Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 60(12): 5209-5215, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28564542

ABSTRACT

Serine hydrolases are susceptible to potent reversible inhibition by boronic acids. Large collections of chemically diverse boronic acid fragments are commercially available because of their utility in coupling chemistry. We repurposed the approximately 650 boronic acid reagents in our collection as a directed fragment library targeting serine hydrolases and related enzymes. Highly efficient hits (LE > 0.6) often result. The utility of the approach is illustrated with the results against autotaxin, a phospholipase implicated in cardiovascular disease.


Subject(s)
Boronic Acids/chemistry , Phosphoric Diester Hydrolases/metabolism , Serine Proteinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Crystallography, X-Ray , Drug Evaluation, Preclinical/methods , Humans , Nitriles/chemistry , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemistry , Small Molecule Libraries/chemistry , Surface Plasmon Resonance
2.
Proc Natl Acad Sci U S A ; 111(1): 173-8, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24347635

ABSTRACT

Covalent inhibition is a reemerging paradigm in kinase drug design, but the roles of inhibitor binding affinity and chemical reactivity in overall potency are not well-understood. To characterize the underlying molecular processes at a microscopic level and determine the appropriate kinetic constants, specialized experimental design and advanced numerical integration of differential equations are developed. Previously uncharacterized investigational covalent drugs reported here are shown to be extremely effective epidermal growth factor receptor (EGFR) inhibitors (kinact/Ki in the range 10(5)-10(7) M(-1)s(-1)), despite their low specific reactivity (kinact ≤ 2.1 × 10(-3) s(-1)), which is compensated for by high binding affinities (Ki < 1 nM). For inhibitors relying on reactivity to achieve potency, noncovalent enzyme-inhibitor complex partitioning between inhibitor dissociation and bond formation is central. Interestingly, reversible binding affinity of EGFR covalent inhibitors is highly correlated with antitumor cell potency. Furthermore, cellular potency for a subset of covalent inhibitors can be accounted for solely through reversible interactions. One reversible interaction is between EGFR-Cys797 nucleophile and the inhibitor's reactive group, which may also contribute to drug resistance. Because covalent inhibitors target a cysteine residue, the effects of its oxidation on enzyme catalysis and inhibitor pharmacology are characterized. Oxidation of the EGFR cysteine nucleophile does not alter catalysis but has widely varied effects on inhibitor potency depending on the EGFR context (e.g., oncogenic mutations), type of oxidation (sulfinylation or glutathiolation), and inhibitor architecture. These methods, parameters, and insights provide a rational framework for assessing and designing effective covalent inhibitors.


Subject(s)
Drug Resistance , Enzyme Inhibitors/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Catalysis , Cell Line, Tumor , Chemistry, Pharmaceutical , Cysteine/chemistry , Drug Design , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , ErbB Receptors/chemistry , Humans , Inhibitory Concentration 50 , Mass Spectrometry , Oxygen/chemistry , Phosphorylation , Protein Binding , Protein Conformation , Quinazolines/chemistry , Signal Transduction
3.
Bioorg Chem ; 39(5-6): 192-210, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21872901

ABSTRACT

Protein kinases are fascinating biological catalysts with a rapidly expanding knowledge base, a growing appreciation in cell regulatory control, and an ascendant role in successful therapeutic intervention. To better understand protein kinases, the molecular underpinnings of phosphoryl group transfer, protein phosphorylation, and inhibitor interactions are examined. This analysis begins with a survey of phosphate group and phosphoprotein properties which provide context to the evolutionary selection of phosphorylation as a central mechanism for biological regulation of most cellular processes. Next, the kinetic and catalytic mechanisms of protein kinases are examined with respect to model aqueous systems to define the elements of catalysis. A brief structural biology overview further delves into the molecular basis of catalysis and regulation of catalytic activity. Concomitant with a prominent role in normal physiology, protein kinases have important roles in the disease state. To facilitate effective kinase drug discovery, classic and emerging approaches for characterizing kinase inhibitors are evaluated including biochemical assay design, inhibitor mechanism of action analysis, and proper kinetic treatment of irreversible inhibitors. As the resulting protein kinase inhibitors can modulate intended and unintended targets, profiling methods are discussed which can illuminate a more complete range of an inhibitor's biological activities to enable more meaningful cellular studies and more effective clinical studies. Taken as a whole, a wealth of protein kinase biochemistry knowledge is available, yet it is clear that a substantial extent of our understanding in this field remains to be discovered which should yield many new opportunities for therapeutic intervention.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Aspartic Acid/metabolism , Catalytic Domain , Drug Discovery , Kinetics , Phosphorylation
4.
Biochemistry ; 50(8): 1412-20, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21222488

ABSTRACT

Human thymidine phosphorylase (hTP) is responsible for thymidine (dT) homeostasis, promotes angiogenesis, and is involved in metabolic inactivation of antiproliferative agents that inhibit thymidylate synthase. Understanding its transition state structure is on the path to design transition state analogues. Arsenolysis of dT by hTP permits kinetic isotope effect (KIE) analysis of the reaction by forming thymine and the chemically unstable 2-deoxyribose 1-arsenate. The transition state for the arsenolytic reaction was characterized using multiple KIEs and computational analysis. Transition state analysis revealed a concerted bimolecular (A(N)D(N)) mechanism. A transition state constrained to match the intrinsic KIE values was found using density functional theory (B3LYP/6-31G*). An active site histidine is implicated as the catalytic base responsible for activation of the arsenate nucleophile and stabilization of the thymine leaving group during the isotopically sensitive step. At the transition state, the deoxyribose ring exhibits significant oxocarbenium ion character with bond breaking (r(C-N) = 2.45 Å) nearly complete and minimal bond making to the attacking nucleophile (r(C-O) = 2.95 Å). The transition state model predicts a deoxyribose conformation with a 2'-endo ring geometry. Transition state structure for the slow hydrolytic reaction of hTP involves a stepwise mechanism [Schwartz, P. A., Vetticatt, M. J., and Schramm, V. L. (2010) J. Am. Chem. Soc. 132, 13425-13433], in contrast to the concerted mechanism described here for arsenolysis.


Subject(s)
Arsenates/chemistry , Arsenates/metabolism , Thymidine Phosphorylase/metabolism , Thymidine/chemistry , Thymidine/metabolism , Biocatalysis , Catalytic Domain , Humans , Kinetics , Models, Molecular , Thymidine Phosphorylase/chemistry
5.
J Am Chem Soc ; 132(38): 13425-33, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20804144

ABSTRACT

Human thymidine phosphorylase (hTP) is responsible for thymidine (dT) homeostasis, and its action promotes angiogenesis. In the absence of phosphate, hTP catalyzes a slow hydrolytic depyrimidination of dT yielding thymine and 2-deoxyribose (dRib). Its transition state was characterized using multiple kinetic isotope effect (KIE) measurements. Isotopically enriched thymidines were synthesized enzymatically from glucose or (deoxy)ribose, and intrinsic KIEs were used to interpret the transition state structure. KIEs from [1'-(14)C]-, [1-(15)N]-, [1'-(3)H]-, [2'R-(3)H]-, [2'S-(3)H]-, [4'-(3)H]-, and [5'-(3)H]dTs provided values of 1.033 ± 0.002, 1.004 ± 0.002, 1.325 ± 0.003, 1.101 ± 0.004, 1.087 ± 0.005, 1.040 ± 0.003, and 1.033 ± 0.003, respectively. Transition state analysis revealed a stepwise mechanism with a 2-deoxyribocation formed early and a higher energetic barrier for nucleophilic attack of a water molecule on the high energy intermediate. An equilibrium exists between the deoxyribocation and reactants prior to the irreversible nucleophilic attack by water. The results establish activation of the thymine leaving group without requirement for phosphate. A transition state constrained to match the intrinsic KIEs was found using density functional theory. An active site histidine (His116) is implicated as the catalytic base for activation of the water nucleophile at the rate-limiting transition state. The distance between the water nucleophile and the anomeric carbon (r(C-O)) is predicted to be 2.3 A at the transition state. The transition state model predicts that deoxyribose adopts a mild 3'-endo conformation during nucleophilic capture. These results differ from the concerted bimolecular mechanism reported for the arsenolytic reaction (Birck, M. R.; Schramm, V. L. J. Am. Chem. Soc. 2004, 126, 2447-2453).


Subject(s)
Thymidine Phosphorylase/metabolism , Thymidine/metabolism , Humans , Hydrolysis , Models, Molecular
6.
Biochemistry ; 46(24): 7284-92, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17503776

ABSTRACT

The photolysis of adenosylcobalamin (coenzyme B12) results in homolytic cleavage of the Co-C5' bond, forming cob(II)alamin and the 5'-deoxyadenosyl radical. In the presence of molecular oxygen, it has been proposed that the primary reaction is interception of the 5'-deoxyadenosyl radical by O2 to form adenosine-5'-aldehyde as the product (Hogenkamp, H. P. C., Ladd, J. N., and Barker, H. A. (1962) J. Biol. Chem. 237, 1950-1952). 5'-Peroxyadenosine is here found to be the initial nucleoside product of this reaction and found to decompose to adenosine-5'-aldehyde. Evidence indicates that 5'-peroxyadenosine arises from the hydrolysis of 5'-peroxyadenosylcobalamin, with the formation of cob(III)alamin. 5'-Peroxyadenosine undergoes further decomposition to adenosine-5'-aldehyde as the major final product of aerobic photolysis as well as to adenosine and adenine as minor products. In a cobalamin-dependent process, 5'-peroxyadenosine becomes re-ligated to cob(III)alamin to form 5'-peroxyadenosylcobalamin, which quickly decomposes to adenosine-5'-aldehyde and cob(III)alamin. This is supported by spectrophotometric observations of both rapidly photolyzed adenosylcobalamin and the reaction of cob(III)alamin with excess 5'-peroxyadenosine. 5'-Peroxyadenosine also slowly undergoes cobalamin-independent decomposition to adenosine-5'-aldehyde and the minor products adenosine and adenine. The present study provides a detailed description of the products initially formed when aqueous, homolytically cleaved adenosylcobalamin reacts with molecular oxygen and provides a detailed description of the behavior of those products subsequent to photolysis.


Subject(s)
Cobamides/chemistry , Cobamides/radiation effects , Adenosine/analogs & derivatives , Adenosine/chemistry , Aerobiosis , In Vitro Techniques , Magnetic Resonance Spectroscopy , Models, Chemical , Molecular Structure , Photolysis , Spectrophotometry , Vitamin B 12/analogs & derivatives , Vitamin B 12/chemistry
7.
Protein Sci ; 16(6): 1157-64, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17525464

ABSTRACT

The reaction of adenosylcobalamin-dependent dioldehydrase with 1,2-propanediol gives rise to a radical intermediate observable by EPR spectroscopy. This reaction requires a monovalent cation such as potassium ion. The radical signal arises from the formation of a radical pair comprised of the Co(II) of cob(II)alamin and a substrate-related radical generated upon hydrogen abstraction by the 5'-deoxyadenosyl radical. The high-field asymmetric doublet arising from the organic radical has allowed investigation of its composition and environment through the use of EPR spectroscopic techniques. To characterize the protonation state of the oxygen substituents in the radical intermediate, X-band EPR spectroscopy was performed in the presence of D(2)O and compared to the spectrum in H(2)O. Results indicate that the unpaired electron of the steady-state radical couples to a proton on the C(1) hydroxyl group. Other spectroscopic experiments were performed, using either potassium or thallous ion as the activating monovalent cation, in an attempt to exploit the magnetic nature of the (205,203)Tl nucleus to identify any intimate interaction of the radical intermediate with the activating cation. The radical intermediate in complex with dioldehydrase, cob(II)alamin and one of the activating monovalent cations was observed using EPR, ENDOR, and ESEEM spectroscopy. The spectroscopic evidence did not implicate a direct coordination of the activating cation and the substrate derived radical intermediate.


Subject(s)
Cations, Monovalent/chemistry , Propanediol Dehydratase/chemistry , Solvents/chemistry , Binding Sites , Cations, Monovalent/metabolism , Cobamides/chemistry , Electron Spin Resonance Spectroscopy/methods , Models, Chemical , Molecular Structure , Potassium/chemistry , Potassium/metabolism , Propanediol Dehydratase/metabolism , Protons , Thallium/chemistry , Thallium/metabolism
8.
Biochemistry ; 46(24): 7293-301, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17516630

ABSTRACT

The complex of dioldehydrase with adenosylcobalamin (coenzyme B12) and potassium ion reacts with molecular oxygen in the absence of a substrate to oxidize coenzyme and inactivate the complex. In this article, high performance liquid chromatography and mass spectral analysis are used to identify the nucleoside products resulting from oxygen inactivation. The product profile indicates that oxygen inactivation proceeds by direct reaction of molecular oxygen with the 5'-deoxyadenosyl radical and cob(II)alamin. Formation of 5'-peroxyadenosine as the initial nucleoside product chemically correlates this reaction with aerobic, aqueous photoinduced homolytic cleavage of adenosylcobalamin (Schwartz, P. A., and Frey, P. A., (2007) Biochemistry, in press), indicating that both reactions proceed through similar chemical intermediates. The oxygen inactivation of the enzyme-coenzyme complex shows an absolute requirement for the same monocations required in catalysis by dioldehydrase. Measurements of the dissociation constants for adenosylcobalamin from potassium-free (Kd = 16 +/- 2 microM) or potassium-bound dioldehydrase (Kd = 0.8 +/- 0.2 microM) reveal that the effect of the monocation in stimulating oxygen sensitivity cannot be explained by an effect on the binding of coenzyme to the enzyme. Cross-linking experiments suggest that the full quaternary structure is assembled in the absence of potassium ion under the experimental conditions. The results indicate that dioldehydrase likely harvests the binding energy of the activating monocation to stimulate the homolytic cleavage of the Co-C5' bond in adenosylcobalamin.


Subject(s)
Cobamides/chemistry , Cobamides/metabolism , Potassium/metabolism , Propanediol Dehydratase/chemistry , Propanediol Dehydratase/metabolism , Carbon/chemistry , Cobalt/chemistry , Kinetics , Models, Molecular , Oxidation-Reduction , Oxygen/metabolism , Propanediol Dehydratase/antagonists & inhibitors , Spectrophotometry
SELECTION OF CITATIONS
SEARCH DETAIL
...