE induced bcl-2 phosphorylation, followed by activation of caspase-9, -3, and -6, but not caspase-8. DNA fragmentation factor and poly(ADP-ribose) polymerase, the downstream substrates of caspase-3 and -6, were cleaved after 3 h of exposure to 3-IAABE, followed by DNA fragmentation. Pretreatment of the cells with inhibitors of caspase-9, -3, or -6, respectively, inhibited the cleavage of DNA fragmentation factor and poly(ADP-ribose) polymerase and thus inhibited the onset of apoptosis. 3-IAABE showed antitumor activities in the panel of 60 National Cancer Institute human tumor cell lines with total growth inhibition in the range of 0.22-4.3 micro M for solid tumor lines and 0.025-0.22 micro M for leukemia/lymphoma cell lines. The 3-IAABU total growth inhibition of phytohemagglutinin-stimulated healthy human lymphocytes was 450-fold greater than that of leukemic cells. 3-IAABE significantly inhibited the growth of human hepatocarcinoma (BEL-7402) in nude mice by 72% in tumor volume, more strongly than did vincristine (43 percent inhibition). Besides being a novel lead for the design of new anticancer tubulin ligands, the activity of 3-IAABE in the cell cycle may also help us to understand the molecular pharmacology of microtubule-active drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Iodoacetamide/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Drug Screening Assays, Antitumor , Humans , Iodoacetamide/analogs & derivatives , Ligands , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Microtubules/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Tubulin/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , U937 Cells/drug effects , Vincristine/pharmacology , Xenograft Model Antitumor Assays
18.
Biochemistry ; 40(19): 5757-64, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11341841

ABSTRACT

Equilibrium measurements of the rate of binding of caldesmon and myosin S1 to actin-tropomyosin from different laboratories have yielded different results and have led to different models of caldesmon function. An alternate approach to answering these questions is to study the kinetics of binding of both caldesmon and S1 to actin. We observed that caldesmon decreased the rate of binding of S1 to actin in a concentration-dependent manner. The inhibition of the rate of S1 binding was enhanced by tropomyosin, but the effect of tropomyosin on the binding was small. Premixing actin with S1 reduced the amplitude (extent) of caldesmon binding in proportion to the fraction of actin that contained bound S1, but the rate of binding of caldesmon to free sites was not greatly altered. No evidence for a stable caldesmon-actin-tropomyosin-S1 complex was observed, although S1 did apparently bind to gaps between caldesmon molecules. These results indicate that experiments involving caldesmon, actin, tropomyosin, and myosin are inherently complex. When the concentration of either S1 or caldesmon is varied, the amount of the other component bound to actin-tropomyosin cannot be assumed to remain fixed. The results are not readily explained by a mechanism in which caldesmon acts only by stabilizing an inactive state of actin-tropomyosin. The results support regulatory mechanisms that involve changes in the actin-S1 interaction.


Subject(s)
Actins/metabolism , Calmodulin-Binding Proteins/metabolism , Iodoacetamide/analogs & derivatives , Myosin Subfragments/antagonists & inhibitors , Myosin Subfragments/metabolism , Tropomyosin/metabolism , Animals , Fluorescein/metabolism , Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Iodoacetamide/metabolism , Kinetics , Light , Oxadiazoles/metabolism , Protein Binding , Pyrenes/metabolism , Rabbits , Salicylates/metabolism , Scattering, Radiation , Spectrometry, Fluorescence , Turkeys
19.
Eur J Biochem ; 268(10): 3042-52, 2001 May.
Article in English | MEDLINE | ID: mdl-11358523

ABSTRACT

Fractionation of the serum of the venomous snake Bothrops jararaca with (NH4)2SO4, followed by phenyl-Sepharose and C4-reversed phase chromatographies, resulted in the isolation of the anti-hemorrhagic factor BJ46a. BJ46a is a potent inhibitor of the SVMPs atrolysin C (class P-I) and jararhagin (P-III) proteolytic activities and B. jararaca venom hemorrhagic activity. The single-chain, acidic (pI 4.55) glycoprotein has a molecular mass of 46 101 atomic mass units determined by MALDI-TOF MS and 79 kDa by gel filtration and dynamic laser light scattering, suggesting a homodimeric structure. mRNA was isolated from the liver of one specimen and transcribed into cDNA. The cDNA pool was amplified by PCR, cloned into a specific vector and used to transform competent cells. Clones containing the complete coding sequence for BJ46a were isolated. The deduced protein sequence was in complete agreement with peptide sequences obtained by Edman degradation. BJ46a is a 322-amino-acid protein containing four putative N-glycosylation sites. It is homologous to the proteinase inhibitor HSF (member of the fetuin family, cystatin superfamily) isolated from the serum of the snake Trimeresurus flavoviridis, having 85% sequence identity. This is the first report of a complete cDNA sequence for an endogenous inhibitor of snake venom metalloproteinases (SVMPs). The sequence reveals that the only proteolytic processing required to obtain the mature protein is the cleavage of the signal peptide. Gel filtration analyses of the inhibitory complexes indicate that inhibition occurs by formation of a noncovalent complex between BJ46a and the proteinases at their metalloproteinase domains. Furthermore, the data shows that the stoichiometry involved in this interaction is of one inhibitor monomer to two enzyme molecules, suggesting an interesting mechanism of metalloproteinase inhibition.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Snake Venoms/enzymology , Viper Venoms/chemistry , Viper Venoms/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Bothrops/blood , Crotalid Venoms/pharmacology , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Endopeptidases/chemistry , Indicators and Reagents/pharmacology , Iodoacetamide/analogs & derivatives , Iodoacetamide/pharmacology , Isoelectric Focusing , Light , Liver/metabolism , Metalloendopeptidases/chemistry , Metalloendopeptidases/pharmacology , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Scattering, Radiation , Sequence Analysis, DNA , Sequence Analysis, Protein , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors , Bothrops jararaca Venom
20.
Biochemistry ; 40(14): 4359-71, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11284692

ABSTRACT

Post-translational modifications of the developmental signaling protein Sonic hedgehog (Shh) by a long-chain fatty acid at the N-terminus and cholesterol at the C-terminus greatly activate the protein in a cell-based signaling assay. To investigate the structural determinants of this activation phenomenon, hydrophobic and hydrophilic moieties have been introduced by chemical and mutagenic methods to the soluble N-terminal signaling domain of Shh and tested in both in vitro and in vivo assays. A wide variety of hydrophobic modifications increased the potency of Shh when added at the N-terminus of the protein, ranging from long-chain fatty acids to hydrophobic amino acids, with EC(50) values from 99 nM for the unmodified protein to 0.6 nM for the myristoylated form. The N-myristoylated Shh was as active as the natural form having both N- and C-terminal modifications. The degree of activation appears to correlate with the hydrophobicity of the modification rather than any specific chemical feature of the adduct; moreover, substitution with hydrophilic moieties decreased activity. Hydrophobic modifications at the C-terminus of Shh resulted in only a 2-3-fold increase in activity, and no activation was found with hydrophobic modification at other surface positions. The N-terminal modifications did not appear to alter the binding affinity of the Shh protein for the transfected receptor protein, Patched, and had no apparent effect on structure as measured by circular dichroism, thermal denaturation, and size determination. Activation of Desert Hh through modification of its N-terminus was also observed, suggesting that this is a common feature of Hh proteins.


Subject(s)
Proteins/chemistry , Proteins/physiology , Trans-Activators , Up-Regulation , Acyl Coenzyme A/chemistry , Amides , Amino Acid Substitution/genetics , Animals , Cell Line , Circular Dichroism , Cysteine/chemistry , Cysteine/genetics , Ethylmaleimide/chemistry , Fatty Acids/chemistry , Formaldehyde/chemistry , Hedgehog Proteins , Humans , Indicators and Reagents , Intracellular Signaling Peptides and Proteins , Iodoacetamide/analogs & derivatives , Iodoacetamide/chemistry , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Mice, Inbred C3H , Patched Receptors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding/genetics , Protein Processing, Post-Translational/genetics , Proteins/genetics , Proteins/metabolism , Receptors, Cell Surface , Signal Transduction/genetics , Spectrometry, Mass, Electrospray Ionization , Sulfhydryl Compounds/chemistry , Thiazoles/chemistry , Thiazoles/metabolism , Thiazolidines , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...