Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
Add more filters

Publication year range
1.
Molecules ; 29(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125005

ABSTRACT

Polarization and charge-transfer interactions play an important role in ligand-receptor complexes containing metals, and only quantum mechanics methods can adequately describe their contribution to the binding energy. In this work, we selected a set of benzenesulfonamide ligands of human Carbonic Anhydrase II (hCA II)-an important druggable target containing a Zn2+ ion in the active site-as a case study to predict the binding free energy in metalloprotein-ligand complexes and designed specialized computational methods that combine the ab initio fragment molecular orbital (FMO) method and GRID approach. To reproduce the experimental binding free energy in these systems, we adopted a machine-learning approach, here named formula generator (FG), considering different FMO energy terms, the hydrophobic interaction energy (computed by GRID) and logP. The main advantage of the FG approach is that it can find nonlinear relations between the energy terms used to predict the binding free energy, explicitly showing their mathematical relation. This work showed the effectiveness of the FG approach, and therefore, it might represent an important tool for the development of new scoring functions. Indeed, our scoring function showed a high correlation with the experimental binding free energy (R2 = 0.76-0.95, RMSE = 0.34-0.18), revealing a nonlinear relation between energy terms and highlighting the relevant role played by hydrophobic contacts. These results, along with the FMO characterization of ligand-receptor interactions, represent important information to support the design of new and potent hCA II inhibitors.


Subject(s)
Carbonic Anhydrase II , Carbonic Anhydrase Inhibitors , Protein Binding , Ligands , Carbonic Anhydrase II/antagonists & inhibitors , Carbonic Anhydrase II/chemistry , Carbonic Anhydrase II/metabolism , Humans , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Thermodynamics , Hydrophobic and Hydrophilic Interactions , Sulfonamides/chemistry , Sulfonamides/pharmacology , Metalloproteins/chemistry , Metalloproteins/antagonists & inhibitors , Metalloproteins/metabolism , Models, Molecular , Machine Learning , Benzenesulfonamides , Binding Sites
2.
Molecules ; 27(4)2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35209064

ABSTRACT

Metalloproteins are a family of proteins characterized by metal ion binding, whereby the presence of these ions confers key catalytic and ligand-binding properties. Due to their ubiquity among biological systems, researchers have made immense efforts to predict the structural and functional roles of metalloproteins. Ultimately, having a comprehensive understanding of metalloproteins will lead to tangible applications, such as designing potent inhibitors in drug discovery. Recently, there has been an acceleration in the number of studies applying machine learning to predict metalloprotein properties, primarily driven by the advent of more sophisticated machine learning algorithms. This review covers how machine learning tools have consolidated and expanded our comprehension of various aspects of metalloproteins (structure, function, stability, ligand-binding interactions, and inhibitors). Future avenues of exploration are also discussed.


Subject(s)
Machine Learning , Metalloproteins/chemistry , Metalloproteins/metabolism , Amino Acid Sequence , Binding Sites , Drug Design , Metalloproteins/antagonists & inhibitors , Models, Molecular , Protein Binding , Protein Stability , Proteolysis , Structure-Activity Relationship
3.
Chem Rev ; 119(2): 1323-1455, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30192523

ABSTRACT

Metalloenzymes are central to a wide range of essential biological activities, including nucleic acid modification, protein degradation, and many others. The role of metalloenzymes in these processes also makes them central for the progression of many diseases and, as such, makes metalloenzymes attractive targets for therapeutic intervention. Increasing awareness of the role metalloenzymes play in disease and their importance as a class of targets has amplified interest in the development of new strategies to develop inhibitors and ultimately useful drugs. In this Review, we provide a broad overview of several drug discovery efforts focused on metalloenzymes and attempt to map out the current landscape of high-value metalloenzyme targets.


Subject(s)
Enzyme Inhibitors/therapeutic use , Metalloproteins/metabolism , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Catalytic Domain , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Matrix Metalloproteinases/chemistry , Matrix Metalloproteinases/metabolism , Metalloproteins/antagonists & inhibitors , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Transferases/antagonists & inhibitors , Transferases/metabolism , Virus Diseases/drug therapy
4.
Proc Natl Acad Sci U S A ; 114(10): 2717-2722, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28209778

ABSTRACT

Natural products harbor unique and complex structures that provide valuable antibiotic scaffolds. With an increase in antibiotic resistance, natural products once again hold promise for new antimicrobial therapies, especially those with unique scaffolds that have been overlooked due to a lack of understanding of how they function. Dithiolopyrrolones (DTPs) are an underexplored class of disulfide-containing natural products, which exhibit potent antimicrobial activities against multidrug-resistant pathogens. DTPs were thought to target RNA polymerase, but conflicting observations leave the mechanisms elusive. Using a chemical genomics screen in Escherichia coli, we uncover a mode of action for DTPs-the disruption of metal homeostasis. We show that holomycin, a prototypical DTP, is reductively activated, and reduced holomycin chelates zinc with high affinity. Examination of reduced holomycin against zinc-dependent metalloenzymes revealed that it inhibits E. coli class II fructose bisphosphate aldolase, but not RNA polymerase. Reduced holomycin also strongly inhibits metallo-ß-lactamases in vitro, major contributors to clinical carbapenem resistance, by removing active site zinc. These results indicate that holomycin is an intracellular metal-chelating antibiotic that inhibits a subset of metalloenzymes and that RNA polymerase is unlikely to be the primary target. Our work establishes a link between the chemical structures of DTPs and their antimicrobial action; the ene-dithiol group of DTPs enables high-affinity metal binding as a central mechanism to inhibit metabolic processes. Our study also validates the use of chemical genomics in characterizing modes of actions of antibiotics and emphasizes the potential of metal-chelating natural products in antimicrobial therapy.


Subject(s)
Escherichia coli/drug effects , Lactams/pharmacology , Pyrroles/chemistry , Toluene/analogs & derivatives , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Biological Products/chemistry , Biological Products/therapeutic use , Catalytic Domain/drug effects , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/genetics , Disulfides/chemistry , Disulfides/therapeutic use , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Genomics , Homeostasis/drug effects , Humans , Lactams/chemistry , Metalloproteins/antagonists & inhibitors , Metalloproteins/genetics , Metals/chemistry , Pyrroles/therapeutic use , Toluene/chemistry , Toluene/therapeutic use , Zinc/metabolism , beta-Lactamases/drug effects , beta-Lactamases/genetics
5.
Org Biomol Chem ; 17(30): 7114-7123, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31294731

ABSTRACT

This review describes the recent advances in the design of novel artificial metalloenzymes and their application in C-H activation reactions. The combination of enzymes and metal or organometallic complexes for the creation of new artificial metalloenzymes has represented a very exciting research line. In particular, the development of proteins with the ability to perform C-H functionalization presents a significant challenge. Here we discuss the development of these processes on natural metalloenzymes by using directed evolution, biotin-(strept)avidin technologies, photocatalytic hybrids or reconstitution of heme-protein technology.


Subject(s)
Metalloproteins/antagonists & inhibitors , Organometallic Compounds/pharmacology , Metalloproteins/chemical synthesis , Metalloproteins/chemistry , Models, Molecular , Molecular Structure , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry
6.
J Chem Inf Model ; 59(9): 3846-3859, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31460757

ABSTRACT

Extensive usage of molecular docking for computer-aided drug discovery resulted in development of numerous programs with versatile scoring and posing algorithms. Selection of the docking program among these vast number of options is central to the outcome of drug discovery. To this end, comparative assessment studies of docking offer valuable insights into the selection of the optimal tool. Despite the availability of various docking assessment studies, the performance difference of docking programs has not been well addressed on metalloproteins which comprise a substantial portion of the human proteome and have been increasingly targeted for treatment of a wide variety of diseases. This study reports comparative assessment of seven docking programs on a diverse metalloprotein set which was compiled for this study. The refined set of the PDBbind (2017) was screened to gather 710 complexes with metal ion(s) closely located to the ligands (<4 Å). The redundancy was eliminated by clustering and overall 213 complexes were compiled as the nonredundant metalloprotein subset of the PDBbind refined. The scoring, ranking, and posing powers of seven noncommercial docking programs, namely, AutoDock4, AutoDock4Zn, AutoDock Vina, Quick Vina 2, LeDock, PLANTS, and UCSF DOCK6, were comprehensively evaluated on this nonredundant set. Results indicated that PLANTS (80%) followed by LeDock (77%), QVina (76%), and Vina (73%) had the most accurate posing algorithms while AutoDock4 (48%) and DOCK6 (56%) were the least successful in posing. Contrary to their moderate-to-high level of posing success, none of the programs was successful in scoring or ranking of the binding affinities (r2 ≈ 0). Screening power was further evaluated by using active-decoy ligand sets for a large compilation of metalloprotein targets. PLANTS stood out among other programs to be able to enrich the active ligand for every target, underscoring its robustness for screening of metalloprotein inhibitors. This study provides useful information for drug discovery studies targeting metalloproteins.


Subject(s)
Databases, Protein , Metalloproteins/metabolism , Molecular Docking Simulation , Drug Evaluation, Preclinical , Ligands , Metalloproteins/antagonists & inhibitors , Metalloproteins/chemistry , Protein Binding , Protein Conformation
7.
J Biol Inorg Chem ; 23(7): 1129-1138, 2018 10.
Article in English | MEDLINE | ID: mdl-30003339

ABSTRACT

Hydroxypyridinethiones (HOPTOs) are strong ligands for metal ions and potentially useful pharmacophores for inhibiting metalloenzymes relevant to human disease. However, HOPTOs have been sparingly used in drug discovery efforts due, in part, to concerns that this scaffold will act as a promiscuous, non-selective metalloenzyme inhibitor, as well as possess poor pharmacokinetics (PK), which may undermine drug candidates containing this functional group. To advance HOPTOs as a useful pharmacophore for metalloenzyme inhibitors, a library of 22 HOPTO isostere compounds has been synthesized and investigated. This library demonstrates that it is possible to maintain the core metal-binding pharmacophore (MBP) while generating diversity in structure, electronics, and PK properties. This HOPTO library has been screened against a set of four different metalloenzymes, demonstrating that while the same metal-binding donor atoms are maintained, there is a wide range of activity between metalloenzyme targets. Overall, this work shows that HOPTO isosteres are useful MBPs and valuable scaffolds for metalloenzyme inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Metalloproteins/antagonists & inhibitors , Organometallic Compounds/pharmacology , Pyridines/pharmacology , Thiones/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Metalloproteins/metabolism , Models, Molecular , Molecular Structure , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Pyridines/chemistry , Thiones/chemistry
8.
Adv Protein Chem Struct Biol ; 141: 539-562, 2024.
Article in English | MEDLINE | ID: mdl-38960485

ABSTRACT

Several species during evolution suffered random mutations in response to various environmental factors, which resulted in the formation of venom in phylogenetically distant species. The composition of the venom of most species is poorly known. Snake venom is well characterized while most species have poorly known composition. In contrast, snake venoms are well characterized which proteins and peptides are the main active and most abundant constituents. 42 protein families have been identified, including metalloproteins known as metalloproteinases. These macromolecules are enzymes with zinc in their active site derived from the disintegrin A and metalloproteinase (ADAM) cellular family and are categorized into three classes (PI, PII and PIII) according to their domain organization. The snake venom metalloproteinases (SVMP) are cytotoxic, neurotoxic, myotoxic and/or hematotoxic with a crucial role in the defense and restraint of prey. In this scenario envenoming represents a danger to human health and has been considered a neglected disease worldwide, particularly in tropical and subtropical countries. Nevertheless, recently advances in "omics" technologies have demonstrated interesting biological activities of SVMPs such as antimicrobial, anticancer, against cardiovascular diseases and nervous system disorders. Metalloproteins have the therapeutic potential to be converted into drugs as other components of the venom have undergone this process (e.g., captopril, tirefiban and eptifibatide). So, this chapter is focused on the metalloproteins found in the secretions of venomous species, highlight some aspects such as structure, biological activity, pharmacological therapeutic potential and on.


Subject(s)
Metalloproteins , Snake Venoms , Animals , Humans , Snake Venoms/metabolism , Snake Venoms/chemistry , Snake Venoms/enzymology , Metalloproteins/metabolism , Metalloproteins/chemistry , Metalloproteins/antagonists & inhibitors
9.
Inorg Chem ; 52(21): 12207-15, 2013 Nov 04.
Article in English | MEDLINE | ID: mdl-23706138

ABSTRACT

An ever-increasing number of metalloproteins are being discovered that play essential roles in physiological processes. Inhibitors of these proteins have significant potential for the treatment of human disease, but clinical success of these compounds has been limited. Herein, zinc(II)-dependent metalloprotein inhibitors in clinical use are reviewed, and the potential for using novel metal-binding groups (MBGs) in the design of these inhibitors is discussed. By using human carbonic anhydrase II as a model system, the nuances of MBG-metal interactions in the context of a protein environment can be probed. Understanding how metal coordination influences inhibitor binding may help in the design of new therapeutics targeting metalloproteins.


Subject(s)
Carbonic Anhydrase II/chemistry , Carbonic Anhydrase II/metabolism , Carbonic Anhydrase Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/chemistry , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Binding Sites , Carbonic Anhydrase II/antagonists & inhibitors , Carbonic Anhydrase II/genetics , Catalytic Domain , Crystallography, X-Ray , Drug Discovery , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Ligands , Metalloproteins/antagonists & inhibitors , Metalloproteins/metabolism , Molecular Targeted Therapy , Mutation , Zinc/metabolism
10.
Biochemistry ; 51(29): 5791-803, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22741521

ABSTRACT

The enzyme N(1)-(5'-phosphoribosyl) adenosine-5'-monophosphate cyclohydrolase (PR-AMP cyclohydrolase) is a Zn(2+) metalloprotein encoded by the hisI gene. It catalyzes the third step of histidine biosynthesis, an uncommon ring-opening of a purine heterocycle for use in primary metabolism. A three-dimensional structure of the enzyme from Methanobacterium thermoautotrophicum has revealed that three conserved cysteine residues occur at the dimer interface and likely form the catalytic site. To investigate the functions of these cysteines in the enzyme from Methanococcus vannielii, a series of biochemical studies were pursued to test the basic hypothesis regarding their roles in catalysis. Inactivation of the enzyme activity by methyl methane thiosulfonate (MMTS) or 5,5'-dithiobis(2-nitrobenzoic acid) (DTNB) also compromised the Zn(2+) binding properties of the protein inducing loss of up to 90% of the metal. Overall reaction stoichiometry and the potassium cyanide (KCN) induced cleavage of the protein suggested that all three cysteines were modified in the process. The enzyme was protected from DTNB-induced inactivation by inclusion of the substrate N(1)-(5'-phosphoribosyl)adenosine 5'-monophosphate; (PR-AMP), while Mg(2+), a metal required for catalytic activity, enhanced the rate of inactivation. Site-directed mutations of the conserved C93, C109, C116 and the double mutant C109/C116 were prepared and analyzed for catalytic activity, Zn(2+) content, and reactivity with DTNB. Substitution of alanine for each of the conserved cysteines showed no measurable catalytic activity, and only the C116A was still capable of binding Zn(2+). Reactions of DTNB with the C109A/C116A double mutant showed that C93 is completely modified within 0.5 s. A model consistent with these data involves a DTNB-induced mixed disulfide linkage between C93 and C109 or C116, followed by ejection of the active site Zn(2+) and provides further evidence that the Zn(2+) coordination site involves the three conserved cysteine residues. The C93 reactivity is modulated by the presence of the Zn(2+) and Mg(2+) and substantiates the role of this residue as a metal ligand. In addition, Mg(2+) ligand binding site(s) indicated by the structural analysis were probed by site-directed mutagenesis of three key aspartate residues flanking the conserved C93 which were shown to have a functional impact on catalysis, cysteine activation, and metal (zinc) binding capacity. The unique amino acid sequence, the dynamic properties of the cysteine ligands involved in Zn(2+) coordination, and the requirement for a second metal (Mg(2+)) are discussed in the context of their roles in catalysis. The results are consistent with a Zn(2+)-mediated activation of H(2)O mechanism involving histidine as a general base that has features similar to but distinct from those of previously characterized purine and pyrimidine deaminases.


Subject(s)
Hydrolases/metabolism , Metalloproteins/metabolism , Methanococcus/enzymology , Zinc/metabolism , Amino Acid Sequence , Catalytic Domain , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Dithionitrobenzoic Acid/pharmacology , Hydrolases/antagonists & inhibitors , Hydrolases/chemistry , Hydrolases/genetics , Magnesium/metabolism , Metalloproteins/antagonists & inhibitors , Metalloproteins/chemistry , Metalloproteins/genetics , Methanococcus/chemistry , Methanococcus/genetics , Models, Molecular , Molecular Sequence Data , Point Mutation , Sequence Alignment
11.
J Am Chem Soc ; 134(17): 7553-7, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22512303

ABSTRACT

Formaldehyde-a rapid and reversible inhibitor of hydrogen evolution by [FeFe]-hydrogenases-binds with a strong potential dependence that is almost complementary to that of CO. Whereas exogenous CO binds tightly to the oxidized state known as H(ox) but very weakly to a state two electrons more reduced, formaldehyde interacts most strongly with the latter. Formaldehyde thus intercepts increasingly reduced states of the catalytic cycle, and density functional theory calculations support the proposal that it reacts with the H-cluster directly, most likely targeting an otherwise elusive and highly reactive Fe-hydrido (Fe-H) intermediate.


Subject(s)
Clostridium acetobutylicum/enzymology , Enzyme Inhibitors/pharmacology , Formaldehyde/pharmacology , Hydrogen/metabolism , Hydrogenase/antagonists & inhibitors , Metalloproteins/antagonists & inhibitors , Carbon Monoxide/metabolism , Clostridium acetobutylicum/chemistry , Hydrogenase/chemistry , Hydrogenase/metabolism , Iron/chemistry , Metalloproteins/chemistry , Metalloproteins/metabolism
12.
Analyst ; 137(23): 5487-90, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23061092

ABSTRACT

Herein, we report the application of synthesized fluorescent, water soluble polymers for post-translational subtyping and differentiation of breast cancer cells in vitro. The fluorescence emission spectra from these polymers were modulated differently in the presence of conditioned cell culture media from various breast cancer cells. These polymers differentiate at a post-translation level possibly due to their ability to interact with extracellular enzymes that are over-expressed in cancerous conditions.


Subject(s)
Breast Neoplasms/classification , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Culture Media, Conditioned , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Female , Fluorescent Dyes , HEK293 Cells , HeLa Cells , Humans , Metalloproteins/analysis , Metalloproteins/antagonists & inhibitors , Polymers , Spectrometry, Fluorescence
13.
J Med Chem ; 64(24): 17706-17727, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34875836

ABSTRACT

Metalloenzymes have critical roles in a wide range of biological processes and are directly involved in many human diseases; hence, they are considered as important targets for therapeutic intervention. The specific characteristics of metal ion(s)-containing active sites make exploitation of metal-binding pharmacophores (MBPs) critical to inhibitor development targeting metalloenzymes. This Perspective focuses on boron-containing MBPs, which display unique binding modes with metalloenzyme active sites, particularly via mimicking native substrates or tetrahedral transition states. The design concepts regarding boron-containing MBPs are highlighted through the case analyses on five distinct classes of clinically relevant nucleophilic metalloenzymes from medicinal chemistry perspectives. The challenges (e.g., selectivity) faced by some boron-containing MBPs and possible strategies (e.g., bioisosteres) for metalloenzyme inhibitor transformation are also discussed.


Subject(s)
Boron/chemistry , Enzyme Inhibitors/pharmacology , Metalloproteins/antagonists & inhibitors , Metals/chemistry , Enzyme Inhibitors/chemistry , Molecular Structure
14.
Metallomics ; 13(3)2021 02 26.
Article in English | MEDLINE | ID: mdl-33638642

ABSTRACT

Inhibitors of indoleamine 2,3-dioxygenase 1 (IDO1) have received wide attention for their roles in cancer immunotherapy. It highlights the important role of metalloenzymes in performing human physiological functions. Herein, the recombinant human IDO1 was expressed and purified successfully, and the protein molecule was characterized by SDS-PAGE, MALDI-TOF mass spectrometry, and metalloenzymology. A series of niacin derivatives were investigated with regard to their inhibition on metalloenzyme IDO1, and the resulting potential anti-cancer activities in cell lines. Among the niacin derivatives, 4,4,4-trifluoro-1-(pyridin-3-yl)-butane-1,3-dione (compound 9) was found to be the most effective inhibitor to IDO1 in HepG-2 cells, with an EC50 of 11 µM with low cytotoxicity. The IC50 value of compound 9 with trifluoroethyl group in enzymatic inhibition was shown to be ∼5 times more potent than a positive control 4-phenylimidazole. The interaction between compound 9 and IDO1 was verified by isothermal titration calorimetry and molecular docking study. The most favorable molecular docking results revealed that functional groups of compound 9 contributed to the binding of 9 to IDO1 through IDO1-heme coordination, H-bond interactions and hydrophobic contacts. Our finding provides a strategy for the development of new inhibitor candidates for the therapeutic inhibition of IDO1.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Design , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Molecular Docking Simulation , Niacin/chemistry , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival , Hep G2 Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Metalloproteins/antagonists & inhibitors , Metalloproteins/metabolism , Structure-Activity Relationship
15.
J Med Chem ; 63(20): 12116-12127, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32940035

ABSTRACT

Immobilized metal-ion affinity chromatography (IMAC) used to purify recombinant proteins features a resin-bound 1:1 Ni(II)-iminodiacetic acid (IDA) complex. This hemi-saturated Ni(II)-IDA system containing exchangeable sites at the metal ion is re-cast as a surrogate of a coordinatively-unsaturated metalloenzyme active site, with utility for selecting compounds with metal-binding groups from mixtures as potential metalloenzyme inhibitors. Exchanging Ni(II) for other metal ions could broaden the scope of metalloenzyme target. This work examined the performance of Cu(II)-, Fe(III)-, Ga(III)-, Ni(II)-, or Zn(II)-IMAC resins to reversibly bind experimental or clinical metalloenzyme inhibitors of Zn(II)-ACE1, Zn(II)-HDAC, Fe(II)/(III)-5-LO or Cu(II)-tyrosinase from a curated mixture (1-17). Each IMAC system gave a distinct selection profile. The Zn(II)-IMAC system selectively bound the thiol-containing Zn(II)-ACE1 inhibitors captopril and omapatrilat, and the Fe(III)-IMAC system selectively bound the Fe(II)/(III)-5-LO inhibitor licofelone, demonstrating a remarkable IMAC-metalloenzyme metal ion match. IMAC provides a simple, water-compatible platform, which could accelerate metalloenzyme inhibitor discovery.


Subject(s)
Coordination Complexes/pharmacology , Drug Discovery , Imino Acids/pharmacology , Metalloproteins/antagonists & inhibitors , Metals, Heavy/pharmacology , Chromatography, Affinity , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Imino Acids/chemistry , Metalloproteins/metabolism , Metals, Heavy/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
16.
Science ; 221(4612): 769-70, 1983 Aug 19.
Article in English | MEDLINE | ID: mdl-6308761

ABSTRACT

Vegetative cells of Clostridium botulinum were shown to contain iron-sulfur proteins that react with added nitrite to form iron-nitric oxide complexes, with resultant destruction of the iron-sulfur cluster. Inactivation of iron-sulfur enzymes (especially ferredoxin) by binding of nitric oxide would almost certainly inhibit growth, and thus is probably the mechanism of botulinal inhibition by nitrite in foods.


Subject(s)
Clostridium botulinum/drug effects , Iron-Sulfur Proteins/antagonists & inhibitors , Metalloproteins/antagonists & inhibitors , Nitrites/pharmacology , Electron Spin Resonance Spectroscopy , Iron , Nitric Oxide
17.
Eur J Med Chem ; 165: 172-197, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30684796

ABSTRACT

Numerous metal-containing enzymes (metalloenzymes) have been considered as drug targets related to diseases such as cancers, diabetes, anemia, AIDS, malaria, bacterial infection, fibrosis, and neurodegenerative diseases. Inhibitors of the metalloenzymes have been developed independently, most of which are mimics of substrates of the corresponding enzymes. However, little attention has been paid to the interactions between inhibitors and active site metal ions. This review is focused on different metal binding fragments and their chelating properties in the metal-containing active binding pockets of metalloenzymes. We have enumerated over one hundred of inhibitors targeting various metalloenzymes and identified over ten kinds of fragments with different binding patterns. Furthermore, we have investigated the inhibitors that are undergoing clinical evaluation in order to help looking for more potential scaffolds bearing metal binding fragments. This review will provide deep insights for the rational design of novel inhibitors targeting the metal-containing binding sites of specific proteins.


Subject(s)
Chelating Agents/therapeutic use , Chemistry, Pharmaceutical/methods , Metalloproteins/antagonists & inhibitors , Animals , Catalytic Domain , Drug Design , Humans
18.
Curr Med Chem ; 26(15): 2748-2785, 2019.
Article in English | MEDLINE | ID: mdl-29984644

ABSTRACT

Epigenetics controls the expression of genes and is responsible for cellular phenotypes. The fundamental basis of these mechanisms involves in part the post-translational modifications (PTMs) of DNA and proteins, in particular, the nuclear histones. DNA can be methylated or demethylated on cytosine. Histones are marked by several modifications including acetylation and/or methylation, and of particular importance are the covalent modifications of lysine. There exists a balance between addition and removal of these PTMs, leading to three groups of enzymes involved in these processes: the writers adding marks, the erasers removing them, and the readers able to detect these marks and participating in the recruitment of transcription factors. The stimulation or the repression in the expression of genes is thus the result of a subtle equilibrium between all the possibilities coming from the combinations of these PTMs. Indeed, these mechanisms can be deregulated and then participate in the appearance, development and maintenance of various human diseases, including cancers, neurological and metabolic disorders. Some of the key players in epigenetics are metalloenzymes, belonging mostly to the group of erasers: the zinc-dependent histone deacetylases (HDACs), the iron-dependent lysine demethylases of the Jumonji family (JMJ or KDM) and for DNA the iron-dependent ten-eleven-translocation enzymes (TET) responsible for the oxidation of methylcytosine prior to the demethylation of DNA. This review presents these metalloenzymes, their importance in human disease and their inhibitors.


Subject(s)
Histone Deacetylase Inhibitors/therapeutic use , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Metalloproteins/antagonists & inhibitors , Animals , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylases/physiology , Humans , Jumonji Domain-Containing Histone Demethylases/physiology , Metalloproteins/physiology
19.
Curr Med Chem ; 26(15): 2690-2714, 2019.
Article in English | MEDLINE | ID: mdl-29611472

ABSTRACT

The utilization of bacterial metalloenzymes, especially ones not having mammalian (human) counterparts, has drawn attention to develop novel antibacterial agents to overcome drug resistance and especially multidrug resistance. In this review, we focus on the recent achievements on the development of inhibitors of bacterial enzymes peptide deformylase (PDF), metallo-ß-lactamase (MBL), methionine aminopeptidase (MetAP) and UDP-3-O-acyl- N-acetylglucosamine deacetylase (LpxC). The state of the art of the design and investigation of inhibitors of bacterial metalloenzymes is presented, and challenges are outlined and discussed.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/enzymology , Bacterial Proteins/antagonists & inhibitors , Metalloproteins/antagonists & inhibitors , beta-Lactamase Inhibitors/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Catalytic Domain , Cell Line, Tumor , Drug Design , Humans , Metalloproteins/chemistry , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , beta-Lactamase Inhibitors/chemistry
20.
Oncogene ; 26(19): 2707-16, 2007 Apr 26.
Article in English | MEDLINE | ID: mdl-17057733

ABSTRACT

Ribosomal proteins were recently shown to regulate p53 activity by abrogating Mdm2-induced p53 degradation (L23, L11, L5) or by enhancing p53 translation (L26). Here, we report that a novel ribosomal protein, RPS27L (S27-like protein), is a direct p53 target. RPS27L, but not its family member RPS27, was identified as a p53 inducible gene in a genome-wide chip-profiling study. Further characterization revealed a p53-dependent induction of RPS27L in multiple cancer cell models. Indeed, a consensus p53-binding site was identified in the first intron of the RPS27L gene and a direct binding of p53 to this site was demonstrated both in vitro and in vivo. Characterization of a luciferase reporter driven by the RPS27L intron fragment revealed a p53-binding site-dependent transaction by wild-type p53, but not by several transactivating-deficient p53 mutants. This transactivation was enhanced by etoposide, a DNA damaging agent that activates p53 and was completely blocked by a dominant-negative p53 mutant. Functionally, overexpression of RPS27L within the physiological inducible levels promoted, whereas siRNA silencing of RPS27L inhibited, apoptosis induced by etoposide. This is the first report, to our knowledge, that p53 directly induces the expression of a ribosomal protein, RPS27L, which in turn promotes apoptosis.


Subject(s)
Apoptosis , Metalloproteins/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Binding Sites , Chromatin Immunoprecipitation , DNA Damage , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic , Gene Silencing , HCT116 Cells/metabolism , Humans , Introns/genetics , Metalloproteins/antagonists & inhibitors , Metalloproteins/chemistry , Metalloproteins/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Messenger , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Transfection , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL