Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
J Pharmacol Exp Ther ; 388(1): 171-180, 2024 01 02.
Article in English | MEDLINE | ID: mdl-37875310

ABSTRACT

Pantothenate kinase-associated neurodegeneration (PKAN) is characterized by a motor disorder with combinations of dystonia, parkinsonism, and spasticity, leading to premature death. PKAN is caused by mutations in the PANK2 gene that result in loss or reduction of PANK2 protein function. PANK2 is one of three kinases that initiate and regulate coenzyme A biosynthesis from vitamin B5, and the ability of BBP-671, an allosteric activator of pantothenate kinases, to enter the brain and elevate coenzyme A was investigated. The metabolic stability, protein binding, and membrane permeability of BBP-671 all suggest that it has the physical properties required to cross the blood-brain barrier. BBP-671 was detected in plasma, liver, cerebrospinal fluid, and brain following oral administration in rodents, demonstrating the ability of BBP-671 to penetrate the brain. The pharmacokinetic and pharmacodynamic properties of orally administered BBP-671 evaluated in cannulated rats showed that coenzyme A (CoA) concentrations were elevated in blood, liver, and brain. BBP-671 elevation of whole-blood acetyl-CoA served as a peripheral pharmacodynamic marker and provided a suitable method to assess target engagement. BBP-671 treatment elevated brain coenzyme A concentrations and improved movement and body weight in a PKAN mouse model. Thus, BBP-671 crosses the blood-brain barrier to correct the brain CoA deficiency in a PKAN mouse model, resulting in improved locomotion and survival and providing a preclinical foundation for the development of BBP-671 as a potential treatment of PKAN. SIGNIFICANCE STATEMENT: The blood-brain barrier represents a major hurdle for drugs targeting brain metabolism. This work describes the pharmacokinetic/pharmacodynamic properties of BBP-671, a pantothenate kinase activator. BBP-671 crosses the blood-brain barrier to correct the neuron-specific coenzyme A (CoA) deficiency and improve motor function in a mouse model of pantothenate kinase-associated neurodegeneration. The central role of CoA and acetyl-CoA in intermediary metabolism suggests that pantothenate kinase activators may be useful in modifying neurological metabolic disorders.


Subject(s)
Pantothenate Kinase-Associated Neurodegeneration , Mice , Animals , Rats , Pantothenate Kinase-Associated Neurodegeneration/drug therapy , Pantothenate Kinase-Associated Neurodegeneration/genetics , Acetyl Coenzyme A/metabolism , Acetyl Coenzyme A/therapeutic use , Coenzyme A/metabolism , Disease Models, Animal , Phosphotransferases (Alcohol Group Acceptor)/genetics , Brain/metabolism
2.
J Biol Chem ; 298(6): 101993, 2022 06.
Article in English | MEDLINE | ID: mdl-35490779

ABSTRACT

Fatty acid kinase (Fak) is a two-component enzyme that generates acyl-phosphate for phospholipid synthesis. Fak consists of a kinase domain protein (FakA) that phosphorylates a fatty acid enveloped by a fatty acid binding protein (FakB). The structural basis for FakB function has been established, but little is known about FakA. Here, we used limited proteolysis to define three separate FakA domains: the amino terminal FakA_N, the central FakA_L, and the carboxy terminal FakA_C. The isolated domains lack kinase activity, but activity is restored when FakA_N and FakA_L are present individually or connected as FakA_NL. The X-ray structure of the monomeric FakA_N captures the product complex with ADP and two Mg2+ ions bound at the nucleotide site. The FakA_L domain encodes the dimerization interface along with conserved catalytic residues Cys240, His282, and His284. AlphaFold analysis of FakA_L predicts the catalytic residues are spatially clustered and pointing away from the dimerization surface. Furthermore, the X-ray structure of FakA_C shows that it consists of two subdomains that are structurally related to FakB. Analytical ultracentrifugation demonstrates that FakA_C binds FakB, and site-directed mutagenesis confirms that a positively charged wedge on FakB meshes with a negatively charged groove on FakA_C. Finally, small angle X-ray scattering analysis is consistent with freely rotating FakA_N and FakA_C domains tethered by flexible linkers to FakA_L. These data reveal specific roles for the three independently folded FakA protein domains in substrate binding and catalysis.


Subject(s)
Staphylococcus aureus , Bacterial Proteins/metabolism , Binding Sites , Catalysis , Crystallography, X-Ray , Fatty Acids/metabolism , Humans , Staphylococcal Infections , Staphylococcus aureus/enzymology , Staphylococcus aureus/metabolism
3.
J Biol Chem ; 298(3): 101676, 2022 03.
Article in English | MEDLINE | ID: mdl-35122790

ABSTRACT

Fatty acid (FA) transfer proteins extract FA from membranes and sequester them to facilitate their movement through the cytosol. Detailed structural information is available for these soluble protein-FA complexes, but the structure of the protein conformation responsible for FA exchange at the membrane is unknown. Staphylococcus aureus FakB1 is a prototypical bacterial FA transfer protein that binds palmitate within a narrow, buried tunnel. Here, we define the conformational change from a "closed" FakB1 state to an "open" state that associates with the membrane and provides a path for entry and egress of the FA. Using NMR spectroscopy, we identified a conformationally flexible dynamic region in FakB1, and X-ray crystallography of FakB1 mutants captured the conformation of the open state. In addition, molecular dynamics simulations show that the new amphipathic α-helix formed in the open state inserts below the phosphate plane of the bilayer to create a diffusion channel for the hydrophobic FA tail to access the hydrocarbon core and place the carboxyl group at the phosphate layer. The membrane binding and catalytic properties of site-directed mutants were consistent with the proposed membrane docked structure predicted by our molecular dynamics simulations. Finally, the structure of the bilayer-associated conformation of FakB1 has local similarities with mammalian FA binding proteins and provides a conceptual framework for how these proteins interact with the membrane to create a diffusion channel from the FA location in the bilayer to the protein interior.


Subject(s)
Bacterial Proteins , Fatty Acid-Binding Proteins , Fatty Acids , Animals , Bacterial Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , Fatty Acids/metabolism , Ligands , Mammals/metabolism , Membranes/chemistry , Membranes/metabolism , Phosphates/metabolism , Protein Conformation , Staphylococcus aureus/chemistry , Staphylococcus aureus/metabolism
4.
J Inherit Metab Dis ; 46(1): 28-42, 2023 01.
Article in English | MEDLINE | ID: mdl-36251252

ABSTRACT

Propionic acidemia (PA, OMIM 606054) is a devastating inborn error of metabolism arising from mutations that reduce the activity of the mitochondrial enzyme propionyl-CoA carboxylase (PCC). The defects in PCC reduce the concentrations of nonesterified coenzyme A (CoASH), thus compromising mitochondrial function and disrupting intermediary metabolism. Here, we use a hypomorphic PA mouse model to test the effectiveness of BBP-671 in correcting the metabolic imbalances in PA. BBP-671 is a high-affinity allosteric pantothenate kinase activator that counteracts feedback inhibition of the enzyme to increase the intracellular concentration of CoA. Liver CoASH and acetyl-CoA are depressed in PA mice and BBP-671 treatment normalizes the cellular concentrations of these two key cofactors. Hepatic propionyl-CoA is also reduced by BBP-671 leading to an improved intracellular C3:C2-CoA ratio. Elevated plasma C3:C2-carnitine ratio and methylcitrate, hallmark biomarkers of PA, are significantly reduced by BBP-671. The large elevations of malate and α-ketoglutarate in the urine of PA mice are biomarkers for compromised tricarboxylic acid cycle activity and BBP-671 therapy reduces the amounts of both metabolites. Furthermore, the low survival of PA mice is restored to normal by BBP-671. These data show that BBP-671 relieves CoA sequestration, improves mitochondrial function, reduces plasma PA biomarkers, and extends the lifespan of PA mice, providing the preclinical foundation for the therapeutic potential of BBP-671.


Subject(s)
Propionic Acidemia , Mice , Animals , Propionic Acidemia/genetics , Methylmalonyl-CoA Decarboxylase/genetics , Methylmalonyl-CoA Decarboxylase/metabolism , Disease Models, Animal , Mitochondria/metabolism , Carnitine
5.
Nucleic Acids Res ; 49(3): 1609-1618, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33469660

ABSTRACT

The endonuclease activity within the influenza virus cap-snatching process is a proven therapeutic target. The anti-influenza drug baloxavir is highly effective, but is associated with resistance mutations that threaten its clinical efficacy. The endonuclease resides within the N-terminal domain of the PA subunit (PAN) of the influenza RNA dependent RNA polymerase, and we report here complexes of PAN with RNA and DNA oligonucleotides to understand its specificity and the structural basis of baloxavir resistance mutations. The RNA and DNA oligonucleotides bind within the substrate binding groove of PAN in a similar fashion, explaining the ability of the enzyme to cleave both substrates. The individual nucleotides occupy adjacent conserved pockets that flank the two-metal active site. However, the 2' OH of the RNA ribose moieties engage in additional interactions that appear to optimize the binding and cleavage efficiency for the natural substrate. The major baloxavir resistance mutation at position 38 is at the core of the substrate binding site, but structural studies and modeling suggest that it maintains the necessary virus fitness via compensating interactions with RNA. These studies will facilitate the development of new influenza therapeutics that spatially match the substrate and are less likely to elicit resistance mutations.


Subject(s)
Endoribonucleases/chemistry , Influenza A Virus, H1N1 Subtype/enzymology , Viral Proteins/chemistry , Antiviral Agents/chemistry , DNA/chemistry , Dibenzothiepins/chemistry , Endoribonucleases/metabolism , Models, Molecular , Morpholines/chemistry , Pyridones/chemistry , RNA/chemistry , Substrate Specificity , Triazines/chemistry , Viral Proteins/metabolism
6.
Bioorg Med Chem ; 52: 116504, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34814071

ABSTRACT

Pantothenate kinase (PANK) is the critical regulator of intracellular levels of coenzyme A and has emerged as an attractive target for treating neurological and metabolic disorders. This report describes the optimization, synthesis, and full structure-activity relationships of a new chemical series of pantothenate competitive PANK inhibitors. Potent drug-like molecules were obtained by optimizing a high throughput screening hit, using lipophilic ligand efficiency (LipE) derived from human PANK3 IC50 values to guide ligand development. X-ray crystal structures of PANK3 with index inhibitors from the optimization were determined to rationalize the emerging structure activity relationships. The analysis revealed a key bidentate hydrogen bonding interaction between pyridazine and R306' as a major contributor to the LipE gain observed in the optimization. A tractable series of PANK3 modulators with nanomolar potency, excellent LipE values, desirable physicochemical properties, and a well-defined structural binding mode was produced from this study.


Subject(s)
Drug Discovery , High-Throughput Screening Assays , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pyridazines/pharmacology , Dose-Response Relationship, Drug , Humans , Hydrogen Bonding , Ligands , Molecular Structure , Pyridazines/chemical synthesis , Pyridazines/chemistry , Structure-Activity Relationship
7.
Angew Chem Int Ed Engl ; 60(51): 26663-26670, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34614283

ABSTRACT

Targeting cereblon (CRBN) is currently one of the most frequently reported proteolysis-targeting chimera (PROTAC) approaches, owing to favorable drug-like properties of CRBN ligands, immunomodulatory imide drugs (IMiDs). However, IMiDs are known to be inherently unstable, readily undergoing hydrolysis in body fluids. Here we show that IMiDs and IMiD-based PROTACs rapidly hydrolyze in commonly utilized cell media, which significantly affects their cell efficacy. We designed novel CRBN binders, phenyl glutarimide (PG) analogues, and showed that they retained affinity for CRBN with high ligand efficiency (LE >0.48) and displayed improved chemical stability. Our efforts led to the discovery of PG PROTAC 4 c (SJ995973), a uniquely potent degrader of bromodomain and extra-terminal (BET) proteins that inhibited the viability of human acute myeloid leukemia MV4-11 cells at low picomolar concentrations (IC50 =3 pM; BRD4 DC50 =0.87 nM). These findings strongly support the utility of PG derivatives in the design of CRBN-directed PROTACs.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Piperidones/chemistry , Ubiquitin-Protein Ligases/chemistry , Humans , Hydrolysis , Proteolysis
8.
J Biol Chem ; 294(44): 16416-16428, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31530637

ABSTRACT

Streptococcus pneumoniae is responsible for the majority of pneumonia, motivating ongoing searches for insights into its physiology that could enable new treatments. S. pneumoniae responds to exogenous fatty acids by suppressing its de novo biosynthetic pathway and exclusively utilizing extracellular fatty acids for membrane phospholipid synthesis. The first step in exogenous fatty acid assimilation is phosphorylation by fatty acid kinase (FakA), whereas bound by a fatty acid-binding protein (FakB). Staphylococcus aureus has two binding proteins, whereas S. pneumoniae expresses three. The functions of these binding proteins were not clear. We determined the SpFakB1- and SpFakB2-binding proteins were bioinformatically related to the two binding proteins of Staphylococcus aureus, and biochemical and X-ray crystallographic analysis showed that SpFakB1 selectively bound saturates, whereas SpFakB2 allows the activation of monounsaturates akin to their S. aureus counterparts. The distinct SpFakB3 enables the utilization of polyunsaturates. The SpFakB3 crystal structure in complex with linoleic acid reveals an expanded fatty acid-binding pocket within the hydrophobic interior of SpFakB3 that explains its ability to accommodate multiple cis double bonds. SpFakB3 also utilizes a different hydrogen bond network than other FakBs to anchor the fatty acid carbonyl and stabilize the protein. S. pneumoniae strain JMG1 (ΔfakB3) was deficient in incorporation of linoleate from human serum verifying the role of FakB3 in this process. Thus, the multiple FakBs of S. pneumoniae permit the utilization of the entire spectrum of mammalian fatty acid structures to construct its membrane.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Fatty Acids/metabolism , Streptococcus pneumoniae/metabolism , Bacterial Proteins/metabolism , Biosynthetic Pathways , Fatty Acid-Binding Proteins/physiology , Fatty Acid-Binding Proteins/ultrastructure , Fatty Acids, Unsaturated/metabolism , Host-Pathogen Interactions/physiology , Humans , Phospholipids/metabolism , Phosphorylation , Serum/chemistry , Staphylococcus aureus/metabolism
9.
J Biol Chem ; 294(1): 38-49, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30429218

ABSTRACT

Fatty acid (FA) kinase produces acyl-phosphate for the synthesis of membrane phospholipids in Gram-positive bacterial pathogens. FA kinase consists of a kinase protein (FakA) that phosphorylates an FA substrate bound to a second module, an FA-binding protein (FakB). Staphylococcus aureus expresses two distinct, but related, FakBs with different FA selectivities. Here, we report the structures of FakB1 bound to four saturated FAs at 1.6-1.93 Å resolution. We observed that the different FA structures are accommodated within a slightly curved hydrophobic cavity whose length is governed by the conformation of an isoleucine side chain at the end of the tunnel. The hydrophobic tunnel in FakB1 prevents the binding of cis-unsaturated FAs, which are instead accommodated by the kinked tunnel within the FakB2 protein. The differences in the FakB interiors are not propagated to the proteins' surfaces, preserving the protein-protein interactions with their three common partners, FakA, PlsX, and PlsY. Using cellular thermal shift analyses, we found that FakB1 binds FA in vivo, whereas a significant proportion of FakB2 does not. Incorporation of exogenous FA into phospholipid in ΔfakB1 and ΔfakB2 S. aureus knockout strains revealed that FakB1 does not efficiently activate unsaturated FAs. FakB2 preferred unsaturated FAs, but also allowed the incorporation of saturated FAs. These results are consistent with a model in which FakB1 primarily functions in the recycling of the saturated FAs produced by S. aureus metabolism, whereas FakB2 activates host-derived oleate, which S. aureus does not produce but is abundant at infection sites.


Subject(s)
Bacterial Proteins/chemistry , Fatty Acid-Binding Proteins/chemistry , Oleic Acid/chemistry , Staphylococcus aureus/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Oleic Acid/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Substrate Specificity
10.
Nucleic Acids Res ; 46(10): 5308-5318, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29718457

ABSTRACT

During infection, bacteriophage T4 produces the MotA transcription factor that redirects the host RNA polymerase to the expression of T4 middle genes. The C-terminal 'double-wing' domain of MotA binds specifically to the MotA box motif of middle T4 promoters. We report the crystal structure of this complex, which reveals a new mode of protein-DNA interaction. The domain binds DNA mostly via interactions with the DNA backbone, but the binding is enhanced in the specific cognate structure by additional interactions with the MotA box motif in both the major and minor grooves. The linker connecting the two MotA domains plays a key role in stabilizing the complex via minor groove interactions. The structure is consistent with our previous model derived from chemical cleavage experiments using the entire transcription complex. α- and ß-d-glucosyl-5-hydroxymethyl-deoxycytosine replace cytosine in T4 DNA, and docking simulations indicate that a cavity in the cognate structure can accommodate the modified cytosine. Binding studies confirm that the modification significantly enhances the binding affinity of MotA for the DNA. Consequently, our work reveals how a DNA modification can extend the uniqueness of small DNA motifs to facilitate the specificity of protein-DNA interactions.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Cytosine/analogs & derivatives , Cytosine/chemistry , Cytosine/metabolism , DNA/chemistry , DNA-Binding Proteins/genetics , Molecular Docking Simulation , Mutagenesis , Protein Conformation , Transcription Factors/genetics , Viral Proteins/genetics
11.
Mol Pharmacol ; 96(1): 73-89, 2019 07.
Article in English | MEDLINE | ID: mdl-31043459

ABSTRACT

Interfering with microtubule dynamics is a well-established strategy in cancer treatment; however, many microtubule-targeting agents are associated with drug resistance and adverse effects. Substantial evidence points to ATP-binding cassette (ABC) transporters as critical players in the development of resistance. Herein, we demonstrate the efficacy of DJ95 (2-(1H-indol-6-yl)-4-(3,4,5-trimethoxyphenyl)-1H-imidazo[4,5-c]pyridine), a novel tubulin inhibitor, in a variety of cancer cell lines, including malignant melanomas, drug-selected resistant cell lines, specific ABC transporter-overexpressing cell lines, and the National Cancer Institute 60 cell line panel. DJ95 treatment inhibited cancer cell migration, caused morphologic changes to the microtubule network foundation, and severely disrupted mitotic spindle formation of mitotic cells. The high-resolution crystal structure of DJ95 in complex with tubulin protein and the detailed molecular interactions confirmed its direct binding to the colchicine site. In vitro pharmacological screening of DJ95 using SafetyScreen44 (Eurofins Cerep-Panlabs) revealed no significant off-target interactions, and pharmacokinetic analysis showed that DJ95 was maintained at therapeutically relevant plasma concentrations for up to 24 hours in mice. In an A375 xenograft model in nude mice, DJ95 inhibited tumor growth and disrupted tumor vasculature in xenograft tumors. These results demonstrate that DJ95 is potent against a variety of cell lines, demonstrated greater potency to ABC transporter-overexpressing cell lines than existing tubulin inhibitors, directly targets the colchicine binding domain, exhibits significant antitumor efficacy, and demonstrates vascular-disrupting properties. Collectively, these data suggest that DJ95 has great potential as a cancer therapeutic, particularly for multidrug resistance phenotypes, and warrants further development. SIGNIFICANCE STATEMENT: Paclitaxel is a widely used tubulin inhibitor for cancer therapy, but its clinical efficacy is often limited by the development of multidrug resistance. In this study, we reported the preclinical characterization of a new tubulin inhibitor DJ95, and demonstrated its abilities to overcome paclitaxel resistance, disrupt tumor vasculature, and exhibit significant antitumor efficacy.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Melanoma/drug therapy , Small Molecule Libraries/administration & dosage , Tubulin Modulators/administration & dosage , Tubulin/chemistry , ATP-Binding Cassette Transporters/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Colchicine/metabolism , Crystallography, X-Ray , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Imidazoles/administration & dosage , Imidazoles/chemistry , Imidazoles/pharmacology , Male , Melanoma/metabolism , Mice , Mice, Nude , Pyridines/administration & dosage , Pyridines/chemistry , Pyridines/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Tubulin/metabolism , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
12.
Proc Natl Acad Sci U S A ; 113(12): 3275-80, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26951671

ABSTRACT

The UvsY recombination mediator protein is critical for efficient homologous recombination in bacteriophage T4 and is the functional analog of the eukaryotic Rad52 protein. During T4 homologous recombination, the UvsX recombinase has to compete with the prebound gp32 single-stranded binding protein for DNA-binding sites and UvsY stimulates this filament nucleation event. We report here the crystal structure of UvsY in four similar open-barrel heptameric assemblies and provide structural and biophysical insights into its function. The UvsY heptamer was confirmed in solution by centrifugation and light scattering, and thermodynamic analyses revealed that the UvsY-ssDNA interaction occurs within the assembly via two distinct binding modes. Using surface plasmon resonance, we also examined the binding of UvsY to both ssDNA and the ssDNA-gp32 complex. These analyses confirmed that ssDNA can bind UvsY and gp32 independently and also as a ternary complex. They also showed that residues located on the rim of the heptamer are required for optimal binding to ssDNA, thus identifying the putative ssDNA-binding surface. We propose a model in which UvsY promotes a helical ssDNA conformation that disfavors the binding of gp32 and initiates the assembly of the ssDNA-UvsX filament.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/physiology , Viral Proteins/chemistry , Viral Proteins/physiology , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Protein Conformation , Structure-Activity Relationship
13.
Proc Natl Acad Sci U S A ; 113(13): 3669-74, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976575

ABSTRACT

The influenza endonuclease is an essential subdomain of the viral RNA polymerase. It processes host pre-mRNAs to serve as primers for viral mRNA and is an attractive target for antiinfluenza drug discovery. Compound L-742,001 is a prototypical endonuclease inhibitor, and we found that repeated passaging of influenza virus in the presence of this drug did not lead to the development of resistant mutant strains. Reduced sensitivity to L-742,001 could only be induced by creating point mutations via a random mutagenesis strategy. These mutations mapped to the endonuclease active site where they can directly impact inhibitor binding. Engineered viruses containing the mutations showed resistance to L-742,001 both in vitro and in vivo, with only a modest reduction in fitness. Introduction of the mutations into a second virus also increased its resistance to the inhibitor. Using the isolated wild-type and mutant endonuclease domains, we used kinetics, inhibitor binding and crystallography to characterize how the two most significant mutations elicit resistance to L-742,001. These studies lay the foundation for the development of a new class of influenza therapeutics with reduced potential for the development of clinical endonuclease inhibitor-resistant influenza strains.


Subject(s)
Endonucleases/antagonists & inhibitors , Endonucleases/genetics , Enzyme Inhibitors/pharmacology , Hydroxybutyrates/pharmacology , Influenza A virus/drug effects , Piperidines/pharmacology , Animals , Catalytic Domain/genetics , Crystallography, X-Ray , Dogs , Drug Resistance, Viral/genetics , Endonucleases/metabolism , Genetic Variation , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A virus/enzymology , Influenza A virus/genetics , Kinetics , Madin Darby Canine Kidney Cells , Models, Molecular , Mutagenesis , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism
14.
J Biol Chem ; 291(12): 6292-303, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26774272

ABSTRACT

Fatty acid kinase (Fak) is a ubiquitous Gram-positive bacterial enzyme consisting of an ATP-binding protein (FakA) that phosphorylates the fatty acid bound to FakB. In Staphylococcus aureus, Fak is a global regulator of virulence factor transcription and is essential for the activation of exogenous fatty acids for incorporation into phospholipids. The 1.2-Å x-ray structure of S. aureus FakB2, activity assays, solution studies, site-directed mutagenesis, and in vivo complementation were used to define the functions of the five conserved residues that define the FakB protein family (Pfam02645). The fatty acid tail is buried within the protein, and the exposed carboxyl group is bound by a Ser-93-fatty acid carboxyl-Thr-61-His-266 hydrogen bond network. The guanidinium of the invariant Arg-170 is positioned to potentially interact with a bound acylphosphate. The reduced thermal denaturation temperatures of the T61A, S93A, and H266A FakB2 mutants illustrate the importance of the hydrogen bond network in protein stability. The FakB2 T61A, S93A, and H266A mutants are 1000-fold less active in the Fak assay, and the R170A mutant is completely inactive. All FakB2 mutants form FakA(FakB2)2 complexes except FakB2(R202A), which is deficient in FakA binding. Allelic replacement shows that strains expressing FakB2 mutants are defective in fatty acid incorporation into phospholipids and virulence gene transcription. These conserved residues are likely to perform the same critical functions in all bacterial fatty acid-binding proteins.


Subject(s)
Bacterial Proteins/chemistry , Fatty Acid-Binding Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Conserved Sequence , Fatty Acid-Binding Proteins/metabolism , Fatty Acids/chemistry , Gene Expression , Hydrogen Bonding , Models, Molecular , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Protein Stability , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
15.
J Biol Chem ; 291(42): 22302-22314, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27555321

ABSTRACT

Pantothenate kinase is the master regulator of CoA biosynthesis and is feedback-inhibited by acetyl-CoA. Comparison of the human PANK3·acetyl-CoA complex to the structures of PANK3 in four catalytically relevant complexes, 5'-adenylyl-ß,γ-imidodiphosphate (AMPPNP)·Mg2+, AMPPNP·Mg2+·pantothenate, ADP·Mg2+·phosphopantothenate, and AMP phosphoramidate (AMPPN)·Mg2+, revealed a large conformational change in the dimeric enzyme. The amino-terminal nucleotide binding domain rotates to close the active site, and this allows the P-loop to engage ATP and facilitates required substrate/product interactions at the active site. Biochemical analyses showed that the transition between the inactive and active conformations, as assessed by the binding of either ATP·Mg2+ or acyl-CoA to PANK3, is highly cooperative indicating that both protomers move in concert. PANK3(G19V) cannot bind ATP, and biochemical analyses of an engineered PANK3/PANK3(G19V) heterodimer confirmed that the two active sites are functionally coupled. The communication between the two protomers is mediated by an α-helix that interacts with the ATP-binding site at its amino terminus and with the substrate/inhibitor-binding site of the opposite protomer at its carboxyl terminus. The two α-helices within the dimer together with the bound ligands create a ring that stabilizes the assembly in either the active closed conformation or the inactive open conformation. Thus, both active sites of the dimeric mammalian pantothenate kinases coordinately switch between the on and off states in response to intracellular concentrations of ATP and its key negative regulators, acetyl(acyl)-CoA.


Subject(s)
Acyl Coenzyme A/chemistry , Mutation, Missense , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Acyl Coenzyme A/metabolism , Allosteric Regulation , Amino Acid Substitution , Humans , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Domains , Protein Structure, Secondary
16.
Proc Natl Acad Sci U S A ; 111(12): 4466-71, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24616519

ABSTRACT

Nucleophosmin (NPM1) is a multifunctional phospho-protein with critical roles in ribosome biogenesis, tumor suppression, and nucleolar stress response. Here we show that the N-terminal oligomerization domain of NPM1 (Npm-N) exhibits structural polymorphism by populating conformational states ranging from a highly ordered, folded pentamer to a highly disordered monomer. The monomer-pentamer equilibrium is modulated by posttranslational modification and protein binding. Phosphorylation drives the equilibrium in favor of monomeric forms, and this effect can be reversed by Npm-N binding to its interaction partners. We have identified a short, arginine-rich linear motif in NPM1 binding partners that mediates Npm-N oligomerization. We propose that the diverse functional repertoire associated with NPM1 is controlled through a regulated unfolding mechanism signaled through posttranslational modifications and intermolecular interactions.


Subject(s)
Biopolymers/chemistry , Nuclear Proteins/chemistry , Amino Acid Sequence , Biopolymers/metabolism , Chromatography, Gel , Humans , Models, Molecular , Molecular Sequence Data , Native Polyacrylamide Gel Electrophoresis , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/metabolism , Nucleophosmin , Phosphorylation , Protein Binding , Protein Conformation
17.
Bioorg Med Chem Lett ; 26(16): 3950-4, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27423480

ABSTRACT

The sulfonamide class of antibiotics has been in continuous use for over 70years. They are thought to act by directly inhibiting dihydropteroate synthase (DHPS), and also acting as prodrugs that sequester pterin pools by forming dead end pterin-sulfonamide conjugates. In this study, eight pterin-sulfonamide conjugates were synthesized using a novel synthetic strategy and their biochemical and microbiological properties were investigated. The conjugates were shown to competitively inhibit DHPS, and inhibition was enhanced by the presence of pyrophosphate that is crucial to catalysis and is known to promote an ordering of the DHPS active site. The co-crystal structure of Yersinia pestis DHPS bound to one of the more potent conjugates revealed a mode of binding that is similar to that of the enzymatic product analog pteroic acid. The antimicrobial activities of the pterin-sulfonamide conjugates were measured against Escherichia coli in the presence and absence of folate precursors and dependent metabolites. These results show that the conjugates have appreciable antibacterial activity and act by an on target, anti-folate pathway mechanism rather than as simple dead end products.


Subject(s)
Anti-Bacterial Agents/chemistry , Dihydropteroate Synthase/antagonists & inhibitors , Pterins/chemistry , Sulfonamides/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Dihydropteroate Synthase/metabolism , Escherichia coli/drug effects , Folic Acid/chemistry , Molecular Docking Simulation , Structure-Activity Relationship , Yersinia pestis/enzymology
18.
J Biol Chem ; 289(32): 22365-76, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24958721

ABSTRACT

The major phospholipid classes of the obligate intracellular bacterial parasite Chlamydia trachomatis are the same as its eukaryotic host except that they also contain chlamydia-made branched-chain fatty acids in the 2-position. Genomic analysis predicts that C. trachomatis is capable of type II fatty acid synthesis (FASII). AFN-1252 was deployed as a chemical tool to specifically inhibit the enoyl-acyl carrier protein reductase (FabI) of C. trachomatis to determine whether chlamydial FASII is essential for replication within the host. The C. trachomatis FabI (CtFabI) is a homotetramer and exhibited typical FabI kinetics, and its expression complemented an Escherichia coli fabI(Ts) strain. AFN-1252 inhibited CtFabI by binding to the FabI·NADH complex with an IC50 of 0.9 µM at saturating substrate concentration. The x-ray crystal structure of the CtFabI·NADH·AFN-1252 ternary complex revealed the specific interactions between the drug, protein, and cofactor within the substrate binding site. AFN-1252 treatment of C. trachomatis-infected HeLa cells at any point in the infectious cycle caused a decrease in infectious titers that correlated with a decrease in branched-chain fatty acid biosynthesis. AFN-1252 treatment at the time of infection prevented the first cell division of C. trachomatis, although the cell morphology suggested differentiation into a metabolically active reticulate body. These results demonstrate that FASII activity is essential for C. trachomatis proliferation within its eukaryotic host and validate CtFabI as a therapeutic target against C. trachomatis.


Subject(s)
Chlamydia trachomatis/metabolism , Fatty Acids/biosynthesis , Amino Acid Sequence , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Benzofurans/pharmacology , Cell Proliferation/drug effects , Chlamydia trachomatis/genetics , Chlamydia trachomatis/pathogenicity , Crystallography, X-Ray , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Enzyme Inhibitors/pharmacology , Fatty Acid Synthase, Type II/antagonists & inhibitors , Fatty Acid Synthase, Type II/genetics , Fatty Acid Synthase, Type II/metabolism , Genes, Bacterial , HeLa Cells , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Structure, Quaternary , Pyrones/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
19.
Nat Chem Biol ; 9(3): 163-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23340338

ABSTRACT

Following DNA damage, nuclear p53 induces the expression of PUMA, a BH3-only protein that binds and inhibits the antiapoptotic BCL-2 repertoire, including BCL-xL. PUMA, unique among BH3-only proteins, disrupts the interaction between cytosolic p53 and BCL-xL, allowing p53 to promote apoptosis via direct activation of the BCL-2 effector molecules BAX and BAK. Structural investigations using NMR spectroscopy and X-ray crystallography revealed that PUMA binding induced partial unfolding of two α-helices within BCL-xL. Wild-type PUMA or a PUMA mutant incapable of causing binding-induced unfolding of BCL-xL equivalently inhibited the antiapoptotic BCL-2 repertoire to sensitize for death receptor-activated apoptosis, but only wild-type PUMA promoted p53-dependent, DNA damage-induced apoptosis. Our data suggest that PUMA-induced partial unfolding of BCL-xL disrupts interactions between cytosolic p53 and BCL-xL, releasing the bound p53 to initiate apoptosis. We propose that regulated unfolding of BCL-xL provides a mechanism to promote PUMA-dependent signaling within the apoptotic pathways.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Protein Unfolding , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-X Protein/metabolism , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Humans , Models, Molecular , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/chemistry , bcl-X Protein/chemistry
20.
Bioorg Med Chem ; 23(13): 3248-59, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25975638

ABSTRACT

Benzoxazolone and benzothiazolone were used as template blocks to develop two series of dimers as anti-inflammatory and analgesic agents based on the concept of bivalent ligands. The first series (I) involved varying the carbon chain lengths extending from the piperazine core to the nitrogen atom of the dibenzo[d]oxazol-2(3H)-one or dibenzo[d]thiazol-2(3H)-one. The second series (II) was designed by changing the attachment point. All compounds were screened for their in vitro anti-inflammatory activity in terms of the inhibition of inducible nitric oxide synthase (iNOS) and nuclear factor kappa B (NF-κB). Seventeen compounds inhibited both targets. Eleven of them exhibited IC50 values below 3µM while five compounds showed IC50 values of 1µM or below. Most of the compounds were found to be devoid of cytotoxicity against mammalian kidney and solid tumors cell lines up to 25µg/mL. In vivo anti-inflammatory and antinociceptive studies revealed that compounds 3j, 5t and 8b have significant anti-inflammatory and analgesic activity comparable to that of indomethacin and ketorolac, respectively.


Subject(s)
Analgesics/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Benzothiazoles/chemical synthesis , Benzoxazoles/chemical synthesis , Edema/drug therapy , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzothiazoles/pharmacology , Benzoxazoles/pharmacology , Carrageenan , Cell Line, Tumor , Chlorocebus aethiops , Dimerization , Drug Design , Edema/chemically induced , Edema/pathology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hindlimb , Humans , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Molecular Docking Simulation , NF-kappa B/antagonists & inhibitors , NF-kappa B/chemistry , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase Type II/metabolism , Rats , Structure-Activity Relationship , Swine , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL