Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
Add more filters










Publication year range
1.
Appl Microbiol Biotechnol ; 106(4): 1543-1556, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35147743

ABSTRACT

Streptomyces roseochromogenes NRRL 3504 is best known as a producer of clorobiocin, a DNA replication inhibitor from the aminocoumarin family of antibiotics. This natural product currently draws attention as a promising adjuvant for co-application with other antibiotics against Gram-negative multidrug-resistant pathogens. Herein, we expand the genetic toolkit for NRRL 3504 by showing that a set of integrative and replicative vectors, not tested previously for this strain, could be conjugally transferred at high frequency from Escherichia coli to NRRL 3504. Using this approach, we leverage a cumate-inducible expression of cluster-situated regulatory gene novG to increase clorobiocin titers by 30-fold (up to approximately 200 mg/L). To our best knowledge, this is the highest level of clorobiocin production reported so far. Our findings set a working ground for further improvement of clorobiocin production as well as for the application of genetic methods to illuminate the cryptic secondary metabolome of NRRL 3504. Key Points • Efficient system for conjugative transfer of plasmids into NRRL 3504 was developed. • Expression of regulatory genes in NRRL 3504 led to increase in clorobiocin titer. • Secondary metabolome of NRRL 3504 becomes an accessible target for genetic manipulations using the expanded vector set and improved intergeneric conjugation protocol.


Subject(s)
Novobiocin , Streptomyces , Anti-Bacterial Agents/pharmacology , Novobiocin/analogs & derivatives , Streptomyces/metabolism
2.
Int J Mol Sci ; 22(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34769152

ABSTRACT

Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is a neurodegenerative disease caused by mutations in the SACS gene, encoding the 520 kDa modular protein sacsin, which comprises multiple functional sequence domains that suggest a role either as a scaffold in protein folding or in proteostasis. Cells from patients with ARSACS display a distinct phenotype including altered organisation of the intermediate filament cytoskeleton and a hyperfused mitochondrial network where mitochondrial respiration is compromised. Here, we used vimentin bundling as a biomarker of sacsin function to test the therapeutic potential of Hsp90 inhibition with the C-terminal-domain-targeted compound KU-32, which has demonstrated mitochondrial activity. This study shows that ARSACS patient cells have significantly increased vimentin bundling compared to control, and this was also present in ARSACS carriers despite them being asymptomatic. We found that KU-32 treatment significantly reduced vimentin bundling in carrier and patient cells. We also found that cells from patients with ARSACS were unable to maintain mitochondrial membrane potential upon challenge with mitotoxins, and that the electron transport chain function was restored upon KU-32 treatment. Our preliminary findings presented here suggest that targeting the heat-shock response by Hsp90 inhibition alleviates vimentin bundling and may represent a promising area for the development of therapeutics for ARSACS.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Muscle Spasticity/drug therapy , Novobiocin/analogs & derivatives , Spinocerebellar Ataxias/congenital , Cell Line , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Muscle Spasticity/metabolism , Novobiocin/pharmacology , Spinocerebellar Ataxias/drug therapy , Spinocerebellar Ataxias/metabolism , Vimentin/metabolism
3.
Int J Antimicrob Agents ; 56(1): 106002, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32361027

ABSTRACT

Burkholderia pseudomallei causes melioidosis, a potentially lethal disease that can establish both chronic and acute infections in humans. It is inherently recalcitrant to many antibiotics, there is a paucity of effective treatment options and there is no vaccine. In the present study, the efficacies of selected aminocoumarin compounds, DNA gyrase inhibitors that were discovered in the 1950s but are not in clinical use for the treatment of melioidosis were investigated. Clorobiocin and coumermycin were shown to be particularly effective in treating B. pseudomallei infection in vivo. A novel formulation with dl-tryptophan or l-tyrosine was shown to further enhance aminocoumarin potency in vivo. It was demonstrated that coumermycin has superior pharmacokinetic properties compared with novobiocin, and the coumermycin in l-tyrosine formulation can be used as an effective treatment for acute respiratory melioidosis in a murine model. Repurposing of existing approved antibiotics offers new resources in a challenging era of drug development and antimicrobial resistance.


Subject(s)
Aminocoumarins/therapeutic use , Burkholderia pseudomallei/drug effects , Melioidosis/drug therapy , Novobiocin/analogs & derivatives , Tryptophan/therapeutic use , Aminocoumarins/pharmacokinetics , Animals , Burkholderia pseudomallei/genetics , Disease Models, Animal , Drug Resistance, Multiple, Bacterial/genetics , Drug Therapy, Combination , Female , Mice , Mice, Inbred BALB C , Moths/microbiology , Novobiocin/pharmacokinetics , Novobiocin/therapeutic use
4.
Sci Signal ; 13(630)2020 05 05.
Article in English | MEDLINE | ID: mdl-32371496

ABSTRACT

Morphine and other opioids are commonly used to treat pain despite their numerous adverse side effects. Modulating µ-opioid receptor (MOR) signaling is one way to potentially improve opioid therapy. In mice, the chaperone protein Hsp90 mediates MOR signaling within the brain. Here, we found that inhibiting Hsp90 specifically in the spinal cord enhanced the antinociceptive effects of morphine in mice. Intrathecal, but not systemic, administration of the Hsp90 inhibitors 17-AAG or KU-32 amplified the effects of morphine in suppressing sensitivity to both thermal and mechanical stimuli in mice. Hsp90 inhibition enabled opioid-induced phosphorylation of the kinase ERK and increased abundance of the kinase RSK in the dorsal horns of the spinal cord, which are heavily populated with primary afferent sensory neurons. The additive effects of Hsp90 inhibition were abolished upon intrathecal inhibition of ERK, RSK, or protein synthesis. This mechanism downstream of MOR, localized to the spinal cord and repressed by Hsp90, may potentially be used to enhance the efficacy and presumably decrease the side effects of opioid therapy.


Subject(s)
Analgesics/pharmacology , Benzoquinones/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , MAP Kinase Signaling System/drug effects , Morphine/pharmacology , Novobiocin/analogs & derivatives , Receptors, Opioid, mu/metabolism , Spine/metabolism , Animals , Benzoquinones/agonists , Female , HSP90 Heat-Shock Proteins/metabolism , Lactams, Macrocyclic/agonists , Male , Mice , Morphine/agonists , Novobiocin/agonists , Novobiocin/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Spine/pathology
5.
J Biol Chem ; 294(16): 6450-6467, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30792306

ABSTRACT

Heat shock protein 90 (Hsp90) is a eukaryotic chaperone responsible for the folding and functional activation of numerous client proteins, many of which are oncoproteins. Thus, Hsp90 inhibition has been intensely pursued, resulting in the development of many potential Hsp90 inhibitors, not all of which are well-characterized. Hsp90 inhibitors not only abrogate its chaperone functions, but also could help us gain insight into the structure-function relationship of this chaperone. Here, using biochemical and cell-based assays along with isothermal titration calorimetry, we investigate KU-32, a derivative of the Hsp90 inhibitor novobiocin (NB), for its ability to modulate Hsp90 chaperone function. Although NB and KU-32 differ only slightly in structure, we found that upon binding, they induce completely opposite conformational changes in Hsp90. We observed that NB and KU-32 both bind to the C-terminal domain of Hsp90, but surprisingly, KU-32 stimulated the chaperone functions of Hsp90 via allosteric modulation of its N-terminal domain, responsible for the chaperone's ATPase activity. In vitro and in silico studies indicated that upon KU-32 binding, Hsp90 undergoes global structural changes leading to the formation of a "partially closed" intermediate that selectively binds ATP and increases ATPase activity. We also report that KU-32 promotes HeLa cell survival and enhances the refolding of an Hsp90 substrate inside the cell. This discovery explains the effectiveness of KU-32 analogs in the management of neuropathies and may facilitate the design of molecules that promote cell survival by enhancing Hsp90 chaperone function and reducing the load of misfolded proteins in cells.


Subject(s)
Enzyme Inhibitors , HSP90 Heat-Shock Proteins , Novobiocin/analogs & derivatives , Protein Folding/drug effects , Allosteric Regulation/drug effects , Cell Survival/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Novobiocin/chemistry , Novobiocin/pharmacology , Protein Binding , Protein Domains
6.
Biophys J ; 116(4): 648-658, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30691677

ABSTRACT

The overexpression of multidrug efflux pumps is an important mechanism of clinical resistance in Gram-negative bacteria. Recently, four small molecules were discovered that inhibit efflux in Escherichia coli and interact with the AcrAB-TolC efflux pump component AcrA. However, the binding site(s) for these molecules was not determined. Here, we combine ensemble docking and molecular dynamics simulations with tryptophan fluorescence spectroscopy, site-directed mutagenesis, and antibiotic susceptibility assays to probe binding sites and effects of binding of these molecules. We conclude that clorobiocin and SLU-258 likely bind at a site located between the lipoyl and ß-barrel domains of AcrA.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carrier Proteins/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Lipoproteins/antagonists & inhibitors , Lipoproteins/metabolism , Membrane Transport Proteins/metabolism , Anti-Bacterial Agents/metabolism , Binding Sites , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Lipoproteins/chemistry , Lipoproteins/genetics , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation , Novobiocin/analogs & derivatives , Novobiocin/metabolism , Novobiocin/pharmacology , Protein Domains
7.
Chem Biol Drug Des ; 93(4): 647-652, 2019 04.
Article in English | MEDLINE | ID: mdl-30570806

ABSTRACT

To maintain the lipid asymmetry of the cell envelope in Gram-negative bacteria, the MlaC protein serves as a lipid transfer factor and delivers phospholipids from the outer to the inner membrane. A strategy of antibiotic discovery is to design a proper compound that can tightly bind to the MlaC protein and inhibit the MlaC function. In this study, we performed virtual screening on multiple MlaC structures obtained from molecular dynamics simulations to identify potential MlaC binders. Our results suggested that clorobiocin is a compound that could bind to the MlaC protein. Through the comparison of the bound geometry between clorobiocin and novobiocin, we pointed out that the methyl-pyrrole group of the noviose sugar in clorobiocin forms hydrophobic interactions with amino acids in the phospholipid binding pocket, which allows the compound to bind deep in the active site. This also explains why clorobiocin shows a tighter binding affinity than novobiocin. Our study highlights a practical path of antibiotic development against Gram-negative bacteria.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/metabolism , Gram-Negative Bacteria/metabolism , Membrane Transport Proteins/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Binding Sites , Gram-Negative Bacteria/drug effects , Membrane Transport Proteins/metabolism , Molecular Docking Simulation , Novobiocin/analogs & derivatives , Novobiocin/chemistry , Novobiocin/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Protein Structure, Tertiary
8.
Behav Brain Res ; 329: 186-190, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28359881

ABSTRACT

Chemotherapy induced cognitive impairment (i.e. chemobrain) involves acute and long-term deficits in memory, executive function, and processing speed. Animal studies investigating these cognitive deficits have had mixed results, potentially due to variability in the complexity of behavioral tasks across experiments. Further, common chemotherapy treatments such as 5-fluorouracil (5-FU) break down myelin integrity corresponding to hippocampal neurodegenerative deficits and mitochondrial dysfunction. There is little evidence, however, of pharmacological treatments that may target mitochondrial dysfunction. Using a differential reinforcement of low rates (DRL) task combining spatial and temporal components, the current study evaluated the preventative effects of the pharmacological agent KU32 on the behavior of rats treated with 5-FU (5-FU+Saline vs. 5FU+KU32). DRL performance was analyzed the day after the first set of injections (D1), the day after the second set of injections (D7) and the last day of the experiment (D14). The 5FU+KU32 group earned significantly more reinforcers on the DRL task at D7 and D14 than the 5FU+Saline group. Further, the 5FU+KU32 group showed significantly better temporal discrimination. The 5FU+KU32 showed within-group improvement in temporal discrimination from D7 to D14. No significant differences were observed in spatial discrimination, however, those in the 5FU+Saline group responded more frequently on T3 compared to the 5FU+KU32 group, highlighting temporal discrimination differences between groups. The current data suggest that KU32 shows promise in the prevention of chemotherapy induced impairments in temporal discrimination.


Subject(s)
Cognition Disorders/chemically induced , Cognition Disorders/prevention & control , Fluorouracil/toxicity , Immunosuppressive Agents/toxicity , Neuroprotective Agents/therapeutic use , Novobiocin/analogs & derivatives , Analysis of Variance , Animals , Disease Models, Animal , Novobiocin/therapeutic use , Rats , Rats, Wistar , Time Factors
9.
PLoS One ; 12(3): e0174665, 2017.
Article in English | MEDLINE | ID: mdl-28355308

ABSTRACT

Acceptor substrate specificity of Streptomyces roseochromogenes prenyltransferase SrCloQ was investigated using different non-genuine phenolic compounds. RP-UHPLC-UV-MSn was used for the tentative annotation and quantification of the prenylated products. Flavonoids, isoflavonoids and stilbenoids with different types of substitution were prenylated by SrCloQ, although with less efficiency than the genuine substrate 4-hydroxyphenylpyruvate. The isoflavan equol, followed by the flavone 7,4'-dihydroxyflavone, were the best non-genuine acceptor substrates. B-ring C-prenylation was in general preferred over A-ring C-prenylation (ratio 5:1). Docking studies of non-genuine acceptor substrates with the B-ring oriented towards the donor substrate dimethylallyl pyrophosphate, showed that the carbonyl group of the C-ring was able to make stabilizing interactions with the residue Arg160, which might determine the preference observed for B-ring prenylation. No reaction products were formed when the acceptor substrate had no phenolic hydroxyl groups. This preference can be explained by the essential hydrogen bond needed between a phenolic hydroxyl group and the residue Glu281. Acceptor substrates with an additional hydroxyl group at the C3' position (B-ring), were mainly O3'-prenylated (> 80% of the reaction products). This can be explained by the proximity of the C3' hydroxyl group to the donor substrate at the catalytic site. Flavones were preferred over isoflavones by SrCloQ. Docking studies suggested that the orientation of the B-ring and of the phenolic hydroxyl group at position C7 (A-ring) of flavones towards the residue Tyr233 plays an important role in this observed preference. Finally, the insights obtained on acceptor substrate specificity and regioselectivity for SrCloQ were extended to other prenyltransferases from the CloQ/NhpB family.


Subject(s)
Bacterial Proteins/metabolism , Dimethylallyltranstransferase/metabolism , Flavonoids/metabolism , Isoflavones/metabolism , Streptomyces/enzymology , Bacterial Proteins/chemistry , Catalytic Domain , Dimethylallyltranstransferase/chemistry , Equol/chemistry , Equol/metabolism , Flavonoids/chemistry , Hydrogen Bonding , Isoflavones/chemistry , Kinetics , Molecular Docking Simulation , Molecular Structure , Novobiocin/analogs & derivatives , Novobiocin/biosynthesis , Novobiocin/chemistry , Phenols/chemistry , Phenols/metabolism , Phenylpyruvic Acids/chemistry , Phenylpyruvic Acids/metabolism , Prenylation , Protein Binding , Protein Structure, Tertiary , Stilbenes/chemistry , Stilbenes/metabolism , Streptomyces/metabolism , Substrate Specificity
10.
Chemistry ; 22(20): 6921-31, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27037933

ABSTRACT

Development of heat shock protein 90 (Hsp90) C-terminal inhibitors has emerged as an exciting strategy for the treatment of cancer. Previous efforts have focused on modifications to the natural products novobiocin and coumermycin. Moreover, variations in both the sugar and amide moieties have been extensively studied, whereas replacements for the coumarin core have received less attention. Herein, 24 cores were synthesized with varying distances and angles between the sugar and amide moieties. Compounds that exhibited good anti-proliferative activity against multiple cancer cell lines and Hsp90 inhibitory activity, were those that placed the sugar and amide moieties between 7.7 and 12.1 Šapart along with angles of 180°.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , Novobiocin/analogs & derivatives , Aminocoumarins/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Coumarins/chemistry , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Novobiocin/chemistry , Structure-Activity Relationship
11.
J Med Chem ; 59(3): 925-33, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26745854

ABSTRACT

Heat shock protein 90 (Hsp90) inhibition by modulation of its N- or C-terminal binding site has become an attractive strategy for the development of anticancer chemotherapeutics. The first Hsp90 C-terminus inhibitor, novobiocin, manifested a relatively high IC50 value of ∼700 µM. Therefore, investigation of the novobiocin scaffold has led to analogues with improved antiproliferative activity (nanomolar concentrations) against several cancer cell lines. During these studies, novobiocin analogues that do not inhibit Hsp90 were identified; however, these analogues demonstrated potent antiproliferative activity. Compound 2, a novobiocin analogue, was identified as a MAPK pathway signaling disruptor that lacked Hsp90 inhibitory activity. In addition, structural modifications of compound 2 were identified that segregated Hsp90 inhibition from MAPK signaling disruption. These studies indicate that compound 2 represents a novel scaffold for disruption of MAPK pathway signaling and may serve as a useful structure for the generation of new anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Novobiocin/pharmacology , Signal Transduction/drug effects , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HSP90 Heat-Shock Proteins/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Molecular Structure , Novobiocin/analogs & derivatives , Novobiocin/chemistry , Structure-Activity Relationship
12.
Mol Pharmacol ; 88(1): 121-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25939977

ABSTRACT

The 90-kDa heat-shock protein (Hsp90) assists in the proper folding of numerous mutated or overexpressed signal transduction proteins that are involved in cancer. Inhibiting Hsp90 consequently is an attractive strategy for cancer therapy as the concomitant degradation of multiple oncoproteins may lead to effective antineoplastic agents. Here we report a novel C-terminal Hsp90 inhibitor, designated KU675, that exhibits potent antiproliferative and cytotoxic activity along with client protein degradation without induction of the heat-shock response in both androgen-dependent and -independent prostate cancer cell lines. In addition, KU675 demonstrates direct inhibition of Hsp90 complexes as measured by the inhibition of luciferase refolding in prostate cancer cells. In direct binding studies, the internal fluorescence signal of KU675 was used to determine the binding affinity of KU675 to recombinant Hsp90α, Hsp90ß, and Hsc70 proteins. The binding affinity (Kd) for Hsp90α was determined to be 191 µM, whereas the Kd for Hsp90ß was 726 µM, demonstrating a preference for Hsp90α. Western blot experiments with four different prostate cancer cell lines treated with KU675 supported this selectivity by inducing the degradation of Hsp90α -: dependent client proteins. KU675 also displayed binding to Hsc70 with a Kd value at 76.3 µM, which was supported in cellular by lower levels of Hsc70-specific client proteins on Western blot analyses. Overall, these findings suggest that KU675 is an Hsp90 C-terminal inhibitor, as well as a dual inhibitor of Hsc70, and may have potential use for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Coumarins/pharmacology , HSC70 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Novobiocin/analogs & derivatives , Phenylurea Compounds/pharmacology , Prostatic Neoplasms/metabolism , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Coumarins/chemical synthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Phenylurea Compounds/chemical synthesis , Protein Binding
13.
PLoS One ; 10(4): e0123314, 2015.
Article in English | MEDLINE | ID: mdl-25928540

ABSTRACT

XN4 might induce DNA damage and apoptotic cell death through reactive oxygen species (ROS). The inhibition of proliferation of K562 and K562/G01 cells was measured by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide). The mRNA levels of NADPH oxidase 1-5 (Nox1-5) genes were evaluated by qRT-PCR. The levels of extracellular reactive oxygen species (ROS), DNA damage, apoptosis, and cell cycle progression were examined by flow cytometry (FCM). Protein levels were analyzed by immunoblotting. XN4 significantly inhibited the proliferation of K562 and K562/G01 cells, with IC50 values of 3.75±0.07 µM and 2.63±0.43 µM, respectively. XN4 significantly increased the levels of Nox4 and Nox5 mRNA, stimulating the generation of intracellular ROS, inducing DNA damage and activating ATM-γ-H2AX signaling, which increased the number of cells in the S and G2/M phase of the cell cycle. Subsequently, XN4 induced apoptotic cell death by activating caspase-3 and PARP. Moreover, the above effects were all reversed by the ROS scavenger N-acetylcysteine (NAC). Additionally, XN4 can induce apoptosis in progenitor/stem cells isolated from CML patients' bone marrow. In conclusion, XN4-induced DNA damage and cell apoptosis in CML cells is mediated by the generation of ROS.


Subject(s)
DNA Damage/drug effects , Novobiocin/pharmacology , Antigens, CD34/genetics , Antigens, CD34/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , DNA Damage/genetics , Flow Cytometry , Humans , K562 Cells , Membrane Proteins/genetics , Membrane Proteins/metabolism , NADPH Oxidase 4 , NADPH Oxidase 5 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Novobiocin/analogs & derivatives , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
14.
Future Med Chem ; 6(14): 1587-605, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25367392

ABSTRACT

Hsp90 is responsible for the conformational maturation of newly synthesized polypeptides (client proteins) and the re-maturation of denatured proteins via the Hsp90 chaperone cycle. Inhibition of the Hsp90 N-terminus has emerged as a clinically relevant strategy for anticancer chemotherapeutics due to the involvement of clients in a variety of oncogenic pathways. Several immunophilins, co-chaperones and partner proteins are also necessary for Hsp90 chaperoning activity. Alternative strategies to inhibit Hsp90 function include disruption of the C-terminal dimerization domain and the Hsp90 heteroprotein complex. C-terminal inhibitors and Hsp90 co-chaperone disruptors prevent cancer cell proliferation similar to N-terminal inhibitors and destabilize client proteins without induction of heat shock proteins. Herein, current Hsp90 inhibitors, the chaperone cycle, and regulation of this cycle will be discussed.


Subject(s)
Drug Discovery , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/pharmacology , HSP90 Heat-Shock Proteins/chemistry , Humans , Models, Molecular , Neoplasms/metabolism , Novobiocin/analogs & derivatives , Novobiocin/pharmacology , Silybin , Silymarin/chemistry , Silymarin/pharmacology
15.
Biochemistry ; 53(30): 5034-41, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25020142

ABSTRACT

Understanding the mechanism of prenyltransferases is important to the design of engineered proteins capable of synthesizing derivatives of naturally occurring therapeutic agents. CloQ is a Mg(2+)-independent aromatic prenyltransferase (APTase) that transfers a dimethylallyl group to 4-hydroxyphenylpyruvate in the biosynthetic pathway for clorobiocin. APTases consist of a common ABBA fold that defines a ß-barrel containing the reaction cavity. Positively charged basic residues line the inside of the ß-barrel of CloQ to activate the pyrophosphate leaving group to replace the function of the Mg(2+) cofactor in other APTases. Classical molecular dynamics simulations of CloQ, its E281G and F68S mutants, and the related NovQ were used to explore the binding of the 4-hydroxyphenylpyruvate (4HPP) and dimethylallyl diphosphate substrates in the reactive cavity and the role of various conserved residues. Hybrid quantum mechanics/molecular mechanics potential of mean force (PMF) calculations show that the effect of the replacement of the Mg(2+) cofactor with basic residues yields a similar activation barrier for prenylation to Mg(2+)-dependent APTases like NphB. The topology of the binding pocket for 4HPP is important for selective prenylation at the ortho position of the ring. Methylation at this position alters the conformation of the substrate for O-prenylation at the phenol group. Further, a two-dimensional PMF scan shows that a "reverse" prenylation product may be a possible target for protein engineering.


Subject(s)
Dimethylallyltranstransferase/chemistry , Magnesium/metabolism , Molecular Dynamics Simulation , Novobiocin/analogs & derivatives , Prenylation/physiology , Quantum Theory , Catalytic Domain , Crystallography, X-Ray , Dimethylallyltranstransferase/physiology , Magnesium/chemistry , Novobiocin/chemistry , Phenylpyruvic Acids/chemistry , Signal Transduction , Static Electricity
16.
J Biol Chem ; 289(36): 25149-65, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25037224

ABSTRACT

The emergence of drug-resistant strains of Mycobacterium tuberculosis makes identification and validation of newer drug targets a global priority. Phosphoserine phosphatase (PSP), a key essential metabolic enzyme involved in conversion of O-phospho-l-serine to l-serine, was characterized in this study. The M. tuberculosis genome harbors all enzymes involved in l-serine biosynthesis including two PSP homologs: Rv0505c (SerB1) and Rv3042c (SerB2). In the present study, we have biochemically characterized SerB2 enzyme and developed malachite green-based high throughput assay system to identify SerB2 inhibitors. We have identified 10 compounds that were structurally different from known PSP inhibitors, and few of these scaffolds were highly specific in their ability to inhibit SerB2 enzyme, were noncytotoxic against mammalian cell lines, and inhibited M. tuberculosis growth in vitro. Surface plasmon resonance experiments demonstrated the relative binding for these inhibitors. The two best hits identified in our screen, clorobiocin and rosaniline, were bactericidal in activity and killed intracellular bacteria in a dose-dependent manner. We have also identified amino acid residues critical for these SerB2-small molecule interactions. This is the first study where we validate that M. tuberculosis SerB2 is a druggable and suitable target to pursue for further high throughput assay system screening.


Subject(s)
Bacterial Proteins/metabolism , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Mycobacterium tuberculosis/enzymology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Amino Acid Sequence , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Humans , Kinetics , Microbial Sensitivity Tests , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Novobiocin/analogs & derivatives , Novobiocin/chemistry , Novobiocin/pharmacology , Phosphoric Monoester Hydrolases/classification , Phosphoric Monoester Hydrolases/genetics , Phylogeny , Protein Binding , Protein Structure, Tertiary , Rosaniline Dyes/chemistry , Rosaniline Dyes/pharmacology , Sequence Homology, Amino Acid , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
17.
J Pharmacol Exp Ther ; 348(2): 281-92, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24263156

ABSTRACT

Impaired neuronal mitochondrial bioenergetics contributes to the pathophysiologic progression of diabetic peripheral neuropathy (DPN) and may be a focal point for disease management. We have demonstrated that modulating heat shock protein (Hsp) 90 and Hsp70 with the small-molecule drug KU-32 ameliorates psychosensory, electrophysiologic, morphologic, and bioenergetic deficits of DPN in animal models of type 1 diabetes. The current study used mouse models of type 1 and type 2 diabetes to determine the relationship of changes in sensory neuron mitochondrial bioenergetics to the onset of and recovery from DPN. The onset of DPN showed a tight temporal correlation with a decrease in mitochondrial bioenergetics in a genetic model of type 2 diabetes. In contrast, sensory hypoalgesia developed 10 weeks before the occurrence of significant declines in sensory neuron mitochondrial bioenergetics in the type 1 model. KU-32 therapy improved mitochondrial bioenergetics in both the type 1 and type 2 models, and this tightly correlated with a decrease in DPN. Mechanistically, improved mitochondrial function following KU-32 therapy required Hsp70, since the drug was ineffective in diabetic Hsp70 knockout mice. Our data indicate that changes in mitochondrial bioenergetics may rapidly contribute to nerve dysfunction in type 2 diabetes, but not type 1 diabetes, and that modulating Hsp70 offers an effective approach toward correcting sensory neuron bioenergetic deficits and DPN in both type 1 and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Neuropathies/prevention & control , HSP70 Heat-Shock Proteins/metabolism , Hypoglycemic Agents/therapeutic use , Mitochondria/drug effects , Novobiocin/analogs & derivatives , Oxidative Phosphorylation/drug effects , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Dose-Response Relationship, Drug , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , HSP70 Heat-Shock Proteins/genetics , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/blood , Hypoglycemic Agents/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Neuritis/prevention & control , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/blood , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/therapeutic use , Novobiocin/administration & dosage , Novobiocin/blood , Novobiocin/pharmacokinetics , Novobiocin/therapeutic use , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism
18.
Assay Drug Dev Technol ; 11(8): 478-88, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24127661

ABSTRACT

The 90 kDa heat-shock protein (Hsp90) and other cochaperones allow for proper folding of nascent or misfolded polypeptides. Cancer cells exploit these chaperones by maintaining the stability of mutated and misfolded oncoproteins and allowing them to evade proteosomal degradation. Inhibiting Hsp90 is an attractive strategy for cancer therapy, as the concomitant degradation of multiple oncoproteins may lead to effective anti-neoplastic agents. Unfortunately, early clinical trials have been disappointing with N-terminal Hsp90 inhibitors, as it is unclear whether the problems that plague current Hsp90 inhibitors in clinical trials are related to on-target or off-target activity. One approach to overcome these pitfalls is to identify structurally diverse scaffolds that improve Hsp90 inhibitory activity in the cancer cell milieu. Utilizing a panel of cancer cell lines that express luciferase, we have designed an in-cell Hsp90-dependent luciferase refolding assay. The assay was optimized using previously identified Hsp90 inhibitors and experimental novobiocin analogues against prostate, colon, and lung cancer cell lines. This assay exhibits good interplate precision (% CV), a signal-to-noise ratio (S/N) of ≥7, and an approximate Z-factor ranging from 0.5 to 0.7. Novobiocin analogues that revealed activity in this assay were examined via western blot experiments for client protein degradation, a hallmark of Hsp90 inhibition. Subsequently, a pilot screen was conducted using the Prestwick library, and two compounds, biperiden and ethoxyquin, revealed significant activity. Here, we report the development of an in-cell Hsp90-dependent luciferase refolding assay that is amenable across cancer cell lines for the screening of inhibitors in their specific milieu.


Subject(s)
Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , High-Throughput Screening Assays/methods , Luciferases/chemistry , Protein Refolding/drug effects , Animals , Biological Assay , Blotting, Western , Cell Line, Tumor , Coloring Agents , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Luciferases/antagonists & inhibitors , Novobiocin/analogs & derivatives , Novobiocin/pharmacology , Protein Denaturation , Rabbits , Reticulocytes/drug effects , Reticulocytes/metabolism , Rhodamines , Small Molecule Libraries
19.
Bioorg Med Chem ; 21(17): 5118-29, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23859777

ABSTRACT

Recent studies have shown that novobiocin (NB), a member of the coumermycin (CA) family of antibiotics with demonstrated DNA gyrase inhibitory activity, inhibits Heat shock protein 90 (HSP90) by binding weakly to a putative ATP-binding site within its C-terminus. To develop more potent HSP90 inhibitors that target this site and to define structure-activity relationships (SARs) for this class of compounds, we have synthesized twenty seven 3-amido-7-noviosylcoumarin analogues starting from NB and CA. These were evaluated for evidence of HSP90 inhibition using several biological assays including inhibition of cell proliferation and cell cycle arrest, induction of the heat shock response, inhibition of luciferase-refolding in vitro, and depletion of the HSP90 client protein c-erbB-2/HER-2/neu (HER2). This SAR study revealed that a substantial increase in biological activity can be achieved by introduction of an indole-2-carboxamide group in place of 4-hydroxy-isopentylbenzamido group at C-3 of NB in addition to removal/derivatization of the 4-hydroxyl group from the coumarin ring. Methylation of the 4-hydroxyl group in the coumarin moiety moderately increased biological activity as shown by compounds 11 and 13. Our most potent new analogue 19 demonstrated biological activities consistent with known HSP90-binding agents, but with greater potency than NB.


Subject(s)
Antineoplastic Agents/chemical synthesis , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Novobiocin/analogs & derivatives , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , HSP90 Heat-Shock Proteins/metabolism , Humans , MCF-7 Cells , Novobiocin/chemical synthesis , Novobiocin/toxicity , Structure-Activity Relationship
20.
Comb Chem High Throughput Screen ; 16(3): 168-79, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22934946

ABSTRACT

The half maximal inhibitory concentration (IC50) has several limitations that make it unsuitable for examining a large number of compounds in cytotoxicity studies, particularly when multiple exposure periods are tested. This article proposes a new approach to measure drug effectiveness, which allows ranking compounds according to their toxic effects on live cells. This effectiveness measure, which combines all exposure times tested, compares the growth rates of a particular cell line in the presence of the compound with its growth rate in the presence of DMSO alone. Our approach allows measuring a wider spectrum of toxicity than the IC50 approach, and allows automatic analyses of a large number of compounds. It can be easily implemented in linear regression software, provides a comparable measure of effectiveness for each investigated compound (both toxic and non-toxic), and allows statistically testing the null hypothesis that a compound is non-toxic versus the alternative that it is toxic. Importantly, our approach allows defining an automated decision rule for deciding whether a compound is significantly toxic. As an illustration, we describe the results of a cellbased study of the cytotoxicity of 24 analogs of novobiocin, a C-terminal inhibitor of heat shock protein 90 (Hsp90); the compounds were ranked in order of cytotoxicity to a panel of 18 cancer cell lines and 1 normal cell line. Our approach may also be a good alternative to computing the half maximal effective concentration (EC50) in studies searching for compounds that promote cell growth.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , High-Throughput Screening Assays/methods , Novobiocin/pharmacology , Antineoplastic Agents/chemistry , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Inhibitory Concentration 50 , Models, Biological , Models, Statistical , Neoplasms/drug therapy , Novobiocin/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL
...