Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Structure ; 18(12): 1587-95, 2010 Dec 08.
Article in English | MEDLINE | ID: mdl-21134638

ABSTRACT

FHA domains are well established as phospho-dependent binding modules mediating signal transduction in Ser/Thr kinase signaling networks in both eukaryotic and prokaryotic species. Although they are unique in binding exclusively to phosphothreonine, the basis for this discrimination over phosphoserine has remained elusive. Here, we attempt to dissect overall binding specificity at the molecular level. We first determined the optimal peptide sequence for Rv0020c FHA domain binding by oriented peptide library screening. This served as a basis for systematic mutagenic and binding analyses, allowing us to derive relative thermodynamic contributions of conserved protein and peptide residues to binding and specificity. Structures of phosphopeptide-bound and uncomplexed Rv0020c FHA domain then directed molecular dynamics simulations which show how the extraordinary discrimination in favor of phosphothreonine occurs through formation of additional hydrogen-bonding networks that are ultimately stabilized by van der Waals interactions of the phosphothreonine γ-methyl group with a conserved pocket on the FHA domain surface.


Subject(s)
Phosphothreonine/metabolism , Phosphothreonine/pharmacology , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Crystallography, X-Ray , Forkhead Transcription Factors/metabolism , Humans , Hydrogen Bonding , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phosphothreonine/chemistry , Protein Binding/genetics , Protein Binding/physiology , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Protein Serine-Threonine Kinases/genetics , Structure-Activity Relationship , Substrate Specificity
2.
J Am Chem Soc ; 132(2): 567-75, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20014831

ABSTRACT

Methyl-coenzyme M reductase (MCR) is the key enzyme in methane formation by methanogenic Archaea. It converts the thioether methyl-coenzyme M and the thiol coenzyme B into methane and the heterodisulfide of coenzyme M and coenzyme B. The catalytic mechanism of MCR and the role of its prosthetic group, the nickel hydrocorphin coenzyme F(430), is still disputed, and no intermediates have been observed so far by fast spectroscopic techniques when the enzyme was incubated with the natural substrates. In the presence of the competitive inhibitor coenzyme M instead of methyl-coenzyme M, addition of coenzyme B to the active Ni(I) state MCR(red1) induces two new species called MCR(red2a) and MCR(red2r) which have been characterized by pulse EPR spectroscopy. Here we show that the two MCR(red2) signals can also be induced by the S-methyl- and the S-trifluoromethyl analogs of coenzyme B. (19)F-ENDOR data for MCR(red2a) and MCR(red2r) induced by S-CF(3)-coenzyme B show that, upon binding of the coenzyme B analog, the end of the 7-thioheptanoyl chain of coenzyme B moves closer to the nickel center of F(430) by more than 2 A as compared to its position in both, the Ni(I) MCR(red1) form and the X-ray structure of the inactive Ni(II) MCR(ox1-silent) form. The finding that the protein is able to undergo a conformational change upon binding of the second substrate helps to explain the dramatic change in the coordination environment induced in the transition from MCR(red1) to MCR(red2) forms and opens the possibility that nickel coordination geometries other than square planar, tetragonal pyramidal, or elongated octahedral might occur in intermediates of the catalytic cycle.


Subject(s)
Oxidoreductases/chemistry , Oxidoreductases/metabolism , Phosphothreonine/analogs & derivatives , Binding Sites , Catalytic Domain/drug effects , Crystallography, X-Ray , Electron Spin Resonance Spectroscopy , Models, Molecular , Phosphothreonine/chemistry , Phosphothreonine/metabolism , Phosphothreonine/pharmacology , Protein Conformation/drug effects
3.
Acta Histochem ; 106(1): 3-10, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15032323

ABSTRACT

In previous studies, we have found that negatively charged, but not uncharged, amino acids and sugars block sea urchin fertilization. These studies were developed from modeling work in non-living systems using derivatized agarose beads that suggested that charge-charge bonding may control at least some adhesive interactions. In the present study, the effects of positively charged, negatively charged and uncharged molecules were examined in the sea urchin sperm-egg system in over 300 individual trials. The results indicate that depending on the specific molecules utilized, both sperm and egg are exquisitely sensitive to charged but not uncharged molecules and to pH changes in sea water caused by some of the charged molecules. It is shown that egg activation, as well as sperm motility and sperm-egg interactions, can be affected by charged molecules. One compound, fructose-1-phosphate blocked fertilization in S. purpuratus sea urchins but not in Lytechinus pictus sea urchins. These findings indicate that charge alone cannot explain all the results. In this case, the presence of a ketone instead of an aldehyde group indicates that species-specific components may control fertilization reactions. The present study is a comprehensive survey of the effects of charge, pH and molecular structure on the fertilization activation continuum in a model system of sea urchins.


Subject(s)
Amino Acids/pharmacology , Fertilization/drug effects , Glucosamine/analogs & derivatives , Glucose-6-Phosphate/analogs & derivatives , Sugar Phosphates/pharmacology , Animals , Arginine/pharmacology , Calcimycin/pharmacology , Calcium/pharmacology , Female , Glucosamine/pharmacology , Glucose-6-Phosphate/pharmacology , Hexosephosphates/pharmacology , Hydrogen-Ion Concentration , Male , Monosaccharides/pharmacology , Phosphoserine/pharmacology , Phosphothreonine/pharmacology , Ribosemonophosphates/pharmacology , Sea Urchins , Seawater/chemistry , Sperm Motility/drug effects , Zinc/pharmacology
4.
Biochem Biophys Res Commun ; 203(1): 408-15, 1994 Aug 30.
Article in English | MEDLINE | ID: mdl-8074685

ABSTRACT

Modulator is an endogenous low-molecular weight regulator of both glucocorticoid and mineralocorticoid receptors, as well as protein kinase C. Analogs of the putative modulator structure have been synthesized. These compounds include 1-O-(3'-carboxypropyl) or (5'-carboxypentyl)-L-glycero-3-phospho-L-serine or L-threonine, and the D-glycerol stereoisomers. These compounds were tested for in vitro modulator activity using the glucocorticoid-receptor complex activation inhibition and steroid-binding stabilization assays. One of the ether phosphoglycerides, 1-O-(5'-carboxypentyl)-L-glycero-3-phospho-L-threonine (H-GPT-1), partially inhibited steroid-receptor complex activation in a dose-dependent manner. However, none of the other compounds exhibited any modulator activity towards the glucocorticoid-receptor complex. Like modulator, H-GPT-1 did not inhibit activated glucocorticoid-receptor complex binding to DNA-cellulose. Surprisingly, in contrast to modulator, H-GPT-1 partially inhibited unoccupied receptor steroid-binding in a dose-dependent manner. These results suggest that although modulator is not exactly mimicked by this compound, H-GPT-1 is the first synthetic organic molecule to exhibit some modulator activity towards the glucocorticoid receptor.


Subject(s)
Glycerophosphates/pharmacology , Liver/metabolism , Phosphothreonine/analogs & derivatives , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Adrenalectomy , Animals , Chromatography, Affinity , Cytosol/metabolism , Glycerophosphates/chemical synthesis , Indicators and Reagents , Kinetics , Male , Phosphothreonine/chemical synthesis , Phosphothreonine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/isolation & purification , Receptors, Mineralocorticoid/drug effects , Receptors, Mineralocorticoid/isolation & purification , Structure-Activity Relationship , Triamcinolone Acetonide/metabolism
5.
J Biol Chem ; 268(25): 18491-5, 1993 Sep 05.
Article in English | MEDLINE | ID: mdl-8395504

ABSTRACT

Purified S-methyl-coenzyme M reductase (methylreductase) exhibits a very low fraction of its in vivo activity, suggesting either enzyme inactivation during cell lysis and chromatographic purification or the lack of an activating component in assay mixtures. Evidence that all methylreductase molecules in the purified protein can catalyze slow substrate turnover is found in a study of turnover-dependent in vitro incorporation of radiolabeled HS-CoM at the enzyme active site (Hartzell, P. L., Donnelly, M. I., and Wolfe, R. S. (1987) J. Biol. Chem. 262, 5581-5586). We have conducted active site titrations of purified methylreductase and of a highly active partially purified preparation (Rospert, S., Bocher, R., Albracht, S. P. J., and Thauer, R. K. (1991) FEBS Lett. 291, 371-375) using the reversible competitive inhibitor bromopropanesulfonate (K(i) = 0.05 microM). Curve fitting the data based on an equilibrium binding model shows that 0.1-1.4% of purified methylreductase has functional inhibitor binding sites while up to 25% of a highly active preparation binds the inhibitor. An EPR titration of highly active methylreductase with this inhibitor is consistent with this result, showing that the MCR-red1 and -red2 EPR signals (Albracht, S. P. J., Ankel-Fuchs, D., Bocher, R., Ellermann, J., Moll, J., van der Zwann, J. W., and Thauer, R. K. (1988) Biochim. Biophys. Acta 955, 86-102) are titrated in parallel with this active fraction. Attempts to observe turnover-dependent uptake of radiolabel from [thio-35S]2-methylthioethane-sulfonate by methylreductase were unsuccessful. These results suggest that the low activity of purified methylreductase is due primarily to low percentages of catalytically competent enzyme.


Subject(s)
Oxidoreductases/metabolism , Alkanesulfonates/pharmacology , Binding Sites , Binding, Competitive , Chromatography, High Pressure Liquid , Disulfides/metabolism , Dithiothreitol/pharmacology , Electron Spin Resonance Spectroscopy , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Euryarchaeota/enzymology , Mesna/metabolism , Oxidation-Reduction , Oxidoreductases/antagonists & inhibitors , Phosphothreonine/analogs & derivatives , Phosphothreonine/pharmacology
6.
J Bacteriol ; 174(3): 1007-12, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1732190

ABSTRACT

The 2-(methylthio)ethanesulfonic acid (CH3-S-CoM) reductase catalyzes the final methane-yielding reaction in fastidiously anaerobic methanogenic archaebacteria. This step involves the reductive demethylation of CH3-S-CoM with reducing equivalents from N-7-(mercaptoheptanoyl)-L-threonine O3-phosphate (HS-HTP) to yield methane and the nonsymmetrical disulfide of 2-mercaptoethanesulfonic acid and HS-HTP. We chemically synthesized modified analogs of CH3-S-CoM (which has two carbons in the ethylene bridge) and of HS-HTP (which has seven carbons in the side chain); analog pairs possessed an overall correct number of side chain carbons (i.e., a total of nine in combination). They were simultaneously added to anaerobic cell extracts of Methanobacterium thermoautotrophicum delta H. The ability of the extracts to reductively demethylate the modified substrates was tested by gas chromatography. We also describe here previously unknown inhibitors of methanogenesis, 6-(methylthio)hexanoyl-L-threonine O3-phosphate (a structural analog of HS-HTP) and sodium bromomethanesulfonic acid (a structural analog of CH3-S-CoM). Both analogs were found to be effective competitive inhibitors with respect to HS-HTP. These substrate analogs were also found to inhibit a recently described photoactivation of homogeneous inactive reductase (K. D. Olson, C. W. McMahon, and R. S. Wolfe, Proc. Natl. Acad. Sci. USA 88:4099-4103, 1991). In addition, we probed the mechanism of action of a potent inhibitor of the enzyme, 2-bromoethanesulfonic acid, a structural analog of CH3-S-CoM.


Subject(s)
Methane/metabolism , Methanobacterium/enzymology , Oxidoreductases/metabolism , Binding, Competitive , Enzyme Activation , Enzyme Inhibitors/pharmacology , Kinetics , Light , Mesna/analogs & derivatives , Mesna/metabolism , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/drug effects , Phosphothreonine/analogs & derivatives , Phosphothreonine/pharmacology , Subcellular Fractions/enzymology , Substrate Specificity , Sulfides/pharmacology
7.
Appl Environ Microbiol ; 57(10): 2891-5, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1746950

ABSTRACT

Methanomicrobium mobile requires a heat-stable factor present in ruminal fluid and in boiled cell extract from Methanobacterium thermoautotrophicum for growth. By comparing the growth of M. mobile with boiled cell extract with that observed with various methanogenic cofactors, we found that 7-mercaptoheptanoylthreonine phosphate (HS-HTP) supported sustained growth of M. mobile, at an optimal concentration of 100 microM. No derivatives or possible biosynthetic precursors of HS-HTP could replace HS-HTP as the sole source of growth factor. Results suggest that the growth requirement might be satisfied by 7-mercaptoheptanoic acid plus a second, unidentified heat-stable factor.


Subject(s)
Growth Substances/pharmacology , Methanomicrobiales/growth & development , Phosphothreonine/analogs & derivatives , Culture Media , Hot Temperature , Methanomicrobiales/drug effects , Phosphothreonine/metabolism , Phosphothreonine/pharmacology , Tungsten/pharmacology
8.
Proc Natl Acad Sci U S A ; 88(10): 4099-103, 1991 May 15.
Article in English | MEDLINE | ID: mdl-1903534

ABSTRACT

Inactive 2-(methylthio)ethanesulfonic acid (CH3-S-CoM) reductase was partially activated by exposure to light. This simplified system replaces the complex enzymatic system of protein components A2, A3a, A3b, and ATP, which previously represented the only available means of reactivating the enzyme. Components necessary for light activation include N-(7-mercaptoheptanoyl)-L-threonine O3-phosphate (HS-HTP), CH3-S-CoM, titanium(III) citrate [Ti(III)Cit], and light above 400 nm. Photoactivation was inhibited by known inhibitors of methanogenesis: 2-bromoethanesulfonate (BES), N-(6-mercaptohexanoyl)-L-threonine O3-phosphate, N-(8-mercaptooctanoyl)-L-threonine O3-phosphate, and sodium dithionite. Methanogenesis continued when the light-activated reaction mixture was incubated in the dark. Although the specific activity was low (35 nmol of CH4 per h per mg of protein) the reaction products methane and the unsymmetrical disulfide of 2-mercaptoethanesulfonate (HS-CoM) and HS-HTP were identified. We were unable to photoactivate a reaction mixture containing the isolated prosthetic group, native F430, or its analogues.


Subject(s)
Euryarchaeota/enzymology , Light , Oxidoreductases/metabolism , Citrates/pharmacology , Citric Acid , Disulfides/metabolism , Enzyme Activation/radiation effects , Mesna/analogs & derivatives , Mesna/pharmacology , Methane/metabolism , Phosphothreonine/analogs & derivatives , Phosphothreonine/pharmacology , Photochemistry
9.
J Bacteriol ; 173(8): 2481-7, 1991 Apr.
Article in English | MEDLINE | ID: mdl-2013570

ABSTRACT

Methyl coenzyme M methylreductase from acetate-grown Methanosarcina thermophila TM-1 was purified 16-fold from a cell extract to apparent homogeneity as determined by native polyacrylamide gel electrophoresis. Ninety-four percent of the methylreductase activity was recovered in the soluble fraction of cell extracts. The estimated native molecular weight of the enzyme was between 132,000 (standard deviation [SD], 1,200) and 141,000 (SD, 1,200). Denaturing polyacrylamide gel electrophoresis revealed three protein bands corresponding to molecular weights of 69,000 (SD, 1,200), 42,000 (SD, 1,200), and 33,000 (SD, 1,200) and indicated a subunit configuration of alpha 1 beta 1 gamma 1. As isolated, the enzyme was inactive but could be reductively reactivated with titanium (III) citrate or reduced ferredoxin. ATP stimulated enzyme reactivation and was postulated to be involved in a conformational change of the inactive enzyme from an unready state to a ready state that could be reductively reactivated. The temperature and pH optima for enzyme activity were 60 degrees C and between 6.5 and 7.0, respectively. The active enzyme contained 1 mol of coenzyme F430 per mol of enzyme (Mr, 144,000). The Kms for 2-(methylthio)ethane-sulfonate and 7-mercaptoheptanoylthreonine phosphate were 3.3 mM and 59 microM, respectively.


Subject(s)
Euryarchaeota/metabolism , Multienzyme Complexes , Oxidoreductases/chemistry , Adenosine Triphosphate/pharmacology , Aldehyde Oxidoreductases/pharmacology , Amino Acid Sequence , Centrifugation, Density Gradient , Citrates/pharmacology , Citric Acid , Electrophoresis, Polyacrylamide Gel , Ferredoxins/pharmacology , Hydrogen-Ion Concentration , Molecular Sequence Data , Oxidoreductases/genetics , Phosphothreonine/analogs & derivatives , Phosphothreonine/pharmacology , Sequence Homology, Nucleic Acid , Temperature
10.
Biochem Biophys Res Commun ; 168(2): 566-73, 1990 Apr 30.
Article in English | MEDLINE | ID: mdl-1692212

ABSTRACT

Arginine vasopressin administration (10(-10)-10(-6) M) to isolated human platelets induces an increase in the specific immunoblotting of a 38 kDa protein revealed by a phosphotyrosine antibody. This signal is biphasic with maximal stimulation within one minute. Neither forskolin (10(-5) M) nor phorbol ester (10(-6) M) produces a similar 38 kDa signal. The specific immunoblotted signals are competitively abolished by 1 mM phosphotyrosine but not phosphoserine or phosphothreonine. Electrophoretic separation at pH 3.5 of the acid hydrolysates of the 38 kDa proteins reveals a vasopressin dependent increase in levels of phosphotyrosine as well as phosphoserine and phosphothreonine. The 38 kDa phosphorylation is also induced by the specific arginine vasopressin V1 receptor agonist (Phe2Orn8Vastocina) and blocked by the V1 receptor antagonist [desGly(NH2)d(CH2)5Tyr(Me) AVPb]. These observations suggest that arginine vasopressin signal transduction may be associated with the tyrosine phosphorylation of a 38 kDa protein.


Subject(s)
Arginine Vasopressin/pharmacology , Blood Platelets/enzymology , Protein-Tyrosine Kinases/metabolism , Angiotensin Receptor Antagonists , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Blotting, Western , Colforsin/pharmacology , Dose-Response Relationship, Drug , Electrophoresis , Humans , In Vitro Techniques , Phorbol Esters/pharmacology , Phosphorylation , Phosphoserine/metabolism , Phosphoserine/pharmacology , Phosphothreonine/metabolism , Phosphothreonine/pharmacology , Phosphotyrosine , Receptors, Vasopressin , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
11.
Biochim Biophys Acta ; 788(1): 58-61, 1984 Jul 17.
Article in English | MEDLINE | ID: mdl-6743663

ABSTRACT

Arylsulfatase B (aryl-sulfate sulfohydrolase, EC 3.1.6.1) purified from rabbit liver is competitively inhibited at modest concentrations by a variety of phosphate esters derived from amino acids, amines and simple sugars. Phospho-L-serine coupled to Sepharose 4B could be used as an affinity column to enhance the purity of a crude preparation of the enzyme. It is suggested that phosphate esters containing functional groups can be used to obtain affinity reagents to purify arylsulfatases and also to probe their active sites.


Subject(s)
Chondro-4-Sulfatase/antagonists & inhibitors , Liver/enzymology , Organophosphates/pharmacology , Organophosphorus Compounds/pharmacology , Sugar Phosphates/pharmacology , Sulfatases/antagonists & inhibitors , Animals , Binding, Competitive , Chemical Phenomena , Chemistry, Physical , Chondro-4-Sulfatase/isolation & purification , Chromatography, Affinity , Glucose-6-Phosphate , Glucosephosphates/pharmacology , Phosphoserine/pharmacology , Phosphothreonine/pharmacology , Rabbits , Structure-Activity Relationship
12.
Neurosci Lett ; 31(1): 59-64, 1982 Jul 20.
Article in English | MEDLINE | ID: mdl-6126848

ABSTRACT

Acidic amino acid analogues varying in their omega-terminal were evaluated: (1) as neuronal excitants or antagonists of excitatory synaptic transmission, and (2) as inhibitors of L-[3H]glutamate binding to synaptic membranes. omega-Phosphonates were antagonists and inhibited L-glutamate binding to the 2-amino-4-phosphonobutyrate (APB)-sensitive population of binding sites; omega-sulfonates and omega-carboxylates were excitants and inhibited L-glutamate binding to APB-sensitive and -insensitive sites. The data indicate that properties of the omega-acidic group are important for establishing the relative potencies of antagonist substances and the overall excitatory/antagonist activity of these analogues.


Subject(s)
Glutamates/metabolism , Hippocampus/metabolism , Receptors, Cell Surface/metabolism , Synaptic Transmission , Aminobutyrates/pharmacology , Animals , Evoked Potentials/drug effects , Glutamic Acid , Hippocampus/drug effects , Phosphoserine/pharmacology , Phosphothreonine/pharmacology , Rats , Receptors, Cell Surface/drug effects , Receptors, Glutamate , Stereoisomerism , Structure-Activity Relationship , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...