Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 107(1): 430-5, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19966283

ABSTRACT

Thermogenesis by resting muscle varies with conditions and plays an active role in homeostasis of body weight. The low metabolic rate of living resting muscles requires that ATP turnover by myosin be inhibited relative to the purified protein in vitro. This inhibition has not been previously seen in in vitro systems. We used quantitative epifluorescence microscopy of fluorescent nucleotides to measure single nucleotide turnovers in relaxed, permeable skeletal muscle fibers. We observed two lifetimes for nucleotide release by myosin: a fast component with a lifetime of approximately 20 s, similar to that of purified myosin, and a slower component with a lifetime of 230 +/- 24 s. We define the latter component to be the "super relaxed state." The fraction of myosins in the super relaxed state was decreased at lower temperatures, by substituting GTP for ATP or by increased levels of myosin phosphorylation. All of these conditions have also been shown to cause increased disorder in the structure of the thick filament. We propose a model in which the structure of the thick filament modulates the nucleotide turnover rates of myosin in relaxed fibers. Modulation of the relative populations of the super relaxed and conventional relaxed states could have a profound effect on muscle thermogenesis, with the capacity to also significantly alter whole-body metabolic rate.


Subject(s)
Adenosine Triphosphate/metabolism , Muscle Fibers, Skeletal/metabolism , Myosins/metabolism , Thermogenesis/physiology , Adenosine Triphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Animals , Energy Metabolism , Fluorescent Dyes/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle Relaxation/physiology , Nucleotides/chemistry , Nucleotides/metabolism , Rabbits
2.
Biochemistry ; 49(5): 1005-13, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20038140

ABSTRACT

In contrast to all other known pyrophosphatases, Moorella thermoacetica pyrophosphatase (mtCBS-PPase) contains nucleotide-binding CBS domains and is thus strongly regulated by adenine nucleotides. Stopped-flow measurements using a fluorescent AMP analogue, 2'(3')-O-(N-methylanthranoyl)-AMP (Mant-AMP), reveal that nucleotide binding to mtCBS-PPase involves a three-step increase in Mant-AMP fluorescence with relaxation times from 0.01 to 100 s, implying conformational changes in the complex. This effect is reversed by AMP. Metal cofactors (Co(2+) and Mg(2+)) enhance the fluorescence signal but are not absolutely required, unlike what is seen when the catalytic reaction is examined. The relaxation times and amplitudes of the fluorescence signals depend on Mant-AMP concentration in a manner suggestive of the presence of a second binding site for Mant-AMP on the protein. Equilibrium fluorescence titration experiments additionally support the presence of two types of AMP binding sites with different affinities, whereas equilibrium dialysis and membrane filtration measurements reveal binding of one AMP molecule per enzyme monomer, implying negative cooperativity in nucleotide binding. The substrate (PP(i)) modulates Mant-AMP binding, leading to a further conformational change in the enzyme-Mant-AMP complex, and stimulates mtCBS-PPase in alkaline medium within a time scale of minutes, via conversion to a more active form. This active form initially comprises only a third of the enzyme, as estimated from kinetic titration with ADP. AMP inhibits both enzyme forms but is unable to independently induce interconversion. Our results collectively suggest that nucleotides and the substrate induce multiple conformational changes in mtCBS-PPase occurring over a wide time scale; the changes are distinct and almost independent.


Subject(s)
Adenosine Diphosphate/chemistry , Adenosine Monophosphate/chemistry , Bacterial Proteins/chemistry , Cystathionine beta-Synthase/chemistry , Inorganic Pyrophosphatase/chemistry , Nucleic Acid Conformation , Thermoanaerobacter/enzymology , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/chemistry , DNA-Binding Proteins/chemistry , Fluorescent Dyes/chemistry , Inorganic Pyrophosphatase/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Substrate Specificity/genetics
3.
Mol Pharmacol ; 75(3): 693-703, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19056899

ABSTRACT

Bacillus anthracis causes anthrax disease and exerts its deleterious effects by the release of three exotoxins: lethal factor, protective antigen, and edema factor (EF), a highly active calmodulin-dependent adenylyl cyclase (AC). However, conventional antibiotic treatment is ineffective against either toxemia or antibiotic-resistant strains. Thus, more effective drugs for anthrax treatment are needed. Previous studies from our laboratory showed that mammalian membranous AC (mAC) exhibits broad specificity for purine and pyrimidine nucleotides ( Mol Pharmacol 70: 878-886, 2006 ). Here, we investigated structural requirements for EF inhibition by natural purine and pyrimidine nucleotides and nucleotides modified with N-methylanthraniloyl (MANT)- or anthraniloyl groups at the 2'(3')-O-ribosyl position. MANT-CTP was the most potent EF inhibitor (K(i), 100 nM) among 16 compounds studied. MANT-nucleotides inhibited EF competitively. Activation of EF by calmodulin resulted in effective fluorescence resonance energy transfer (FRET) from tryptophan and tyrosine residues located in the vicinity of the catalytic site to MANT-ATP, but FRET to MANT-CTP was only small. Mutagenesis studies revealed that Phe586 is crucial for FRET to MANT-ATP and MANT-CTP and that the mutations N583Q, K353A, and K353R differentially alter the inhibitory potencies of MANT-ATP and MANT-CTP. Docking approaches relying on crystal structures of EF indicate similar binding modes of the MANT nucleotides with subtle differences in the region of the nucleobases. In conclusion, like mAC, EF accommodates both purine and pyrimidine nucleotides. The unique preference of EF for the base cytosine offers an excellent starting point for the development of potent and selective EF inhibitors.


Subject(s)
Adenylyl Cyclases/metabolism , Anthrax Vaccines/metabolism , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Purine Nucleotides/metabolism , Pyrimidine Nucleotides/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Adenylyl Cyclases/chemistry , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Animals , Anthrax Vaccines/chemistry , Anthrax Vaccines/genetics , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Catalysis , Cattle , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , Mutagenesis, Site-Directed , Protein Binding/genetics , Purine Nucleotides/chemistry , Pyrimidine Nucleotides/chemistry , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/metabolism
4.
J Mol Biol ; 377(1): 193-205, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18237742

ABSTRACT

DEAD-box proteins are ATPase enzymes that destabilize and unwind duplex RNA. Quantitative knowledge of the ATPase cycle parameters is critical for developing models of helicase activity. However, limited information regarding the rate and equilibrium constants defining the ATPase cycle of RNA helicases is available, including the distribution of populated biochemical intermediates, the catalytic step(s) that limits the enzymatic reaction cycle, and how ATP utilization and RNA interactions are linked. We present a quantitative kinetic and equilibrium characterization of the ribosomal RNA (rRNA)-activated ATPase cycle mechanism of DbpA, a DEAD-box rRNA helicase implicated in ribosome biogenesis. rRNA activates the ATPase activity of DbpA by promoting a conformational change after ATP binding that is associated with hydrolysis. Chemical cleavage of bound ATP is reversible and occurs via a gamma-phosphate attack mechanism. ADP-P(i) and RNA binding display strong thermodynamic coupling, which causes DbpA-ADP-P(i) to bind rRNA with >10-fold higher affinity than with bound ATP, ADP or in the absence of nucleotide. The rRNA-activated steady-state ATPase cycle of DbpA is limited both by ATP hydrolysis and by P(i) release, which occur with comparable rates. Consequently, the predominantly populated biochemical states during steady-state cycling are the ATP- and ADP-P(i)-bound intermediates. Thermodynamic linkage analysis of the ATPase cycle transitions favors a model in which rRNA duplex destabilization is linked to strong rRNA and nucleotide binding. The presented analysis of the DbpA ATPase cycle reaction mechanism provides a rigorous kinetic and thermodynamic foundation for developing testable hypotheses regarding the functions and molecular mechanisms of DEAD-box helicases.


Subject(s)
Adenosine Triphosphatases/metabolism , DEAD-box RNA Helicases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/metabolism , Hydrolysis , Kinetics , Oxygen Isotopes , Peptidyl Transferases/metabolism , Phosphates/metabolism , RNA, Bacterial/metabolism , RNA, Ribosomal/metabolism , Thermodynamics
5.
Biochemistry ; 46(33): 9654-64, 2007 Aug 21.
Article in English | MEDLINE | ID: mdl-17661443

ABSTRACT

The interaction of gamma-amido-ATP (ATPN) and its 2'(3')-O-methylanthraniloyl derivative (mantATPN) with skeletal myosin subfragment 1 (S1) and actomyosin (actoS1) was studied in stopped-flow experiments. Tryptophan fluorescence and fluorescence of the mant label or light scattering were measured simultaneously. Information about the binding of mant nucleotides was obtained from the quenching of tryptophan fluorescence by the mant label. The parameters of various kinetic models were fitted to the experimental traces. The high-fluorescence state of S1 forms with ATPN at a rate of 95 s-1 ("open-closed" transition); the transition is only slowly reversible, in contrast to the very fast equilibrium seen with its better known isomer AMPPNP [Urbanke, C., and Wray, J. (2001) Biochem. J. 358, 165-173]. The stabilization of the closed state of myosin by ATPN may be due to the formation of a complex with a pentacoordinated amido-gamma-phosphate, from which ATPN can dissociate at a rate of 0.005 s-1 or be hydrolyzed by cleavage of the beta-gamma bond at a rate of 2.5 x 10(-4) s-1. A corresponding actoS1-ATPN complex with myosin in the "closed" conformation is the first detectable intermediate in the association of actin and S1-ATPN, giving an experimental access to a state analogous to a key intermediate in the cross-bridge cycle.


Subject(s)
Actins/chemistry , Adenylyl Imidodiphosphate/analogs & derivatives , Myosins/chemistry , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/chemistry , Adenylyl Imidodiphosphate/chemistry , Animals , Dictyostelium/metabolism , Fluorescence , Protein Conformation , Spectrometry, Fluorescence , Tryptophan/analysis , ortho-Aminobenzoates/chemistry
6.
Biochemistry ; 45(30): 9074-84, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16866353

ABSTRACT

The dsRNA-activated protein kinase, PKR, plays a pivotal role in the cellular antiviral response. PKR contains an N-terminal dsRNA binding domain (dsRBD) and a C-terminal kinase domain. An autoinhibition model has been proposed in which latent PKR exists in a closed conformation where the substrate binding cleft of the kinase is blocked by the dsRBD. Binding to dsRNA activates the enzyme by inducing an open conformation and enhancing dimerization. We have tested this model by characterizing the affinity and kinetics of binding of a nucleotide substrate to PKR. The fluorescent nucleotide mant-AMPPNP binds to unactivated PKR with a Kd of approximately 30 microM, and the affinity is not strongly affected by autophosphorylation or binding to dsRNA. We observe biphasic binding kinetics in which the fast phase depends on ligand concentration but the slow phase is ligand-independent. The kinetic data fit to a two-step model of ligand binding followed by a slow conformation change. The kinetics are also not strongly affected by phosphorylation state or dsRNA binding. Thus, the equilibrium and kinetic data indicate that the substrate accessibility of the kinase is not modulated by PKR activation state as predicted by the autoinhibition model. In atomic force microscopy images, monomers of the latent protein are resolved with three separate regions linked by flexible, bridgelike structures. The resolution of the individual domains in the images supports a model in which unactivated PKR exists in an open conformation where the kinase domain is accessible and capable of binding substrate.


Subject(s)
DNA-Binding Proteins/metabolism , Nucleotides/metabolism , eIF-2 Kinase/metabolism , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/chemistry , Adenylyl Imidodiphosphate/metabolism , Binding Sites , Binding, Competitive , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/chemistry , Dimerization , Enzyme Activation/physiology , Fluorescence Polarization , Kinetics , Nucleotides/chemistry , Phosphorylation , Protein Binding , Protein Conformation , Spectrometry, Fluorescence , Substrate Specificity , eIF-2 Kinase/chemistry
7.
FEBS Lett ; 518(1-3): 97-100, 2002 May 08.
Article in English | MEDLINE | ID: mdl-11997025

ABSTRACT

On binding to myosin subfragment 1 (S1), the gamma-amido derivative of ATP (ATPgammaNH2), an isomer of adenosine 5'-[beta,gamma-imido]-triphosphate (AMPPNP), induces a larger increase in the intrinsic (tryptophan) fluorescence than is seen with ATP. A binding constant of 1.7x10(7) M(-1) was measured for ATPgammaNH2, compared to 2.1-2.4x10(7) M(-1) for AMPPNP. ATPgammaNH2 was hydrolyzed only very slowly by S1. ATPgammaNH2 appears to stabilize the 'closed' conformation of S1, and does so without cleavage of the beta-gamma phosphate bond. The dissociation of actin-S1 by ATPgammaNH2 and that of S1.ATPgammaNH2 by actin are both strikingly slow.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Myosin Subfragments/chemistry , Actomyosin/metabolism , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/metabolism , Animals , Fluorescence , Hydrolysis , Kinetics , Myosin Subfragments/metabolism , Rabbits , Temperature , Tryptophan/chemistry
8.
J Biol Chem ; 275(41): 31857-68, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-10903310

ABSTRACT

Benzoyl-CoA reductase catalyzes the two-electron transfer from a reduced ferredoxin to the aromatic ring of benzoyl-CoA; this reaction is coupled to stoichiometrical ATP hydrolysis. A very low reduction potential (less than -1 V) is required for the first electron transfer to the aromatic ring. In this work the nature of the redox centers of purified benzoyl-CoA reductase from Thauera aromatica was studied by EPR and Mössbauer spectroscopy. The results obtained indicated the presence of three [4Fe-4S] clusters. Redox titration studies revealed that the reduction potentials of all three clusters were below -500 mV. The previously reported S = 7/2 state of the enzyme during benzoyl-CoA-independent ATPase activity (Boll, M., Albracht, S. J. P., and Fuchs, G. (1997) Eur. J. Biochem. 244, 840-851) was confirmed by Mössbauer spectroscopy. Inactivation by oxygen was associated with the irreversible conversion of part of the [4Fe-4S] clusters to [3Fe-4S] clusters. Acetylene stimulated the benzoyl-CoA-independent ATPase activity and induced novel EPR signals with g(av) >2. The presence of simple cubane clusters in benzoyl-CoA reductase as the sole redox-active metal centers demonstrates novel aspects of [4Fe-4S] clusters since they adopt the role of elemental sodium or lithium which are used as electron donors in the analogous chemical Birch reduction of aromatic rings.


Subject(s)
Iron-Sulfur Proteins/chemistry , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/chemistry , Thauera/enzymology , Acetylene/pharmacology , Acyl Coenzyme A/metabolism , Acyl Coenzyme A/pharmacology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/pharmacology , Affinity Labels , Azides/pharmacology , Binding Sites , Dithionite/pharmacology , Electron Spin Resonance Spectroscopy , Enzyme Activation/drug effects , Oxidation-Reduction , Oxygen/metabolism , Oxygen/pharmacology , Spectroscopy, Mossbauer , Temperature
9.
Biochemistry ; 39(8): 2106-22, 2000 Feb 29.
Article in English | MEDLINE | ID: mdl-10684661

ABSTRACT

The kinetic mechanism of binding nucleotide cofactors to the Escherichia coli primary replicative helicase DnaB protein has been studied, using the fluorescence stopped-flow technique. The experiments have been performed with fluorescent ATP and ADP analogues bearing the modification on the ribose, MANT-AMP-PNP and MANT-ADP, and on the base, epsilonAMP-PNP and epsilonADP. Association of the DnaB helicase with nucleotide cofactors is characterized by four relaxation times that indicate that the binding occurs by a minimum of four-steps. The simplest mechanism which can describe the data is a four-step sequential process where the bimolecular binding step is followed by three isomerization steps. This mechanism is described by the following equation: [equation in text]. The binding mechanism is independent of the location of the nucleotide cofactor modification and is an intrinsic property of the DnaB helicase-nucleotide system. Quantitative amplitude analyses, using the matrix projection operator technique, allowed us to determine specific fluorescence changes accompanying the formation of all intermediates relative to the fluorescence of the free nucleotide. It shows that the major conformational change of the DnaB helicase-nucleotide complex occurs in the formation of the (H-N)(1). Moreover, the value of the bimolecular rate constant, k(1), is 3-4 orders of magnitude lower than the value expected for the diffusion-controlled reaction. These results indicate that the determined first step includes formation of the collision and an additional transition of the enzyme-nucleotide complex. The obtained results provide evidence of profoundly different conformational states of the ribose and base regions of the nucleotide-binding site in different intermediates. The sequential nature of the mechanism of the nucleotide binding to the DnaB helicase indicates the lack of the existence of a kinetically significant conformational equilibrium of the helicase protomer and the DnaB hexamer prior to the binding. The significance of these results for the functioning of the DnaB helicase is discussed.


Subject(s)
DNA Helicases/metabolism , Fluorescent Dyes , Nucleotides/metabolism , Ribose/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/metabolism , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Computer Simulation , DNA Helicases/chemistry , DnaB Helicases , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Kinetics , Models, Chemical , Models, Statistical , Protein Conformation , Spectrometry, Fluorescence , Thermodynamics , Time Factors , ortho-Aminobenzoates/metabolism
10.
Proc Natl Acad Sci U S A ; 96(16): 9106-11, 1999 Aug 03.
Article in English | MEDLINE | ID: mdl-10430903

ABSTRACT

Members of the kinesin superfamily are force-generating ATPases that drive movement and influence cytoskeleton organization in cells. Often, more than one kinesin is implicated in a cellular process, and many kinesins are proposed to have overlapping functions. By using conventional kinesin as a model system, we have developed an approach to activate or inhibit a specific kinesin allele in the presence of other similar motor proteins. Modified ATP analogs are described that do not activate either conventional kinesin or another superfamily member, Eg5. However, a kinesin allele with Arg-14 in its nucleotide binding pocket mutated to alanine can use a subset of these nucleotide analogs to drive microtubule gliding. Cyclopentyl-ATP is one such analog. Cyclopentyl-adenylylimidodiphosphate, a nonhydrolyzable form of this analog, inhibits the mutant allele in microtubule-gliding assays, but not wild-type kinesin or Eg5. We anticipate that the incorporation of kinesin mutants and allele-specific activators and inhibitors in in vitro assays should clarify the role of individual motor proteins in complex cellular processes.


Subject(s)
Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/pharmacology , Kinesins/drug effects , Kinesins/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/metabolism , Binding Sites , Dimerization , Humans , Kinesins/chemistry , Kinetics , Macromolecular Substances , Models, Molecular , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Structure-Activity Relationship
11.
Biochemistry ; 38(28): 9063-8, 1999 Jul 13.
Article in English | MEDLINE | ID: mdl-10413480

ABSTRACT

S-Adenosylmethionine (AdoMet) synthetase catalyzes the only known route of biosynthesis of the primary in vivo alkylating agent. Inhibitors of this enzyme could provide useful modifiers of biological methylation and polyamine biosynthetic processes. The AdoMet synthetase catalyzed reaction converts ATP and L-methionine to AdoMet, PP(i), and P(i), with formation of tripolyphosphate as a tightly bound intermediate. This work describes a nonhydrolyzable analogue of the tripolyphosphate (PPP(i)) reaction intermediate, diimidotriphosphate (O(3)P-NH-PO(2)-NH-PO(3)(5)(-)), as a potent inhibitor. In the presence of AdoMet, PNPNP is a slow-binding inhibitor with an overall inhibition constant (K(i)) of 2 nM and a dissociation rate of 0.6 h(-)(1). In contrast, in the absence of AdoMet PNPNP is a classical competitive inhibitor with a K(i) of 0.5 microM, a slightly higher affinity than PPP(i) itself (K(i) = 3 microM). The imido analogue of the product pyrophosphate, imidodiphosphate (O(3)P-NH-PO(3)(4)(-)) also displays slow onset inhibition only in the presence of AdoMet, with a K(i) of 0.8 microM, compared to K(i) of 250 microM for PP(i). Circular dichroism spectra of the unliganded enzyme and various complexes are indistinguishable indicating that the protein secondary structure is not greatly altered upon complex formation, suggesting local rearrangements at the active site during the slow binding process. A model based on ionization of the bridging -NH- moiety is presented which could account for the potent inhibition by PNP and PNPNP.


Subject(s)
Diphosphates/chemistry , Enzyme Inhibitors/chemistry , Methionine Adenosyltransferase/antagonists & inhibitors , Methionine Adenosyltransferase/metabolism , Polyphosphates/chemistry , Acid Anhydride Hydrolases/antagonists & inhibitors , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/chemistry , Amino Acid Substitution/genetics , Arginine/genetics , Binding, Competitive/genetics , Diphosphates/pharmacology , Diphosphonates/chemistry , Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Hydrolysis/drug effects , Leucine/genetics , Methionine Adenosyltransferase/genetics , Mutagenesis, Site-Directed , Polyphosphates/pharmacology
12.
Eur J Biochem ; 252(1): 155-61, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9523725

ABSTRACT

Limited trypsin digestion of isolated F1 removed 15 and 7 amino acids from the N-termini of the alpha and beta subunits respectively and left other subunits untouched as shown by electrophoresis, immunoblotting and protein sequencing. The cooperativity for ATP hydrolysis by soluble F1 was impaired by trypsin digestion. The Km2 obtained from Eadie-Hofstee plots apparently decreased in trypsin-digested F1 but the affinity for adenosine 5'-[beta,gamma-imido]triphosphate (AdoPP[NH]P) and GTP hydrolysis was not influenced. The inhibition of ATP hydrolysis by ADP was attenuated by trypsin digestion. Trypsin digestion of F1 did not affect its capacity to bind to Fo nor did it alter the sensitivity of ATP hydrolysis in the F1Fo reconstituted system to oligomycin and N,N'-dicyclohexylcarbodiimide. The cleavage of the alpha and beta subunits did, on the other hand impair: (a) the ATP-driven proton pumping in the reconstituted F1Fo complex: (b) the inhibition by F1 of passive proton conduction in Fo; (c) the inhibition of passive proton conduction in Fo by AdoPP[NH]P binding to F1. These results show that the limited cleavage of the N-termini of the alpha and beta subunits, located on the top of F1, results in decoupling of catalysis from proton transport. The possible relationship of these observations with the binding change rotatory model of the F1Fo ATP synthase is discussed.


Subject(s)
Mitochondria, Heart/enzymology , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/physiology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/metabolism , Amino Acid Sequence , Animals , Catalysis , Cattle , Dicyclohexylcarbodiimide/pharmacology , Fluorescent Dyes/metabolism , Guanosine Triphosphate/metabolism , Hydrolysis , Kinetics , Molecular Sequence Data , Oligomycins/pharmacology , Peptide Fragments/chemistry , Protons , Sequence Analysis , Trypsin/metabolism , Urea/pharmacology
13.
Biochim Biophys Acta ; 1144(3): 419-25, 1993 Oct 04.
Article in English | MEDLINE | ID: mdl-8399286

ABSTRACT

Two ATP analogs, 2- and 8-azidoadenyl-5'-yl imidodiphosphate, were synthesized, purified and utilized as inhibitors of soluble beef heart mitochondrial F1-ATPase under non-photolytical conditions. In the range of 5 microM to 3 mM ATP, the initial rates of ATP hydrolysis in the presence and absence of the inhibiting ATP analogs can be adequately described by two pairs of Km and Vmax values (3 microM, 8.5 mumol ATP/min per mg; 255 microM, 42.0 mumol ATP/min per mg). With increasing inhibitor concentrations, the apparent Km,2 increases as in competitive inhibition, while Vmax,1 decreases as in non-competitive inhibition. The Ki values derived for both types of inhibition are similar, but strongly different for 2- and 8-azido-AMP-PNP (4 microM and 460 microM, respectively). The decrease of the high-affinity Vmax is compensated by an increase in low-affinity catalysis, resulting in a constant sum of maximal velocities. These data can be described by a model where two sites interact with negative cooperativity in binding of substrate.


Subject(s)
Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/analogs & derivatives , Mitochondria, Heart/enzymology , Proton-Translocating ATPases/antagonists & inhibitors , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemical synthesis , Adenylyl Imidodiphosphate/pharmacology , Animals , Cattle , Kinetics , Proton-Translocating ATPases/metabolism
14.
J Biol Chem ; 260(23): 12875-83, 1985 Oct 15.
Article in English | MEDLINE | ID: mdl-2413035

ABSTRACT

Movements of DNA polymerase III holoenzyme (holoenzyme) in replicating a template multiprimed with synthetic pentadecadeoxynucleotides (15-mers) annealed at known positions on a single-stranded circular or linear DNA have been analyzed. After extension of one 15-mer on a multiprimed template, holoenzyme moves downstream in the direction of chain elongation to the next primer. Holoenzyme readily traverses a duplex, even 400 base pairs long, to exploit its 3'-hydroxyl end as the next available primer. This downstream polarity likely results from an inability to diffuse upstream along single-stranded DNA. These holoenzyme movements, unlike formation of the initial complex with a primer, do not require ATP. Time elapsed between completion of a chain and initiation on the next downstream primer is rapid (1 s or less); dissociation of holoenzyme to form a complex with another primed template is slow (1-2 min). Thus, holoenzyme diffuses rapidly only on duplex DNA, probably in both directions, and forms an initiation complex with the first primer encountered. Based on these findings, schemes can be considered for holoenzyme action at the replication fork of a duplex chromosome.


Subject(s)
DNA Polymerase III/metabolism , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Escherichia coli/enzymology , Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/pharmacology , Bacteriophage phi X 174/genetics , Base Composition , DNA Restriction Enzymes , DNA, Circular/metabolism , DNA, Single-Stranded/metabolism , DNA, Viral/metabolism , Deoxyadenine Nucleotides/metabolism , Deoxyribonucleotides/metabolism , Kinetics , Nucleic Acid Hybridization , RNA/metabolism , Templates, Genetic , Thymine Nucleotides/metabolism
15.
Biochim Biophys Acta ; 787(2): 113-21, 1984 Jun 14.
Article in English | MEDLINE | ID: mdl-6375727

ABSTRACT

Acrylamide fluorescence quenching of 1,N6- ethenoadenylyl imidodiphosphate (e-AMPPNP) bound reversibly to the ATP binding sites of heavy meromyosin was investigated. In the presence of a 6-fold excess of heavy meromyosin over e-AMPPNP, a modified Stern-Volmer plot was not a linear function of the inverse of the concentration of acrylamide used as a quencher. By analyzing the plot, two Stern-Volmer constants (0.89 M-1 and 13 M-1) were obtained for bound e-AMPPNP. About 94% of bound e-AMPPNP had the Stern-Volmer constant of 0.89 M-1 and 6% of bound e-AMPPNP had the Stern-Volmer constant of 13 M-1. Moreover, in the presence of a 10-fold excess of e-AMPPNP over heavy meromyosin, a curved Stern-Volmer plot was obtained. It was found from this plot that 50% of bound e-AMPPNP had the Stern-Volmer constant of 13 M-1 and the remainder had the Stern-Volmer constant of 0.89 M-1. From these and the above data, it was concluded that one of the two ATP-binding sites of heavy meromyosin binds e-AMPPNP much more tightly than the other site does, and that the fluorescent group of e-AMPPNP bound to the former site is strongly isolated from the solvent as compared with that of e-AMPPNP bound to the latter site.


Subject(s)
Adenosine Triphosphate/metabolism , Myosin Subfragments/metabolism , Adenylyl Imidodiphosphate/analogs & derivatives , Animals , Binding Sites , Fluorescent Dyes , Kinetics , Muscles/metabolism , Myosins/metabolism , Peptide Fragments/metabolism , Protein Binding , Rabbits
16.
Nucleic Acids Res ; 11(23): 8369-87, 1983 Dec 10.
Article in English | MEDLINE | ID: mdl-6324083

ABSTRACT

A modified phosphotriester method has been employed for the efficient chemical synthesis of long-chain deoxyribooligonucleotides. During the course of this work, a general and rapid procedure was developed for the preparation of 24-62-mers in solution. Preparative reversed phase column chromatography on silanized silica gel was used to purify triester intermediates starting from 10-mers. The rapid synthesis of 32-mer and 42-mer on glass and silica gel supports using suitably protected 2-8-mer blocks as coupling units has been also accomplished. In particular, a convenient procedure for the solid-phase synthesis of oligonucleotide blocks bearing 3'-terminal phosphodiester groups is described.


Subject(s)
Oligodeoxyribonucleotides/chemical synthesis , Oligonucleotides/chemical synthesis , Adenylyl Imidodiphosphate/analogs & derivatives , Chromatography, High Pressure Liquid , Glass , Imidazoles , Indicators and Reagents , Silicon Dioxide , Structure-Activity Relationship
17.
Biochim Biophys Acta ; 682(1): 124-34, 1982 Oct 18.
Article in English | MEDLINE | ID: mdl-6215942

ABSTRACT

(1)N-4-Azido-2-nitrophenyl-gamma-[3H]aminobutyryl-AdoPP[NH] P(NAP4-AdoPP[NH]P) a photoactivable derivative of 5-adenylyl imidodiphosphate (AdoPP[NH]P), was synthesized. (2) Binding of [3H]NAP4-AdoPP[NH]P to soluble ATPase from beef heart mitochondria (F1) was studied in the absence of photoirradiation, and compared to that of [3H]AdoPP[NH]P. The photoactivable derivative of AdoPP[NH]P was found to bind to F1 with high affinity, like AdoPP[NH]P. Once [3H]NAP4-AdoPP[NH]P had bound to F1 in the dark, it could be released by AdoPP[NH]P, ADP and ATP, but not at all by NAP4 or AMP. Furthermore, preincubation of F1 with unlabeled AdoPP[NH]P, ADP, or ATP prevented the covalent labeling of the enzyme by [3H]NAP4-AdoPP[NH]P upon photoirradiation. (3) Photoirradiation of F1 by [3H]NAP4-AdoPP[NH]P resulted in covalent photolabeling and concomitant inactivation of the enzyme. Full inactivation corresponded to the binding of about 2 mol [3H]NAP4-AdoPP[NH]P/mol F1. Photolabeling by NAP4-AdoPP[NH]P was much more efficient in the presence than in the absence of MgCl2. (4) Bound [3H]NAP4-AdoPP[NH]P was localized on the alpha- and beta- subunits of F1. At low concentrations (less than 10 microM), bound [3H]NAP4-AdoPP[NH]P was predominantly localized on the alpha-subunit; at concentrations equal to, or greater than 75 microM, both alpha- and beta-subunits were equally labeled. (5) The extent of inactivation was independent of the nature of the photolabeled subunit (alpha or beta), suggesting that each of the two subunits, alpha and beta, is required for the activity of F1. (6) The covalently photolabeled F1 was able to form a complex with aurovertin, as does native F1. The ADP-induced fluorescence enhancement was more severely inhibited than the fluorescence quenching caused by ATP. The precentage of inactivation of F1 was virtually the same as the percentage of inhibition of the ATP-induced fluorescence quenching, suggestion that fluorescence quenching is related to the binding of ATP to the catalytic site of F1.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenylyl Imidodiphosphate/analogs & derivatives , Azides , Mitochondria, Heart/enzymology , Proton-Translocating ATPases/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Adenylyl Imidodiphosphate/pharmacology , Animals , Binding Sites , Cattle , Edetic Acid/pharmacology , Kinetics , Magnesium/pharmacology , Magnesium Chloride , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...