Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Nature ; 584(7821): 479-483, 2020 08.
Article in English | MEDLINE | ID: mdl-32788728

ABSTRACT

Lipopolysaccharide (LPS) resides in the outer membrane of Gram-negative bacteria where it is responsible for barrier function1,2. LPS can cause death as a result of septic shock, and its lipid A core is the target of polymyxin antibiotics3,4. Despite the clinical importance of polymyxins and the emergence of multidrug resistant strains5, our understanding of the bacterial factors that regulate LPS biogenesis is incomplete. Here we characterize the inner membrane protein PbgA and report that its depletion attenuates the virulence of Escherichia coli by reducing levels of LPS and outer membrane integrity. In contrast to previous claims that PbgA functions as a cardiolipinĀ transporter6-9, our structural analyses and physiological studies identify a lipid A-binding motif along the periplasmic leaflet of the inner membrane. Synthetic PbgA-derived peptides selectively bind to LPS in vitro and inhibit the growth of diverse Gram-negative bacteria, including polymyxin-resistant strains. Proteomic, genetic and pharmacological experiments uncover a model in which direct periplasmic sensing of LPS by PbgA coordinates the biosynthesis of lipid A by regulating the stability of LpxC, a key cytoplasmic biosynthetic enzyme10-12. In summary, we find that PbgA has an unexpected but essential role in the regulation of LPS biogenesis, presents a new structural basis for the selective recognition of lipids, and provides opportunities for future antibiotic discovery.


Subject(s)
Cell Membrane/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/chemistry , Escherichia coli/pathogenicity , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Amino Acid Motifs , Bacterial Outer Membrane/chemistry , Bacterial Outer Membrane/metabolism , Binding Sites , Cell Membrane/metabolism , Enzyme Stability , Escherichia coli/cytology , Escherichia coli/drug effects , Genes, Essential , Hydrolases/chemistry , Hydrolases/metabolism , Lipid A/chemistry , Lipid A/metabolism , Lipopolysaccharides/biosynthesis , Microbial Sensitivity Tests , Microbial Viability/drug effects , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Periplasm/chemistry , Periplasm/metabolism , Protein Binding , Virulence
2.
Biochemistry ; 62(3): 633-644, 2023 02 07.
Article in English | MEDLINE | ID: mdl-34985287

ABSTRACT

Autophagy-related proteins (Atgs) drive the lysosome-mediated degradation pathway, autophagy, to enable the clearance of dysfunctional cellular components and maintain homeostasis. In humans, this process is driven by the mammalian Atg8 (mAtg8) family of proteins comprising the LC3 and GABARAP subfamilies. The mAtg8 proteins play essential roles in the formation and maturation of autophagosomes and the capture of specific cargo through binding to the conserved LC3-interacting region (LIR) sequence within target proteins. Modulation of interactions of mAtg8 with its target proteins via small-molecule ligands would enable further interrogation of their function. Here we describe unbiased fragment and DNA-encoded library (DEL) screening approaches for discovering LC3 small-molecule ligands. Both strategies resulted in compounds that bind to LC3, with the fragment hits favoring a conserved hydrophobic pocket in mATG8 proteins, as detailed by LC3A-fragment complex crystal structures. Our findings demonstrate that the malleable LIR-binding surface can be readily targeted by fragments; however, rational design of additional interactions to drive increased affinity proved challenging. DEL libraries, which combine small, fragment-like building blocks into larger scaffolds, yielded higher-affinity binders and revealed an unexpected potential for reversible, covalent ligands. Moreover, DEL hits identified possible vectors for synthesizing fluorescent probes or bivalent molecules for engineering autophagic degradation of specific targets.


Subject(s)
Autophagy , Microtubule-Associated Proteins , Humans , Animals , Microtubule-Associated Proteins/metabolism , Ligands , Autophagy-Related Protein 8 Family/chemistry , Autophagosomes/metabolism , Mammals/metabolism
3.
Biochem Soc Trans ; 48(4): 1323-1336, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32794575

ABSTRACT

The proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates plasma LDL cholesterol levels by binding to the liver LDL receptor (LDLR) and promoting its degradation. Therefore, PCSK9 has become a compelling new therapeutic target for lipid lowering and the prevention of cardiovascular disease. PCSK9 contains two regions of conformational flexibility, the N-terminal regions of the prodomain and of the catalytic domain. The recognition that the latter region, the so-called P' helix, is able to transition from an α-helical to a disordered state gave rise to new strategies to develop small molecule inhibitors of PCSK9 for lipid lowering. In the ordered state the P' helix is buried in a groove of the PCSK9 catalytic domain located next to the main LDLR binding site. The transition to a disordered state leaves the groove site vacated and accessible for compounds to antagonize LDLR binding. By use of a groove-directed phage display strategy we were able to identify several groove-binding peptides. Based on structural information of PCSK9-peptide complexes, a minimized groove-binding peptide was generated and utilized as an anchor to extend towards the adjacent main LDLR binding site, either by use of a phage-displayed peptide extension library, or by appending organic moieties to yield organo-peptides. Both strategies led to antagonists with pharmacologic activities in cell-based assays. The intricate bipartite mechanism of the potent organo-peptide inhibitors was revealed by structural studies, showing that the core peptide occupies the N-terminal groove, while the organic moiety interacts with the LDLR binding site to create antagonism. These findings validate the PCSK9 groove as an attractive target site and should inspire the development of a new class of small molecule antagonists of PCSK9.


Subject(s)
Anticholesteremic Agents/chemistry , Cholesterol, LDL/blood , Drug Design , Proprotein Convertase 9/metabolism , Serine Proteinase Inhibitors/chemistry , Animals , Anticholesteremic Agents/pharmacology , Binding Sites , Humans , PCSK9 Inhibitors , Proprotein Convertase 9/chemistry , Receptors, LDL/metabolism , Serine Proteinase Inhibitors/pharmacology
4.
Article in English | MEDLINE | ID: mdl-29339384

ABSTRACT

The outer membrane is an essential structural component of Gram-negative bacteria that is composed of lipoproteins, lipopolysaccharides, phospholipids, and integral Ɵ-barrel membrane proteins. A dedicated machinery, called the Lol system, ensures proper trafficking of lipoproteins from the inner to the outer membrane. The LolCDE ABC transporter is the inner membrane component, which is essential for bacterial viability. Here, we report a novel pyrrolopyrimidinedione compound, G0507, which was identified in a phenotypic screen for inhibitors of Escherichia coli growth followed by selection of compounds that induced the extracytoplasmic σE stress response. Mutations in lolC, lolD, and lolE conferred resistance to G0507, suggesting LolCDE as its molecular target. Treatment of E. coli cells with G0507 resulted in accumulation of fully processed Lpp, an outer membrane lipoprotein, in the inner membrane. Using purified protein complexes, we found that G0507 binds to LolCDE and stimulates its ATPase activity. G0507 still binds to LolCDE harboring a Q258K substitution in LolC (LolCQ258K), which confers high-level resistance to G0507 in vivo but no longer stimulates ATPase activity. Our work demonstrates that G0507 has significant promise as a chemical probe to dissect lipoprotein trafficking in Gram-negative bacteria.


Subject(s)
Gram-Negative Bacteria/metabolism , Lipoproteins/metabolism , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gram-Negative Bacteria/drug effects , Lipoproteins/genetics , Mutation/genetics , Protein Transport/drug effects , Protein Transport/genetics
5.
Article in English | MEDLINE | ID: mdl-30104274

ABSTRACT

There is a critical need for new antibacterial strategies to counter the growing problem of antibiotic resistance. In Gram-negative bacteria, the outer membrane (OM) provides a protective barrier against antibiotics and other environmental insults. The outer leaflet of the outer membrane is primarily composed of lipopolysaccharide (LPS). Outer membrane biogenesis presents many potentially compelling drug targets as this pathway is absent in higher eukaryotes. Most proteins involved in LPS biosynthesis and transport are essential; however, few compounds have been identified that inhibit these proteins. The inner membrane ABC transporter MsbA carries out the first essential step in the trafficking of LPS to the outer membrane. We conducted a biochemical screen for inhibitors of MsbA and identified a series of quinoline compounds that kill Escherichia coli through inhibition of its ATPase and transport activity, with no loss of activity against clinical multidrug-resistant strains. Identification of these selective inhibitors indicates that MsbA is a viable target for new antibiotics, and the compounds we identified serve as useful tools to further probe the LPS transport pathway in Gram-negative bacteria.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Lipopolysaccharides/metabolism , Anti-Bacterial Agents/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Escherichia coli/drug effects
6.
J Biol Chem ; 289(2): 942-55, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24225950

ABSTRACT

PCSK9 (proprotein convertase subtilisin/kexin type 9) is a negative regulator of the hepatic LDL receptor, and clinical studies with PCSK9-inhibiting antibodies have demonstrated strong LDL-c-lowering effects. Here we screened phage-displayed peptide libraries and identified the 13-amino acid linear peptide Pep2-8 as the smallest PCSK9 inhibitor with a clearly defined mechanism of inhibition that has been described. Pep2-8 bound to PCSK9 with a KD of 0.7 Āµm but did not bind to other proprotein convertases. It fully restored LDL receptor surface levels and LDL particle uptake in PCSK9-treated HepG2 cells. The crystal structure of Pep2-8 bound to C-terminally truncated PCSK9 at 1.85 ƅ resolution showed that the peptide adopted a strand-turn-helix conformation, which is remarkably similar to its solution structure determined by NMR. Consistent with the functional binding site identified by an Ala scan of PCSK9, the structural Pep2-8 contact region of about 400 ƅ(2) largely overlapped with that contacted by the EGF(A) domain of the LDL receptor, suggesting a competitive inhibition mechanism. Consistent with this, Pep2-8 inhibited LDL receptor and EGF(A) domain binding to PCSK9 with IC50 values of 0.8 and 0.4 Āµm, respectively. Remarkably, Pep2-8 mimicked secondary structural elements of the EGF(A) domain that interact with PCSK9, notably the Ɵ-strand and a discontinuous short α-helix, and it engaged in the same Ɵ-sheet hydrogen bonds as EGF(A) does. Although Pep2-8 itself may not be amenable to therapeutic applications, this study demonstrates the feasibility of developing peptidic inhibitors to functionally relevant sites on PCSK9.


Subject(s)
Oligopeptides/pharmacology , Proprotein Convertases/metabolism , Receptors, LDL/metabolism , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive/drug effects , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Hep G2 Cells , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptide Library , Proprotein Convertase 9 , Proprotein Convertases/chemistry , Proprotein Convertases/genetics , Protein Binding/drug effects , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, LDL/chemistry , Receptors, LDL/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics
7.
Bioorg Med Chem Lett ; 25(3): 529-41, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25556090

ABSTRACT

Herein we report the optimization efforts to ameliorate the potent CYP3A4 time-dependent inhibition (TDI) and low aqueous solubility exhibited by a previously identified lead compound from our NAMPT inhibitor program (1, GNE-617). Metabolite identification studies pinpointed the imidazopyridine moiety present in 1 as the likely source of the TDI signal, and replacement with other bicyclic systems was found to reduce or eliminate the TDI finding. A strategy of reducing the number of aromatic rings and/or lowering cLogD7.4 was then employed to significantly improve aqueous solubility. These efforts culminated in the discovery of 42, a compound with no evidence of TDI, improved aqueous solubility, and robust efficacy in tumor xenograft studies.


Subject(s)
Cytochrome P-450 CYP3A/chemistry , Enzyme Inhibitors/chemistry , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Animals , Binding Sites , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Crystallography, X-Ray , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/chemistry , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/toxicity , Dogs , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Female , Half-Life , Humans , Kinetics , Madin Darby Canine Kidney Cells , Mice , Mice, Nude , Molecular Dynamics Simulation , Neoplasms/drug therapy , Neoplasms/pathology , Nicotinamide Phosphoribosyltransferase/metabolism , Protein Binding , Protein Structure, Tertiary , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Pyrimidines/toxicity , Solubility , Structure-Activity Relationship , Thermodynamics , Transplantation, Heterologous , Water/chemistry
8.
Proc Natl Acad Sci U S A ; 109(14): 5299-304, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22431598

ABSTRACT

The Ras gene is frequently mutated in cancer, and mutant Ras drives tumorigenesis. Although Ras is a central oncogene, small molecules that bind to Ras in a well-defined manner and exert inhibitory effects have not been uncovered to date. Through an NMR-based fragment screen, we identified a group of small molecules that all bind to a common site on Ras. High-resolution cocrystal structures delineated a unique ligand-binding pocket on the Ras protein that is adjacent to the switch I/II regions and can be expanded upon compound binding. Structure analysis predicts that compound-binding interferes with the Ras/SOS interactions. Indeed, selected compounds inhibit SOS-mediated nucleotide exchange and prevent Ras activation by blocking the formation of intermediates of the exchange reaction. The discovery of a small-molecule binding pocket on Ras with functional significance provides a new direction in the search of therapeutically effective inhibitors of the Ras oncoprotein.


Subject(s)
Nucleotides/metabolism , Son of Sevenless Proteins/metabolism , ras Proteins/metabolism , Binding Sites , Cell Line , Humans , Ligands , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , ras Proteins/chemistry
9.
Proc Natl Acad Sci U S A ; 109(47): 19368-73, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23134728

ABSTRACT

The protein kinase v-akt murine thymoma viral oncogene homolog (AKT), a key regulator of cell survival and proliferation, is frequently hyperactivated in human cancers. Intramolecular pleckstrin homology (PH) domain-kinase domain (KD) interactions are important in maintaining AKT in an inactive state. AKT activation proceeds after a conformational change that dislodges the PH from the KD. To understand these autoinhibitory interactions, we generated mutations at the PH-KD interface and found that most of them lead to constitutive activation of AKT. Such mutations are likely another mechanism by which activation may occur in human cancers and other diseases. In support of this likelihood, we found somatic mutations in AKT1 at the PH-KD interface that have not been previously described in human cancers. Furthermore, we show that the AKT1 somatic mutants are constitutively active, leading to oncogenic signaling. Additionally, our studies show that the AKT1 mutants are not effectively inhibited by allosteric AKT inhibitors, consistent with the requirement for an intact PH-KD interface for allosteric inhibition. These results have important implications for therapeutic intervention in patients with AKT mutations at the PH-KD interface.


Subject(s)
Neoplasms/enzymology , Neoplasms/genetics , Oncogenes/genetics , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Enzyme Activation/drug effects , Humans , Mice , Models, Molecular , Mutant Proteins/metabolism , Mutation/genetics , NIH 3T3 Cells , Protein Binding/drug effects , Protein Binding/genetics , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
10.
Bioorg Med Chem Lett ; 24(3): 954-62, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24433859

ABSTRACT

The fragment-based identification of two novel and potent biochemical inhibitors of the nicotinamide phosphoribosyltransferase (NAMPT) enzyme is described. These compounds (51 and 63) incorporate an amide moiety derived from 3-aminopyridine, and are thus structurally distinct from other known anti-NAMPT agents. Each exhibits potent inhibition of NAMPT biochemical activity (IC50=19 and 15 nM, respectively) as well as robust antiproliferative properties in A2780 cell culture experiments (IC50=121 and 99 nM, respectively). However, additional biological studies indicate that only inhibitor 51 exerts its A2780 cell culture effects via a NAMPT-mediated mechanism. The crystal structures of both 51 and 63 in complex with NAMPT are also independently described.


Subject(s)
Amides/chemical synthesis , Amides/pharmacology , Aminopyridines/chemical synthesis , Cytokines/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Amides/chemistry , Aminopyridines/chemistry , Aminopyridines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Crystallography, X-Ray , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , Models, Molecular , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL