Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Med Chem ; 63(22): 13578-13594, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32910655

ABSTRACT

SHP2 is a nonreceptor protein tyrosine phosphatase encoded by the PTPN11 gene and is involved in cell growth and differentiation via the MAPK signaling pathway. SHP2 also plays an important role in the programed cell death pathway (PD-1/PD-L1). As an oncoprotein as well as a potential immunomodulator, controlling SHP2 activity is of high therapeutic interest. As part of our comprehensive program targeting SHP2, we identified multiple allosteric binding modes of inhibition and optimized numerous chemical scaffolds in parallel. In this drug annotation report, we detail the identification and optimization of the pyrazine class of allosteric SHP2 inhibitors. Structure and property based drug design enabled the identification of protein-ligand interactions, potent cellular inhibition, control of physicochemical, pharmaceutical and selectivity properties, and potent in vivo antitumor activity. These studies culminated in the discovery of TNO155, (3S,4S)-8-(6-amino-5-((2-amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-3-methyl-2-oxa-8-azaspiro[4.5]decan-4-amine (1), a highly potent, selective, orally efficacious, and first-in-class SHP2 inhibitor currently in clinical trials for cancer.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Neoplasms/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Antineoplastic Agents/therapeutic use , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Macaca fascicularis , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Rats , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
2.
IEEE Trans Haptics ; 12(3): 232-246, 2019.
Article in English | MEDLINE | ID: mdl-31352355

ABSTRACT

As haptics have become an ingrained part of our wearable experience, particularly through phones, smartwatches, and fitness trackers, significant research effort has been conducted to find new ways of using wearable haptics to convey information, especially while we are on-the-go. In this paper, instead of focusing on aspects of haptic information design, such as tacton encoding methods, actuators, and technical fabrication of devices, we address the more general recurring issues and "gotchas" that arise when moving from core haptic perceptual studies and in-lab wearable experiments to real world testing of wearable vibrotactile haptic systems. We summarize key issues for practitioners to take into account when designing and carrying out in-the-wild wearable haptic user studies, as well as for user studies in a lab environment that seek to simulate real-world conditions. We include not only examples from published work and commercial sources, but also hard-won illustrative examples derived from issues and failures from our own haptic studies. By providing a broad-based, accessible overview of recurring issues, we expect that both novice and experienced haptic researchers will find suggestions that will improve their own mobile wearable haptic studies.


Subject(s)
Feedback, Sensory , Research Design , Touch Perception , Wearable Electronic Devices , Equipment Design , Humans , Physical Stimulation , Touch , User-Computer Interface , Wireless Technology
3.
ACS Med Chem Lett ; 8(2): 151-156, 2017 Feb 09.
Article in English | MEDLINE | ID: mdl-28197303

ABSTRACT

High throughput screening and subsequent hit validation identified 4-isopropyl-3-(2-((1-phenylethyl)amino)pyrimidin-4-yl)oxazolidin-2-one as a potent inhibitor of IDH1R132H. Synthesis of the four separate stereoisomers identified the (S,S)-diastereomer (IDH125, 1f) as the most potent isomer. This also showed reasonable cellular activity and excellent selectivity vs IDH1wt. Initial structure-activity relationship exploration identified the key tolerances and potential for optimization. X-ray crystallography identified a functionally relevant allosteric binding site amenable to inhibitors, which can penetrate the blood-brain barrier, and aided rational optimization. Potency improvement and modulation of the physicochemical properties identified (S,S)-oxazolidinone IDH889 (5x) with good exposure and 2-HG inhibitory activity in a mutant IDH1 xenograft mouse model.

4.
Structure ; 25(3): 506-513, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28132785

ABSTRACT

Oncogenic IDH1 and IDH2 mutations contribute to cancer via production of R-2-hydroxyglutarate (2-HG). Here, we characterize two structurally distinct mutant- and isoform-selective IDH1 inhibitors that inhibit 2-HG production. Both bind to an allosteric pocket on IDH1, yet shape it differently, highlighting the plasticity of this site. Oncogenic IDH1R132H mutation destabilizes an IDH1 "regulatory segment," which otherwise restricts compound access to the allosteric pocket. Regulatory segment destabilization in wild-type IDH1 promotes inhibitor binding, suggesting that destabilization is critical for mutant selectivity. We also report crystal structures of oncogenic IDH2 mutant isoforms, highlighting the fact that the analogous segment of IDH2 is not similarly destabilized. This intrinsic stability of IDH2 may contribute to observed inhibitor IDH1 isoform selectivity. Moreover, discrete residues in the IDH1 allosteric pocket that differ from IDH2 may also guide IDH1 isoform selectivity. These data provide a deeper understanding of how IDH1 inhibitors achieve mutant and isoform selectivity.


Subject(s)
Enzyme Inhibitors/pharmacology , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/genetics , Neoplasms/genetics , Small Molecule Libraries/pharmacology , Allosteric Regulation , Allosteric Site , Crystallography, X-Ray , Glutarates/metabolism , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Protein Binding , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/genetics
5.
PLoS One ; 11(9): e0162107, 2016.
Article in English | MEDLINE | ID: mdl-27603211

ABSTRACT

BACKGROUND: Our daily activities imply displacements on various types of soil. For persons with gait disorder or losing functional autonomy, walking on some types of soil could be challenging because of the risk of falling it represents. METHODS: In this paper, we present, in a first part, the use of an enactive shoe for an automatic differentiation of several types of soil. In a second part, using a second improved prototype (an enactive insole), twelve participants with Parkinson's disease (PD) and nine age-matched controls have performed the Timed Up and Go (TUG) test on six types of soil with and without cueing. The frequency of the cueing was set at 10% above the cadence computed at the lower risk of falling (walking over the concrete). Depending on the cadence computed at the lower risk, the enactive insole activates a vibrotactile cueing aiming to improve gait and balance control. Finally, a risk index is computed using gait parameters in relation to given type of soil. RESULTS: The frequency analysis of the heel strike vibration allows the differentiation of various types of soil. The risk computed is associated to an appropriate rhythmic cueing in order to improve balance and gait impairment. The results show that a vibrotactile cueing could help to reduce the risk of falling. CONCLUSIONS: Firstly, this paper demonstrates the feasibility of reducing the risk of falling while walking on different types of soil using vibrotactile cueing. We found a significant difference and a significant decrease in the computed risks of falling for most of types of soil especially for deformable soils which can lead to fall. Secondly, heel strike provides an approximation of the impulse response of the soil that can be analyzed with time and frequency-domain modeling. From these analyses, an index is computed enabling differentiation the types of soil.


Subject(s)
Accidental Falls/prevention & control , Cues , Foot Orthoses , Risk , Soil , Touch/physiology , Vibration , Acceleration , Aged , Case-Control Studies , Demography , Electronics , Female , Fourier Analysis , Gait/physiology , Humans , Male , Middle Aged , Parkinson Disease/physiopathology , Signal Processing, Computer-Assisted , Walking/physiology
6.
J Med Chem ; 59(17): 7773-82, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27347692

ABSTRACT

SHP2 is a nonreceptor protein tyrosine phosphatase (PTP) encoded by the PTPN11 gene involved in cell growth and differentiation via the MAPK signaling pathway. SHP2 also purportedly plays an important role in the programmed cell death pathway (PD-1/PD-L1). Because it is an oncoprotein associated with multiple cancer-related diseases, as well as a potential immunomodulator, controlling SHP2 activity is of significant therapeutic interest. Recently in our laboratories, a small molecule inhibitor of SHP2 was identified as an allosteric modulator that stabilizes the autoinhibited conformation of SHP2. A high throughput screen was performed to identify progressable chemical matter, and X-ray crystallography revealed the location of binding in a previously undisclosed allosteric binding pocket. Structure-based drug design was employed to optimize for SHP2 inhibition, and several new protein-ligand interactions were characterized. These studies culminated in the discovery of 6-(4-amino-4-methylpiperidin-1-yl)-3-(2,3-dichlorophenyl)pyrazin-2-amine (SHP099, 1), a potent, selective, orally bioavailable, and efficacious SHP2 inhibitor.


Subject(s)
Antineoplastic Agents/chemistry , Piperidines/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Pyrazines/chemistry , Pyrimidines/chemistry , Administration, Oral , Allosteric Regulation , Allosteric Site , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Female , Heterografts , High-Throughput Screening Assays , Humans , Male , Mice, Inbred C57BL , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Piperidines/chemical synthesis , Piperidines/pharmacokinetics , Piperidines/pharmacology , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Pyrazines/chemical synthesis , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Structure-Activity Relationship
7.
Biochemistry ; 55(15): 2269-77, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27030275

ABSTRACT

The proto-oncogene PTPN11 encodes a cytoplasmic protein tyrosine phosphatase, SHP2, which is required for normal development and sustained activation of the Ras-MAPK signaling pathway. Germline mutations in SHP2 cause developmental disorders, and somatic mutations have been identified in childhood and adult cancers and drive leukemia in mice. Despite our knowledge of the PTPN11 variations associated with pathology, the structural and functional consequences of many disease-associated mutants remain poorly understood. Here, we combine X-ray crystallography, small-angle X-ray scattering, and biochemistry to elucidate structural and mechanistic features of three cancer-associated SHP2 variants harboring single point mutations within the N-SH2:PTP interdomain autoinhibitory interface. Our findings directly compare the impact of each mutation on autoinhibition of the phosphatase and advance the development of structure-guided and mutation-specific SHP2 therapies.


Subject(s)
Neoplasms/genetics , Point Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Amino Acid Substitution/genetics , Cell Transformation, Neoplastic/genetics , Crystallography, X-Ray , Enzyme Activation/genetics , Humans , Leukemia/genetics , Ligands , Models, Molecular , Oncogenes/genetics , Protein Structure, Tertiary/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Mas , Scattering, Small Angle , Structure-Activity Relationship
8.
Chem Biol ; 22(1): 87-97, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25544045

ABSTRACT

The highly conserved 70 kDa heat shock proteins (Hsp70) play an integral role in proteostasis such that dysregulation has been implicated in numerous diseases. Elucidating the precise role of Hsp70 family members in the cellular context, however, has been hampered by the redundancy and intricate regulation of the chaperone network, and relatively few selective and potent tools. We have characterized a natural product, novolactone, that targets cytosolic and ER-localized isoforms of Hsp70 through a highly conserved covalent interaction at the interface between the substrate-binding and ATPase domains. Biochemical and structural analyses indicate that novolactone disrupts interdomain communication by allosterically inducing a conformational change in the Hsp70 protein to block ATP-induced substrate release and inhibit refolding activities. Thus, novolactone is a valuable tool for exploring the requirements of Hsp70 chaperones in diverse cellular contexts.


Subject(s)
Abietanes/metabolism , Biological Products/metabolism , HSP70 Heat-Shock Proteins/metabolism , Abietanes/chemistry , Adenosine Triphosphatases/metabolism , Allosteric Regulation , Binding Sites , Biological Products/chemistry , Cell Line , Crystallography, X-Ray , Endoplasmic Reticulum/metabolism , Genome, Fungal , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , Humans , Molecular Dynamics Simulation , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Substrate Specificity
9.
Biochemistry ; 52(39): 6866-78, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24000826

ABSTRACT

Protein lysine methyltransferases (PKMTs) are key players in epigenetic regulation and have been associated with a variety of diseases, including cancers. The catalytic subunit of Polycomb Repressive Complex 2, EZH2 (EC 2.1.1.43), is a PKMT and a member of a family of SET domain lysine methyltransferases that catalyze the transfer of a methyl group from S-adenosyl-l-methionine to lysine 27 of histone 3 (H3K27). Wild-type (WT) EZH2 primarily catalyzes the mono- and dimethylation of H3K27; however, a clinically relevant active site mutation (Y641F) has been shown to alter the reaction specificity, dominantly catalyzing trimethylation of H3K27, and has been linked to tumor genesis and maintenance. Herein, we explore the chemical mechanism of methyl transfer by EZH2 and its Y641F mutant with pH-rate profiles and solvent kinetic isotope effects (sKIEs) using a short peptide derived from histone H3 [H3(21-44)]. A key component of the chemical reaction is the essential deprotonation of the ε-NH3(+) group of lysine to accommodate subsequent methylation. This deprotonation has been suggested by independent studies (1) to occur prior to binding to the enzyme (by bulk solvent) or (2) to be facilitated within the active site following binding, either (a) by the enzyme itself or (b) by a water molecule with access to the binding pocket. Our pH-rate and sKIE data best support a model in which lysine deprotonation is enzyme-dependent and at least partially rate-limiting. Furthermore, our experimental data are in agreement with prior computational models involving enzyme-dependent solvent deprotonation through a channel providing bulk solvent access to the active site. The mechanism of deprotonation and the rate-limiting catalytic steps appear to be unchanged between the WT and Y641F mutant enzymes, despite their activities being highly dependent on different substrate methylation states, suggesting determinants of substrate and product specificity in EZH2 are independent of catalytic events limiting the steady-state rate.


Subject(s)
Lysine/metabolism , Polycomb Repressive Complex 2/metabolism , Protons , Biocatalysis , Hydrogen-Ion Concentration , Lysine/chemistry , Models, Molecular , Molecular Structure , Mutation , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/genetics
10.
J Med Chem ; 56(17): 7049-59, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23879431

ABSTRACT

Tankyrases 1 and 2 are members of the poly(ADP-ribose) polymerase (PARP) family of enzymes that modulate Wnt pathway signaling. While amide- and lactam-based nicotinamide mimetics that inhibit tankyrase activity, such as XAV939, are well-known, herein we report the discovery and evaluation of a novel nicotinamide isostere that demonstrates selectivity over other PARP family members. We demonstrate the utilization of lipophilic efficiency-based structure-efficiency relationships (SER) to rapidly drive the evaluation of this series. These efforts led to a series of selective, cell-active compounds with solubility, physicochemical, and in vitro properties suitable for further optimization.


Subject(s)
Amines/pharmacology , Tankyrases/antagonists & inhibitors , Triazoles/pharmacology , Amines/chemistry , Animals , Enzyme-Linked Immunosorbent Assay , Male , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Triazoles/chemistry
11.
J Med Chem ; 56(16): 6495-511, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23844574

ABSTRACT

Tankyrase 1 and 2 have been shown to be redundant, druggable nodes in the Wnt pathway. As such, there has been intense interest in developing agents suitable for modulating the Wnt pathway in vivo by targeting this enzyme pair. By utilizing a combination of structure-based design and LipE-based structure efficiency relationships, the core of XAV939 was optimized into a more stable, more efficient, but less potent dihydropyran motif 7. This core was combined with elements of screening hits 2, 19, and 33 and resulted in highly potent, selective tankyrase inhibitors that are novel three pocket binders. NVP-TNKS656 (43) was identified as an orally active antagonist of Wnt pathway activity in the MMTV-Wnt1 mouse xenograft model. With an enthalpy-driven thermodynamic signature of binding, highly favorable physicochemical properties, and high lipophilic efficiency, NVP-TNKS656 is a novel tankyrase inhibitor that is well suited for further in vivo validation studies.


Subject(s)
Acetamides/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Pyrimidinones/pharmacology , Tankyrases/antagonists & inhibitors , Acetamides/administration & dosage , Acetamides/chemistry , Administration, Oral , Animals , Area Under Curve , Biological Availability , Enzyme Inhibitors/administration & dosage , Mice , Models, Molecular , Pyrimidinones/administration & dosage , Pyrimidinones/chemistry , Structure-Activity Relationship
12.
J Med Chem ; 55(3): 1127-36, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22260203

ABSTRACT

The Wnt signaling pathway is critical to the regulation of key cellular processes. When deregulated, it has been shown to play a crucial role in the growth and progression of multiple human cancers. The identification of small molecule modulators of Wnt signaling has proven challenging, largely due to the relative paucity of druggable nodes in this pathway. Several recent publications have identified small molecule inhibitors of the Wnt pathway, and tankyrase (TNKS) inhibition has been demonstrated to antagonize Wnt signaling via axin stabilization. Herein, we report the early hit assessment of a series of compounds previously reported to antagonize Wnt signaling. We report the biophysical, computational characterization, structure-activity relationship, and physicochemical properties of a novel series of [1,2,4]triazol-3-ylsulfanylmethyl)-3-phenyl-[1,2,4]oxadiazole inhibitors of TNKS1 and 2. Furthermore, a cocrystal structure of compound 24 complexed to TNKS1 demonstrates an alternate binding mode for PARP family member proteins that does not involve interactions with the nicotinamide binding pocket.


Subject(s)
Adenosine/metabolism , Models, Molecular , Oxadiazoles/chemical synthesis , Sulfides/chemical synthesis , Tankyrases/antagonists & inhibitors , Triazoles/chemical synthesis , Wnt Signaling Pathway/drug effects , Adenosine/chemistry , Binding Sites , Crystallography, X-Ray , HEK293 Cells , Humans , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Protein Conformation , Structure-Activity Relationship , Sulfides/chemistry , Sulfides/pharmacology , Triazoles/chemistry , Triazoles/pharmacology
13.
Biochemistry ; 47(43): 11310-20, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18826255

ABSTRACT

The pseudomonal phytotoxin syringomycin E and related nonribosomal peptides contain an L- threo-beta-hydroxyaspartyl residue at the eighth position of the lipodepsipeptide backbone as part of a conserved nonproteinogenic tripeptide motif. Informatic analysis of the P. syringae genome suggests only one putative non-heme iron hydroxylase, AspH. On heterologous expression in Escherichia coli AspH shows robust catalytic activity with free L-Asp and L-Asp thioesters to make beta-OH-Asp but yields the erythro diastereomer rather than the threo configuration that is found in syringomycin. Further analysis of the Syr gene cluster indicated that SyrP, previously annotated as the gene regulatory protein for the five-gene Syr cluster, is actually homologous to the known non-heme mononuclear iron hydroxylase TauD. Indeed, purified SyrP acts on Asp tethered as the protein-bound S-pantetheinyl thioester on the eighth module of the SyrE megasynthetase. The hydroxylation gives the anticipated L- threo-3-OH-Asp diastereomer found in syringomycin. The knockout of syrP abolishes the production of the mature syringomycin E, while knockout of aspH has no effect on syringomycin production.


Subject(s)
Mixed Function Oxygenases/genetics , Peptides, Cyclic/genetics , Pseudomonas syringae/genetics , Toxins, Biological/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Hydroxylation , Mixed Function Oxygenases/metabolism , Peptides, Cyclic/metabolism , Pseudomonas syringae/metabolism , Toxins, Biological/metabolism
14.
Proc Natl Acad Sci U S A ; 105(36): 13321-6, 2008 Sep 09.
Article in English | MEDLINE | ID: mdl-18768797

ABSTRACT

Andrimid is a hybrid nonribosomal peptide-polyketide antibiotic that blocks the carboxyl-transfer reaction of bacterial acetyl-CoA carboxylase (ACC) and thereby inhibits fatty acid biosynthesis with submicromolar potency. The andrimid biosynthetic gene cluster from Pantoea agglomerans encodes an admT gene with homology to the acetyl-CoA carboxyltransferase (CT) beta-subunit gene accD. Escherichia coli cells overexpressing admT showed resistance to andrimid. Co-overproduction of AdmT with E. coli CT alpha-subunit AccA allowed for the in vitro reconstitution of an active heterologous tetrameric CT A(2)T(2) complex. A subsequent andrimid-inhibition assay revealed an IC(50) of 500 nM for this hybrid A(2)T(2) in contrast to that of 12 nM for E. coli CT A(2)D(2). These results validated that AdmT is an AccD homolog that confers resistance in the andrimid producer. Mutagenesis studies guided by the x-ray crystal structure of the E. coli A(2)D(2) complex disclosed a single amino acid mutation of AdmT (L203M) responsible for 5-fold andrimid sensitivity (IC(50) = 100 nM). Complementarily, the E. coli AccD mutant M203L became 5-fold more resistant in the CT assays. This observation allowed for bioinformatic identification of several Vibrio cholerae strains in which accD genes encode the Met<-->Leu switches, and their occurrences correlate predictively with sensitivities to andrimid in vivo.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/metabolism , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Acetyl-CoA Carboxylase/genetics , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Computational Biology , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Polyenes/chemistry , Polyenes/metabolism , Polyenes/pharmacology , Protein Structure, Tertiary , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Pyrroles/chemistry , Pyrroles/metabolism , Pyrroles/pharmacology , Sequence Alignment , Sequence Homology, Amino Acid , Vibrio cholerae/drug effects
15.
ACS Chem Biol ; 3(9): 542-54, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18652473

ABSTRACT

The antibiotic andrimid, a nanomolar inhibitor of bacterial acetyl coenzyme A carboxylase, is generated on an unusual polyketide/nonribosomal peptide enzyme assembly line in that all thiolation (T) domains/small-molecule building stations are on separate proteins. In addition, a transglutaminase homologue is used to condense andrimid building blocks together on the andrimid assembly line. The first two modules of the andrimid assembly line yields an octatrienoyl-beta-Phe-thioester tethered to the AdmI T domain, with amide bond formation carried out by a free-standing transglutaminase homologue AdmF. Analysis of the aminomutase AdmH reveals its specific conversion from l-Phe to (S)-beta-Phe, which in turn is activated by AdmJ and ATP to form (S)-beta-Phe-aminoacyl-AMP. AdmJ then transfers the (S)-beta-Phe moiety to one of the free-standing T domains, AdmI, but not AdmA, which instead gets loaded with an octatrienoyl group by other enzymes. AdmF, the amide synthase, will accept a variety of acyl groups in place of the octatrienoyl donor if presented on either AdmA or AdmI. AdmF will also use either stereoisomer of phenylalanine or beta-Phe when presented on AdmA and AdmI, but not when placed on noncognate T domains. Further, we show the polyketide synthase proteins responsible for the polyunsaturated acyl cap can be bypassed in vitro with N-acetylcysteamine as a low-molecular-weight acyl donor to AdmF and also in vivo in an Escherichia coli strain bearing the andrimid biosynthetic gene cluster with a knockout in admA.


Subject(s)
Enzyme Inhibitors/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Amides/metabolism , Aminoacylation , Phenylalanine/metabolism , Polyenes/metabolism , Pyrroles/metabolism , Succinimides/metabolism , Transglutaminases/metabolism
16.
Nature ; 448(7155): 824-7, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-17653193

ABSTRACT

The unrelenting emergence of antibiotic-resistant bacterial pathogens demands the investigation of antibiotics with new modes of action. The pseudopeptide antibiotic andrimid is a nanomolar inhibitor of the bacterial acetyl-CoA carboxylase that catalyses the first committed step in prokaryotic fatty acid biosynthesis. Recently, the andrimid (adm) biosynthetic gene cluster was isolated and heterologously expressed in Escherichia coli. This establishes a heterologous biological host in which to rapidly probe features of andrimid formation and to use biosynthetic engineering to make unnatural variants of this important and promising new class of antibiotics. Bioinformatic analysis of the adm cluster revealed a dissociated biosynthetic assembly system lacking canonical amide synthases between the first three carrier protein domains. Here we report that AdmF, a transglutaminase (TGase) homologue, catalyses the formation of the first amide bond, an N-acyl-beta-peptide link, in andrimid biosynthesis. Hence, AdmF is a newly discovered biosynthetic enzyme that acts as a stand-alone amide synthase between protein-bound, thiotemplated substrates in an antibiotic enzymatic assembly line. TGases (enzyme class (EC) 2.3.2.13) normally catalyse the cross-linking of (poly)peptides by creating isopeptidic bonds between the gamma-carboxamide group of a glutamine side chain of one protein and various amine donors, including lysine side chains. To the best of our knowledge, the present study constitutes the first report of a TGase-like enzyme recruited for the assembly of an antibiotic. Moreover, genome mining using the AdmF sequence yielded additional TGases in unassigned natural product biosynthetic pathways. With many more microbial genomes being sequenced, such a strategy could potentially unearth biosynthetic pathways producing new classes of antibiotics.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Bacteria/enzymology , Transglutaminases/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/classification , Bacteria/genetics , Bacteria/metabolism , Biological Products/biosynthesis , Biological Products/chemistry , Catalysis , Escherichia coli , Genes, Bacterial/genetics , Multigene Family/genetics , Phenylalanine/chemistry , Phenylalanine/metabolism , Polyenes/chemistry , Polyenes/metabolism , Protein Structure, Tertiary , Pyrroles/chemistry , Pyrroles/metabolism , Transglutaminases/chemistry , Transglutaminases/genetics
17.
J Bacteriol ; 189(11): 4038-45, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17416660

ABSTRACT

Sphingomonas wittichii RW1 degrades chlorinated dibenzofurans and dibenzo-p-dioxins via meta cleavage. We used inverse PCR to amplify dxnB2, a gene encoding one of three meta-cleavage product (MCP) hydrolases identified in the organism that are homologues of BphD involved in biphenyl catabolism. Purified DxnB2 catalyzed the hydrolysis of 8-OH 2-hydroxy-6-oxo-6-phenylhexa-2,4-dienoate (HOPDA) approximately six times faster than for HOPDA at saturating substrate concentrations. Moreover, the specificity of DxnB2 for HOPDA (k(cat)/K(m) = 1.2 x 10(7) M(-1) s(-1)) was about half that of the BphDs of Burkholderia xenovorans LB400 and Rhodococcus globerulus P6, two potent polychlorinated biphenyl (PCB)-degrading strains. Interestingly, DxnB2 transformed 3-Cl and 4-OH HOPDAs, compounds that inhibit the BphDs and limit PCB degradation. DxnB2 had a higher specificity for 9-Cl HOPDA than for HOPDA but a lower specificity for 8-Cl HOPDA (k(cat)/K(m) = 1.7 x 10(6) M(-1) s(-1)), the chlorinated analog of 8-OH HOPDA produced during dibenzofuran catabolism. Phylogenetic analyses based on structure-guided sequence alignment revealed that DxnB2 belongs to a previously unrecognized class of MCP hydrolases, evolutionarily divergent from the BphDs although the physiological substrates of both enzyme types are HOPDAs. However, both classes of enzymes have mainly small hydrophobic residues lining the subsite that binds the C-6 phenyl of HOPDA, in contrast to the bulky hydrophobic residues (Phe106, Phe135, Trp150, and Phe197) found in the class II enzymes that prefer substrates possessing a C-6 alkyl. Thr196 and/or Asn203 appears to be an important determinant of specificity for DxnB2, potentially forming hydrogen bonds with the 8-OH substituent. This study demonstrates that the substrate specificities of evolutionarily divergent hydrolases may be useful for degrading mixtures of pollutants, such as PCBs.


Subject(s)
Bacterial Proteins/metabolism , Benzofurans/metabolism , Hydrolases/metabolism , Sphingomonas/enzymology , Amino Acid Sequence , Bacterial Proteins/genetics , Benzofurans/chemistry , Cloning, Molecular , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Dibenzofurans, Polychlorinated , Dioxins/chemistry , Dioxins/metabolism , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/metabolism , Hydrolases/genetics , Kinetics , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sphingomonas/genetics , Sphingomonas/metabolism , Substrate Specificity
19.
J Biol Chem ; 281(41): 30933-40, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16920719

ABSTRACT

Prokaryotic glutathione S-transferases are as diverse as their eukaryotic counterparts but are much less well characterized. BphK from Burkholderia xenovorans LB400 consumes two GSH molecules to reductively dehalogenate chlorinated 2-hydroxy-6-oxo-6-phenyl-2,4-dienoates (HOPDAs), inhibitory polychlorinated biphenyl metabolites. Crystallographic structures of two ternary complexes of BphK were solved to a resolution of 2.1A. In the BphK-GSH-HOPDA complex, GSH and HOPDA molecules occupy the G- and H-subsites, respectively. The thiol nucleophile of the GSH molecule is positioned for SN2 attack at carbon 3 of the bound HOPDA. The respective sulfur atoms of conserved Cys-10 and the bound GSH are within 3.0A, consistent with product release and the formation of a mixed disulfide intermediate. In the BphK-(GSH)2 complex, a GSH molecule occupies each of the two subsites. The three sulfur atoms of the two GSH molecules and Cys-10 are aligned suitably for a disulfide exchange reaction that would regenerate the resting enzyme and yield disulfide-linked GSH molecules. A second conserved residue, His-106, is adjacent to the thiols of Cys-10 and the GSH bound to the G-subsite and thus may stabilize a transition state in the disulfide exchange reaction. Overall, the structures support and elaborate a proposed dehalogenation mechanism for BphK and provide insight into the plasticity of the H-subsite.


Subject(s)
Burkholderia/enzymology , Glutathione Transferase/chemistry , Binding Sites , Crystallography, X-Ray , Disulfides/chemistry , Escherichia coli/metabolism , Fatty Acids, Unsaturated/chemistry , Glutathione/chemistry , Polychlorinated Biphenyls/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary
20.
J Bacteriol ; 188(12): 4424-30, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16740949

ABSTRACT

BphK is a glutathione S-transferase of unclear physiological function that occurs in some bacterial biphenyl catabolic (bph) pathways. We demonstrated that BphK of Burkholderia xenovorans strain LB400 catalyzes the dehalogenation of 3-chloro 2-hydroxy-6-oxo-6-phenyl-2,4-dienoates (HOPDAs), compounds that are produced by the cometabolism of polychlorinated biphenyls (PCBs) by the bph pathway and that inhibit the pathway's hydrolase. A one-column protocol was developed to purify heterologously produced BphK. The purified enzyme had the greatest specificity for 3-Cl HOPDA (kcat/Km, approximately 10(4) M(-1) s(-1)), which it dechlorinated approximately 3 orders of magnitude more efficiently than 4-chlorobenzoate, a previously proposed substrate of BphK. The enzyme also catalyzed the dechlorination of 5-Cl HOPDA and 3,9,11-triCl HOPDA. By contrast, BphK did not detectably transform HOPDA, 4-Cl HOPDA, or chlorinated 2,3-dihydroxybiphenyls. The BphK-catalyzed dehalogenation proceeded via a ternary-complex mechanism and consumed 2 equivalents of glutathione (GSH) (Km for GSH in the presence of 3-Cl HOPDA, approximately 0.1 mM). A reaction mechanism consistent with the enzyme's specificity is proposed. The ability of BphK to dehalogenate inhibitory PCB metabolites supports the hypothesis that this enzyme was recruited to facilitate PCB degradation by the bph pathway.


Subject(s)
Burkholderia/enzymology , Glutathione Transferase/metabolism , Polychlorinated Biphenyls/metabolism , Diltiazem/analogs & derivatives , Fatty Acids, Unsaturated/metabolism , Glutathione Transferase/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...