Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
ChemMedChem ; 14(1): 78-82, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30376607

ABSTRACT

Antibiotics with novel mechanisms of action are desperately needed to combat the increasing rates of multidrug-resistant infections. Bacterial pantothenate kinase (PanK) has emerged as a target of interest to cut off the biosynthesis of coenzyme A. Herein we report the results of an in vitro high-throughput screen of over 10 000 small molecules against Bacillus anthracis PanK, as well as a follow-up screen of hits against PanK isolated from Pseudomonas aeruginosa and Burkholderia cenocepacia. Nine hits are structurally categorized and analyzed to set the stage for future drug development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacillus anthracis/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Bacillus anthracis/enzymology , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
2.
J Biol Chem ; 291(46): 24188-24199, 2016 Nov 11.
Article in English | MEDLINE | ID: mdl-27679486

ABSTRACT

The enzyme cytochrome c oxidase (CcO) or complex IV (EC 1.9.3.1) is a large transmembrane protein complex that serves as the last enzyme in the respiratory electron transport chain of eukaryotic mitochondria. CcO promotes the switch from glycolytic to oxidative phosphorylation (OXPHOS) metabolism and has been associated with increased self-renewal characteristics in gliomas. Increased CcO activity in tumors has been associated with tumor progression after chemotherapy failure, and patients with primary glioblastoma multiforme and high tumor CcO activity have worse clinical outcomes than those with low tumor CcO activity. Therefore, CcO is an attractive target for cancer therapy. We report here the characterization of a CcO inhibitor (ADDA 5) that was identified using a high throughput screening paradigm. ADDA 5 demonstrated specificity for CcO, with no inhibition of other mitochondrial complexes or other relevant enzymes, and biochemical characterization showed that this compound is a non-competitive inhibitor of cytochrome c When tested in cellular assays, ADDA 5 dose-dependently inhibited the proliferation of chemosensitive and chemoresistant glioma cells but did not display toxicity against non-cancer cells. Furthermore, treatment with ADDA 5 led to significant inhibition of tumor growth in flank xenograft mouse models. Importantly, ADDA 5 inhibited CcO activity and blocked cell proliferation and neurosphere formation in cultures of glioma stem cells, the cells implicated in tumor recurrence and resistance to therapy in patients with glioblastoma. In summary, we have identified ADDA 5 as a lead CcO inhibitor for further optimization as a novel approach for the treatment of glioblastoma and related cancers.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Electron Transport Complex IV/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Glioma , Neoplasm Proteins/antagonists & inhibitors , Animals , Cell Line, Tumor , Cytochromes c/metabolism , Electron Transport Complex IV/metabolism , Glioma/drug therapy , Glioma/enzymology , Humans , Mice , Neoplasm Proteins/metabolism , Xenograft Model Antitumor Assays
3.
Oncotarget ; 7(20): 29492-506, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27121052

ABSTRACT

The binding of hepatocyte growth factor (HGF) to its receptor MET activates a signaling cascade that promotes cell survival, proliferation, cell scattering, migration and invasion of malignant cells. HGF is secreted by cancer cells or by tumor-associated fibroblasts as pro-HGF, an inactive precursor. A key step in the regulation of HGF/MET signaling is proteolytic processing of pro-HGF to its active form by one of the three serine proteases, matriptase, hepsin or HGF activator (HGFA).We developed SRI 31215, a small molecule that acts as a triplex inhibitor of matriptase, hepsin and HGFA and mimics the activity of HAI-1/2, endogenous inhibitors of HGF activation. We demonstrated that SRI 31215 inhibits fibroblast-induced MET activation, epithelial-mesenchymal transition and migration of cancer cells. SRI 31215 overcomes primary resistance to cetuximab and gefitinib in HGF-producing colon cancer cells and prevents fibroblast-mediated resistance to EGFR inhibitors. Thus, SRI 31215 blocks signaling between cancer cells and fibroblasts and inhibits the tumor-promoting activity of cancer-associated fibroblasts.Aberrant HGF/MET signaling supports cell survival, proliferation, angiogenesis, invasion and metastatic spread of cancer cells, establishing HGF and MET as valid therapeutic targets. Our data demonstrate that inhibitors of HGF activation, such as SRI 31215, merit investigation as potential therapeutics in tumors that are addicted to HGF/MET signaling. The findings reported here also indicate that inhibitors of HGF activation overcome primary and acquired resistance to anti-EGFR therapy, providing a rationale for concurrent inhibition of EGFR and HGF to prevent therapeutic resistance and to improve the outcome of cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamidines/pharmacology , Hepatocyte Growth Factor/antagonists & inhibitors , Protein Precursors/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrimidinones/pharmacology , Signal Transduction/drug effects , Cancer-Associated Fibroblasts/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition/drug effects , Humans , Protein Precursors/drug effects
4.
ACS Med Chem Lett ; 7(2): 177-81, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26985294

ABSTRACT

In this letter we report first nonpeptide inhibitors of hepatocyte growth factor (HGF) activation. These compounds inhibit the three proteases (matriptase, hepsin, and HGF activator) required for HGF maturation. We show that 6, 8a, 8b, and 8d block activation of fibroblast-derived pro-HGF, thus preventing fibroblast-induced scattering of DU145 prostate cancer cells. Compound 6 (SRI 31215) is very soluble (91 µM) and has excellent microsome stability (human t 1/2 = 162 min; mouse t 1/2 = 296 min). In mouse 6 has an in vivo t 1/2 = 5.8 h following IV administration. The high solubility of 6 and IV t 1/2 make this compound a suitable prototype "triplex inhibitor" for the study of the inhibition of HGF activation in vivo.

5.
Biochem J ; 473(8): 1027-35, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26846349

ABSTRACT

Historically, drugs used in the treatment of cancers also tend to cause damage to healthy cells while affecting cancer cells. Therefore, the identification of novel agents that act specifically against cancer cells remains a high priority in the search for new therapies. In contrast with normal cells, most cancer cells contain multiple centrosomes which are associated with genome instability and tumorigenesis. Cancer cells can avoid multipolar mitosis, which can cause cell death, by clustering the extra centrosomes into two spindle poles, thereby enabling bipolar division. Kinesin-like protein KIFC1 plays a critical role in centrosome clustering in cancer cells, but is not essential for normal cells. Therefore, targeting KIFC1 may provide novel insight into selective killing of cancer cells. In the present study, we identified a small-molecule KIFC1 inhibitor, SR31527, which inhibited microtubule (MT)-stimulated KIFC1 ATPase activity with an IC50 value of 6.6 µM. By using bio layer interferometry technology, we further demonstrated that SR31527 bound directly to KIFC1 with high affinity (Kd=25.4 nM). Our results from computational modelling and saturation-transfer difference (STD)-NMR experiments suggest that SR31527 bound to a novel allosteric site of KIFC1 that appears suitable for developing selective inhibitors of KIFC1. Importantly, SR31527 prevented bipolar clustering of extra centrosomes in triple negative breast cancer (TNBC) cells and significantly reduced TNBC cell colony formation and viability, but was less toxic to normal fibroblasts. Therefore, SR31527 provides a valuable tool for studying the biological function of KIFC1 and serves as a potential lead for the development of novel therapeutic agents for breast cancer treatment.


Subject(s)
Drug Discovery , Kinesins/antagonists & inhibitors , Kinesins/metabolism , Thiadiazoles/chemistry , Thiadiazoles/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Discovery/methods , Humans , Kinesins/chemistry , Protein Binding/physiology , Protein Structure, Secondary , Thiadiazoles/pharmacology
6.
J Biol Chem ; 289(47): 32937-51, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25228699

ABSTRACT

Pathogenic mutations in the LRRK2 gene can cause late-onset Parkinson disease. The most common mutation, G2019S, resides in the kinase domain and enhances activity. LRRK2 possesses the unique property of cis-autophosphorylation of its own GTPase domain. Because high-resolution structures of the human LRRK2 kinase domain are not available, we used novel high-throughput assays that measured both cis-autophosphorylation and trans-peptide phosphorylation to probe the ATP-binding pocket. We disclose hundreds of commercially available activity-selective LRRK2 kinase inhibitors. Some compounds inhibit cis-autophosphorylation more strongly than trans-peptide phosphorylation, and other compounds inhibit G2019S-LRRK2 more strongly than WT-LRRK2. Through exploitation of structure-activity relationships revealed through high-throughput analyses, we identified a useful probe inhibitor, SRI-29132 (11). SRI-29132 is exquisitely selective for LRRK2 kinase activity and is effective in attenuating proinflammatory responses in macrophages and rescuing neurite retraction phenotypes in neurons. Furthermore, the compound demonstrates excellent potency, is highly blood-brain barrier-permeant, but suffers from rapid first-pass metabolism. Despite the observed selectivity of SRI-29132, docking models highlighted critical interactions with residues conserved in many protein kinases, implying a unique structural configuration for the LRRK2 ATP-binding pocket. Although the human LRRK2 kinase domain is unstable and insoluble, we demonstrate that the LRRK2 homolog from ameba can be mutated to approximate some aspects of the human LRRK2 ATP-binding pocket. Our results provide a rich resource for LRRK2 small molecule inhibitor development. More broadly, our results provide a precedent for the functional interrogation of ATP-binding pockets when traditional approaches to ascertain structure prove difficult.


Subject(s)
Adenosine Triphosphate/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Biocatalysis/drug effects , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Hep G2 Cells , Humans , Kinetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutation , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Pyridazines/chemistry , Pyridazines/metabolism , Pyridazines/pharmacology , Sequence Homology, Amino Acid , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
8.
J Mol Biol ; 342(3): 953-70, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15342249

ABSTRACT

We report three crystal structures of the Mycobacterium tuberculosis cell division protein FtsZ, as the citrate, GDP, and GTPgammaS complexes, determined at 1.89, 2.60, and 2.08A resolution. MtbFtsZ crystallized as a tight, laterally oriented dimer distinct from the longitudinal polymer observed for alphabeta-tubulin. Mutational data on Escherichia coli FtsZ suggest that this dimer interface is important for proper protofilament and "Z-ring" assembly and function. An alpha-to-beta secondary structure conformational switch at the dimer interface is spatially analogous to, and has many of the hallmarks of, the Switch I conformational changes exhibited by G-proteins upon activation. The presence of a gamma-phosphate in the FtsZ active site modulates the conformation of the "tubulin" loop T3 (spatially analogous to the G-protein Switch II); T3 switching upon gamma-phosphate ligation is directly coupled to the alpha-to-beta switch by steric overlap. The dual conformational switches observed here for the first time in an FtsZ link GTP binding and hydrolysis to FtsZ (and tubulin) lateral assembly and Z-ring contraction, and they are suggestive of an underappreciated functional analogy between FtsZ, tubulin and G-proteins.


Subject(s)
Bacterial Proteins/chemistry , Cytoskeletal Proteins/chemistry , Mycobacterium tuberculosis/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Binding Sites , Crystallography, X-Ray , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA, Bacterial/genetics , Dimerization , GTP-Binding Proteins/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine Diphosphate/metabolism , Hydrogen Bonding , Models, Molecular , Mycobacterium tuberculosis/genetics , Protein Conformation , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
FEMS Microbiol Lett ; 232(1): 101-5, 2004 Mar 12.
Article in English | MEDLINE | ID: mdl-15019741

ABSTRACT

The gene for dihydrofolate reductase of Mycobacterium tuberculosis was amplified by polymerase chain reaction (PCR) from M. tuberculosis H37Rv strain genomic DNA. The protein was expressed in inclusion bodies in high yield in Escherichia coli under the control of the T7 promoter. Active enzyme was obtained by refolding from guanidine HCl and after a single chromatography step the sample was > 99% homogeneous with a specific activity of approximately 15.5 micromol min(-1) mg(-1). Mass spectrometry analysis confirmed the expected mass of 17.6 kDa. Gel filtration of the enzyme indicated that it was a monomer. Steady-state kinetic parameters were determined and the effect of pH and KCl on the enzyme examined. Methotrexate and trimethoprim inhibited the enzyme.


Subject(s)
Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Amino Acid Sequence , Chromatography, Liquid , Cloning, Molecular , Enzyme Inhibitors/pharmacology , Enzyme Stability , Escherichia coli/enzymology , Escherichia coli/genetics , Hydrogen-Ion Concentration , Methotrexate/pharmacology , Molecular Sequence Data , Molecular Weight , Potassium Chloride , Recombinant Proteins/biosynthesis , Sequence Alignment , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/isolation & purification , Trimethoprim/pharmacology
10.
J Antimicrob Chemother ; 50(1): 111-4, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12096015

ABSTRACT

Compounds originally designed as putative tubulin inhibitors were tested as antitubercular agents for inhibition of the Mycobacterium tuberculosis analogue of tubulin, FtsZ. Initial screening of 200 2-alkoxycarbonylpyridines found several that inhibited M. tuberculosis growth. Two compounds, SRI-3072 and SRI-7614, inhibited FtsZ polymerization and were equipotent against susceptible and single-drug-resistant strains of M. tuberculosis. In addition, SRI-3072 reduced the growth of M. tuberculosis in mouse bone marrow macrophages. Our results suggest that these types of compound might be developed into antitubercular drugs effective against the current multidrug-resistant strains of M. tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Azepines/pharmacology , Bacterial Proteins/drug effects , Carbamates/pharmacology , Cytoskeletal Proteins , Mycobacterium tuberculosis/drug effects , Pteridines/pharmacology , Tuberculosis, Multidrug-Resistant/drug therapy , Animals , Antitubercular Agents/therapeutic use , Antitubercular Agents/toxicity , Azepines/therapeutic use , Azepines/toxicity , Bacterial Proteins/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/microbiology , Carbamates/therapeutic use , Carbamates/toxicity , Chlorocebus aethiops , GTP Phosphohydrolases/metabolism , Inhibitory Concentration 50 , Macrophages/drug effects , Macrophages/microbiology , Mice , Microbial Sensitivity Tests , Mycobacterium tuberculosis/metabolism , Pteridines/therapeutic use , Pteridines/toxicity , Vero Cells
11.
J Mol Biol ; 320(3): 677-93, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-12096917

ABSTRACT

The crystal structures of two human dihydrofolate reductase (hDHFR) ternary complexes, each with bound NADPH cofactor and a lipophilic antifolate inhibitor, have been determined at atomic resolution. The potent inhibitors 6-([5-quinolylamino]methyl)-2,4-diamino-5-methylpyrido[2,3-d]pyrimidine (SRI-9439) and (Z)-6-(2-[2,5-dimethoxyphenyl]ethen-1-yl)-2,4-diamino-5-methylpyrido[2,3-d]pyrimidine (SRI-9662) were developed at Southern Research Institute against Toxoplasma gondii DHFR-thymidylate synthase. The 5-deazapteridine ring of each inhibitor adopts an unusual puckered conformation that enables the formation of identical contacts in the active site. Conversely, the quinoline and dimethoxybenzene moieties exhibit distinct binding characteristics that account for the differences in inhibitory activity. In both structures, a salt-bridge is formed between Arg70 in the active site and Glu44 from a symmetry-related molecule in the crystal lattice that mimics the binding of methotrexate to DHFR.


Subject(s)
Tetrahydrofolate Dehydrogenase/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray , Folic Acid Antagonists/chemistry , Humans , Hydrogen Bonding , In Vitro Techniques , Macromolecular Substances , Models, Molecular , Molecular Sequence Data , NADP/chemistry , Protein Conformation , Pyrimidines/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Toxoplasma/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...