Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Curr Issues Mol Biol ; 45(10): 8138-8151, 2023 Oct 07.
Article in English | MEDLINE | ID: mdl-37886957

ABSTRACT

EGFR tyrosine kinase inhibitors (TKIs) are the first-line treatment for advanced EGFR-mutated non-small-cell lung cancer (NSCLC). However, NSCLC patients with wild-type EGFR and KRAS mutation are ineligible for EGFR-TKIs. Therefore, the discovery of new therapeutic agents is urgently needed for NSCLC patients who cannot receive targeted therapies. Natural products possess tremendous chemical diversity and have been extensively investigated for their anticancer activity. In this study, we found that Cucurbitacin E (Cu E), a triterpene of cucurbitacins widely presented in the edible plants of the Cucurbitaceae family, significantly inhibits the viability and proliferation of A549 cells that harbor wild-type EGFR and KRAS mutation. Our results revealed that Cu E increases cell-cycle arrest at G2/M and subG1 phase. Mechanistically, Cu E significantly inhibits the phosphorylation and protein levels of regulatory proteins and hinders G2/M cell-cycle progression. Meanwhile, the treatment of Cu E resulted in DNA damage response and apoptosis. For the first time, we observed that Cu E induces incomplete autophagy as evidenced by increased LC3B-II expression and p62-accumulation. Knockdown of p62 rescued the cells from Cu E-mediated anti-proliferative effect, apoptosis, DNA damage, and ROS production. These findings suggest that Cu E is a promising drug candidate for NSCLC.

2.
Mar Drugs ; 21(2)2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36827114

ABSTRACT

Bladder cancer (BC) is one of the most prevalent cancers worldwide. However, the recurrence rate and five-year survival rate have not been significantly improved in advanced BC, and new therapeutic strategies are urgently needed. The anticancer activity of stellettin B (SP-2), a triterpene isolated from the marine sponge Rhabdastrella sp., was evaluated with the MTT assay as well as PI and Annexin V/7-AAD staining. Detailed mechanisms were elucidated through an NGS analysis, protein arrays, and Western blotting. SP-2 suppressed the viability of BC cells without severe toxicity towards normal uroepithelial cells, and it increased apoptosis with the activation of caspase 3/8/9, PARP, and γH2AX. The phosphorylation of FGFR3 and its downstream targets were downregulated by SP-2. Meanwhile, it induced autophagy in BC cells as evidenced by LC3-II formation and p62 downregulation. The inhibition of autophagy using pharmacological inhibitors or through an ATG5-knockout protected RT-112 cells from SP-2-induced cell viability suppression and apoptosis. In addition, the upregulation of DAPK2 mRNA and protein expression also contributed to SP-2-induced cytotoxicity and apoptosis. In RT-112 cells, an FGFR3-TACC3-knockout caused the downregulation of DAPK2, autophagy, and apoptosis. In conclusion, this is the first study demonstrating that SP-2 exhibits potent anti-BC activity by suppressing the FGFR3-TACC3/Akt/mTOR pathway, which further activates a novel autophagy/DAPK2/apoptosis signaling cascade.


Subject(s)
Porifera , Triterpenes , Urinary Bladder Neoplasms , Animals , Death-Associated Protein Kinases , Apoptosis , Triterpenes/pharmacology , Urinary Bladder Neoplasms/drug therapy , Autophagy , Porifera/metabolism , Cell Line, Tumor , Proto-Oncogene Proteins c-akt/metabolism
3.
J Nat Prod ; 84(2): 259-267, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33459007

ABSTRACT

Seven new polyhydroxylated oleanane-type triterpene saponins, arenarosides A-G (1-7), together with four known compounds, were isolated from an ethanol extract of the aerial parts of the Vietnamese plant Polycarpaea arenaria. The chemical structures of the newly isolated oleanane saponins were elucidated on the basis of spectroscopic and spectrometric analysis, especially 2D NMR and HRMS. Biological evaluation revealed that 3, 4, 6, and 7 showed moderate activities against four human cancer cell lines (A549, HTC116, PC3, and RT112) with IC50 values of 6.0-9.9 µM, and 3, 4, 5, and 7 also displayed promising antiangiogenesis effects with IC50 values <5 µM in the test system used. Among the isolates, arenaroside D (4) exhibited the most potent inhibitory effects, not only in cancer cell proliferation but also in angiogenic activities. Preliminary SAR studies revealed that the presence of an acetyl group at C-22 in oleanane-type triterpene saponins increases these bioactivities.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Caryophyllaceae/chemistry , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , Angiogenesis Inhibitors/isolation & purification , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Line, Tumor , Humans , Oleanolic Acid/isolation & purification , Oleanolic Acid/pharmacology , Phytochemicals/isolation & purification , Phytochemicals/pharmacology , Plant Components, Aerial/chemistry , Saponins/isolation & purification , Vietnam
4.
Molecules ; 25(3)2020 Jan 28.
Article in English | MEDLINE | ID: mdl-32012805

ABSTRACT

In folk medicine, Stahlianthus thorelii Gagnep. has been used to treat diseases related to inflammation, ulcers, and cancer. There are no reports concerning the chemical components and bioactivities of S. thorelii; thus, this study aims to explore the phytochemicals, quantify the main compounds, and test the anticancer activity of isolates from S. thorelii. Dried rhizomes were extracted with 95% ethanol and, then, partitioned, fractionated, and isolated. On the basis of the result of the antiproliferative activity of the fractions, seven isolates were yielded and were identified by spectroscopic analyses. The inhibition of cancer proliferation was determined by an MTT assay and the deployed IC50 to value their efficacy. Seven compounds containing one new C-benzylated dihydrochalcone derivative, thorechalcone A (1) and 2-7 were isolated from S. thorelii. In terms of the bioactivity, compounds 1 and 3 displayed promising antiproliferative activity (WiDr, A549, and HepG2), with IC50 values <40 µM. The HPLC-UV method of quantification of two major compounds (3 and 4) was also validated. This study presented the isolations of antiproliferative potentials of new chalcone and known flavonoid derivatives from S. thorelii. The validated simple, accurate, and rapid HPLC method could be deployed for the quality control of herbal drugs.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Chalcones/isolation & purification , Flavonoids/isolation & purification , Zingiberaceae/chemistry , A549 Cells , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chalcones/chemistry , Chalcones/pharmacology , Chromatography, High Pressure Liquid , Flavonoids/chemistry , Flavonoids/pharmacology , Hep G2 Cells , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Molecular Structure , Phytochemicals/isolation & purification , Phytochemicals/pharmacology
5.
J Clin Invest ; 130(2): 699-714, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31845908

ABSTRACT

HSP27 is highly expressed in, and supports oncogene addiction of, many cancers. HSP27 phosphorylation is a limiting step for activation of this protein and a target for inhibition, but its highly disordered structure challenges rational structure-guided drug discovery. We performed multistep biochemical, structural, and computational experiments to define a spherical 24-monomer complex composed of 12 HSP27 dimers with a phosphorylation pocket flanked by serine residues between their N-terminal domains. Ivermectin directly binds this pocket to inhibit MAPKAP2-mediated HSP27 phosphorylation and depolymerization, thereby blocking HSP27-regulated survival signaling and client-oncoprotein interactions. Ivermectin potentiated activity of anti-androgen receptor and anti-EGFR drugs in prostate and EGFR/HER2-driven tumor models, respectively, identifying a repurposing approach for cotargeting stress-adaptive responses to overcome resistance to inhibitors of oncogenic pathway signaling.


Subject(s)
Heat-Shock Proteins , Ivermectin , Molecular Chaperones , Neoplasms, Experimental , Receptor, ErbB-2 , A549 Cells , Animals , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ivermectin/chemistry , Ivermectin/pharmacology , Mice , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Domains , Protein Multimerization , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
6.
Molecules ; 24(7)2019 Apr 08.
Article in English | MEDLINE | ID: mdl-30965662

ABSTRACT

Six new dammarane-type saponins, gypenosides CP1-6 (16), along with 19 known compounds 7⁻25, were isolated and characterized from the aerial parts of Gynostemma pentaphyllum. Among these compounds, eight dammarane-type saponins, 2, 5, 6, 7, 11, 12, 13, and 15, exhibited the greatest antiproliferative effects against two human tumor cell lines (A549 and HepG2).


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Gynostemma/chemistry , Saponins/isolation & purification , A549 Cells , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Hep G2 Cells , Humans , Molecular Structure , Plant Components, Aerial/chemistry , Plant Extracts/chemistry , Saponins/chemistry , Saponins/pharmacology , Triterpenes , Dammaranes
7.
Biochem J ; 474(5): 851-864, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28049758

ABSTRACT

Cathepsin K (CatK) is the predominant mammalian bone-degrading protease and thus an ideal target for antiosteoporotic drug development. Rodent models of osteoporosis are preferred due to their close reflection of the human disease and their ease of handling, genetic manipulation and economic affordability. However, large differences in the potency of CatK inhibitors for the mouse/rat vs. the human protease orthologs have made it impossible to use rodent models. This is even more of a problem considering that the most advanced CatK inhibitors, including odanacatib (ODN) and balicatib, failed in human clinical trials due to side effects and rodent models are not available to investigate the mechanism of these failures. Here, we elucidated the structural elements of the potency differences between mouse and human CatK (hCatK) using ODN. We determined and compared the structures of inhibitor-free mouse CatK (mCatK), hCatK and ODN bound to hCatK. Two structural differences were identified and investigated by mutational analysis. Humanizing subsite 2 in mCatK led to a 5-fold improvement of ODN binding, whereas the replacement of Tyr61 in mCatK with Asp resulted in an hCatK with comparable ODN potency. Combining both sites further improved the inhibition of the mCatK variant. Similar results were obtained for balicatib. These findings will allow the generation of transgenic CatK mice that will facilitate the evaluation of CatK inhibitor adverse effects and to explore routes to avoid them.


Subject(s)
Benzamides/chemistry , Biphenyl Compounds/chemistry , Bone Density Conservation Agents/chemistry , Cathepsin K/antagonists & inhibitors , Piperazines/chemistry , Protease Inhibitors/chemistry , Amino Acid Sequence , Animals , Benzamides/metabolism , Binding Sites , Biphenyl Compounds/metabolism , Bone Density Conservation Agents/metabolism , Cathepsin K/chemistry , Cathepsin K/genetics , Cathepsin K/metabolism , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Kinetics , Ligands , Mice , Mutagenesis, Site-Directed , Piperazines/metabolism , Protease Inhibitors/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Structural Homology, Protein
8.
J Nat Prod ; 79(8): 1962-70, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27498895

ABSTRACT

Natural products are an important source of novel drug scaffolds. The highly variable and unpredictable timelines associated with isolating novel compounds and elucidating their structures have led to the demise of exploring natural product extract libraries in drug discovery programs. Here we introduce affinity crystallography as a new methodology that significantly shortens the time of the hit to active structure cycle in bioactive natural product discovery research. This affinity crystallography approach is illustrated by using semipure fractions of an actinomycetes culture extract to isolate and identify a cathepsin K inhibitor and to compare the outcome with the traditional assay-guided purification/structural analysis approach. The traditional approach resulted in the identification of the known inhibitor antipain (1) and its new but lower potency dehydration product 2, while the affinity crystallography approach led to the identification of a new high-affinity inhibitor named lichostatinal (3). The structure and potency of lichostatinal (3) was verified by total synthesis and kinetic characterization. To the best of our knowledge, this is the first example of isolating and characterizing a potent enzyme inhibitor from a partially purified crude natural product extract using a protein crystallographic approach.


Subject(s)
Biological Products/pharmacology , Cathepsin K/antagonists & inhibitors , Lichens/chemistry , Peptides/isolation & purification , Peptides/pharmacology , Antipain/chemistry , Antipain/pharmacology , Biological Products/chemical synthesis , Biological Products/chemistry , British Columbia , Crystallography, X-Ray , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry
9.
ACS Cent Sci ; 2(3): 154-161, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27066537

ABSTRACT

Selective inhibitors of human pancreatic α-amylase (HPA) are an effective means of controlling blood sugar levels in the management of diabetes. A high-throughput screen of marine natural product extracts led to the identification of a potent (Ki = 10 pM) peptidic HPA inhibitor, helianthamide, from the Caribbean sea anemone Stichodactyla helianthus. Active helianthamide was produced in Escherichia coli via secretion as a barnase fusion protein. X-ray crystallographic analysis of the complex of helianthamide with porcine pancreatic α-amylase revealed that helianthamide adopts a ß-defensin fold and binds into and across the amylase active site, utilizing a contiguous YIYH inhibitory motif. Helianthamide represents the first of a novel class of glycosidase inhibitors and provides an unusual example of functional malleability of the ß-defensin fold, which is rarely seen outside of its traditional role in antimicrobial peptides.

10.
FEBS Lett ; 590(8): 1143-51, 2016 04.
Article in English | MEDLINE | ID: mdl-27000970

ABSTRACT

As part of a search for selective, mechanism-based covalent inhibitors of human pancreatic α-amylase we describe the chemoenzymatic synthesis of the disaccharide analog α-glucosyl epi-cyclophellitol, demonstrate its stoichiometric reaction with human pancreatic α-amylase and evaluate the time dependence of its inhibition. X-ray crystallographic analysis of the covalent derivative so formed confirms its reaction at the active site with formation of a covalent bond to the catalytic nucleophile D197. The structure illuminates the interactions with the active site and confirms OH4' on the nonreducing end sugar as a good site for attachment of fluorescent tags in generating probes for localization and quantitation of amylase in vivo.


Subject(s)
Cyclohexanols/pharmacology , Disaccharides/pharmacology , Epoxy Compounds/pharmacology , Pancreatic alpha-Amylases/chemistry , Pancreatic alpha-Amylases/metabolism , Catalytic Domain , Computer Simulation , Humans , Hydrogen Bonding , Inositol/analogs & derivatives , Inositol/chemistry , Inositol/metabolism , Kinetics , Mass Spectrometry , Models, Molecular , Water , X-Ray Diffraction
11.
Nat Chem Biol ; 11(9): 691-6, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26214255

ABSTRACT

The complex plant flavonol glycoside montbretin A is a potent (Ki = 8 nM) and specific inhibitor of human pancreatic α-amylase with potential as a therapeutic for diabetes and obesity. Controlled degradation studies on montbretin A, coupled with inhibition analyses, identified an essential high-affinity core structure comprising the myricetin and caffeic acid moieties linked via a disaccharide. X-ray structural analyses of the montbretin A-human α-amylase complex confirmed the importance of this core structure and revealed a novel mode of glycosidase inhibition wherein internal π-stacking interactions between the myricetin and caffeic acid organize their ring hydroxyls for optimal hydrogen bonding to the α-amylase catalytic residues D197 and E233. This novel inhibitory motif can be reproduced in a greatly simplified analog, offering potential for new strategies for glycosidase inhibition and therapeutic development.


Subject(s)
Drug Design , Enzyme Inhibitors/chemistry , Flavonols/chemistry , Glycosides/chemistry , alpha-Amylases/chemistry , Binding Sites , Caffeic Acids/chemistry , Carbohydrate Sequence , Enzyme Inhibitors/chemical synthesis , Flavones/chemistry , Flavonoids/chemistry , Gene Expression , Humans , Hydrogen Bonding , Hydrolysis , Ligands , Models, Molecular , Molecular Sequence Data , Pichia/genetics , Pichia/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Trisaccharides/chemistry , alpha-Amylases/antagonists & inhibitors , alpha-Amylases/genetics
12.
Proc Natl Acad Sci U S A ; 111(49): 17474-9, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25422423

ABSTRACT

Cathepsin K is the major collagenolytic protease in bone that facilitates physiological as well as pathological bone degradation. Despite its key role in bone remodeling and for being a highly sought-after drug target for the treatment of osteoporosis, the mechanism of collagen fiber degradation by cathepsin K remained elusive. Here, we report the structure of a collagenolytically active cathepsin K protein dimer. Cathepsin K is organized into elongated C-shaped protease dimers that reveal a putative collagen-binding interface aided by glycosaminoglycans. Molecular modeling of collagen binding to the dimer indicates the participation of nonactive site amino acid residues, Q21 and Q92, in collagen unfolding. Mutations at these sites as well as perturbation of the dimer protein-protein interface completely inhibit cathepsin-K-mediated fiber degradation without affecting the hydrolysis of gelatin or synthetic peptide. Using scanning electron microscopy, we demonstrate the specific binding of cathepsin K at the edge of the fibrillar gap region of collagen fibers, which suggest initial cleavage events at the N- and C-terminal ends of tropocollagen molecules. Edman degradation analysis of collagen fiber degradation products revealed those initial cleavage sites. We propose that one cathepsin K molecule binds to collagen-bound glycosaminoglycans at the gap region and recruits a second protease molecule that provides an unfolding and cleavage mechanism for triple helical collagen. Removal of collagen-associated glycosaminoglycans prevents cathepsin K binding and subsequently fiber hydrolysis. Cathepsin K dimer and glycosaminoglycan binding sites represent novel targeting sites for the development of nonactive site-directed second-generation inhibitors of this important drug target.


Subject(s)
Cathepsin K/chemistry , Collagen/chemistry , Amino Acids/chemistry , Binding Sites , Bone Remodeling , Bone and Bones/metabolism , Crystallography, X-Ray , Glycosaminoglycans/chemistry , Humans , Hydrolysis , Microscopy, Electron , Models, Molecular , Mutagenesis , Osteoporosis , Peptide Hydrolases/chemistry , Pichia , Protein Denaturation , Protein Folding , Protein Multimerization , Protein Structure, Tertiary , Substrate Specificity
13.
Biochim Biophys Acta ; 1834(12): 2546-53, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23954305

ABSTRACT

The citrate synthase (CS) of Escherichia coli is an allosteric hexameric enzyme specifically inhibited by NADH. The crystal structure of wild type (WT) E. coli CS, determined by us previously, has no substrates bound, and part of the active site is in a highly mobile region that is shifted from the position needed for catalysis. The CS of Acetobacter aceti has a similar structure, but has been successfully crystallized with bound substrates: both oxaloacetic acid (OAA) and an analog of acetyl coenzyme A (AcCoA). We engineered a variant of E. coli CS wherein five amino acids in the mobile region have been replaced by those in the A. aceti sequence. The purified enzyme shows unusual kinetics with a low affinity for both substrates. Although the crystal structure without ligands is very similar to that of the WT enzyme (except in the mutated region), complexes are formed with both substrates and the allosteric inhibitor NADH. The complex with OAA in the active site identifies a novel OAA-binding residue, Arg306, which has no functional counterpart in other known CS-OAA complexes. This structure may represent an intermediate in a multi-step substrate binding process where Arg306 changes roles from OAA binding to AcCoA binding. The second complex has the substrate analog, S-carboxymethyl-coenzyme A, in the allosteric NADH-binding site and the AcCoA site is not formed. Additional CS variants unable to bind adenylates at the allosteric site show that this second complex is not a factor in positive allosteric activation of AcCoA binding.


Subject(s)
Acetobacter/enzymology , Acetyl Coenzyme A/chemistry , Citrate (si)-Synthase/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , NADP/chemistry , Acetobacter/genetics , Acetyl Coenzyme A/genetics , Acetyl Coenzyme A/metabolism , Allosteric Regulation , Animals , Catalytic Domain , Citrate (si)-Synthase/genetics , Citrate (si)-Synthase/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , NADP/genetics , NADP/metabolism , Protein Binding , Swine
14.
Glycobiology ; 23(9): 1075-83, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23735230

ABSTRACT

Trehalose synthase (TreS) catalyzes the reversible conversion of maltose into trehalose in mycobacteria as one of three biosynthetic pathways to this nonreducing disaccharide. Given the importance of trehalose to survival of mycobacteria, there has been considerable interest in understanding the enzymes involved in its production; indeed the structures of the key enzymes in the other two pathways have already been determined. Herein, we present the first structure of TreS from Mycobacterium smegmatis, thereby providing insights into the catalytic machinery involved in this intriguing intramolecular reaction. This structure, which is of interest both mechanistically and as a potential pharmaceutical target, reveals a narrow and enclosed active site pocket within which intramolecular substrate rearrangements can occur. We also present the structure of a complex of TreS with acarbose, revealing a hitherto unsuspected oligosaccharide-binding site within the C-terminal domain. This may well provide an anchor point for the association of TreS with glycogen, thereby enhancing its role in glycogen biosynthesis and degradation.


Subject(s)
Acarbose/metabolism , Glucosyltransferases/chemistry , Glucosyltransferases/metabolism , Mycobacterium smegmatis/enzymology , Acarbose/chemistry , Acarbose/pharmacology , Amino Acid Sequence , Biocatalysis/drug effects , Catalytic Domain/drug effects , Glucosyltransferases/antagonists & inhibitors , Molecular Sequence Data , Protein Binding , Protein Conformation , Sequence Alignment , Structure-Activity Relationship
15.
Article in English | MEDLINE | ID: mdl-19478443

ABSTRACT

Autotransporters (ATs) are proteins that deliver effectors (the passenger domain) to the surface of Gram-negative bacteria by the type V secretion pathway. The passenger domain of BrkA, a Bordetella pertussis autotransporter mediating serum resistance and adherence, was cloned in a pET expression system and overexpressed in Escherichia coli. The gene product was correctly refolded, purified to homogeneity and crystallized. The crystals diffracted to 2.8 A resolution. The space group was assumed to be P4(1)2(1)2, with unit-cell parameters a = b = 108.19, c = 115.35 A.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bordetella pertussis/chemistry , Membrane Transport Proteins/chemistry , Amino Acid Sequence , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/isolation & purification , Bacterial Outer Membrane Proteins/physiology , Bordetella pertussis/genetics , Circular Dichroism , Cloning, Molecular , Crystallization , Data Collection , Escherichia coli/genetics , Inclusion Bodies/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/physiology , Molecular Sequence Data , Protein Folding , Protein Sorting Signals , Protein Structure, Tertiary/genetics , Proteins/genetics , Proteins/metabolism , Secretory Pathway , Sequence Homology, Amino Acid , Statistics as Topic , Temperature , X-Ray Diffraction
16.
J Biol Chem ; 284(12): 7897-902, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19139097

ABSTRACT

The Syd protein has been implicated in the Sec-dependent transport of polypeptides across the bacterial inner membrane. Using Nanodiscs, we here provide direct evidence that Syd binds the SecY complex, and we demonstrate that interaction involves the two electropositive and cytosolic loops of the SecY subunit. We solve the crystal structure of Syd and together with cysteine cross-link analysis, we show that a conserved concave and electronegative groove constitutes the SecY-binding site. At the membrane, Syd decreases the activity of the translocon containing loosely associated SecY-SecE subunits, whereas in detergent solution Syd disrupts the SecYEG heterotrimeric associations. These results support the role of Syd in proofreading the SecY complex biogenesis and point to the electrostatic nature of the Sec channel interaction with its cytosolic partners.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Membrane Proteins/chemistry , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Binding/physiology , Protein Structure, Quaternary/physiology , Protein Structure, Secondary/physiology , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport/physiology , SEC Translocation Channels , Static Electricity
17.
J Biol Chem ; 278(37): 35435-43, 2003 Sep 12.
Article in English | MEDLINE | ID: mdl-12824188

ABSTRACT

The citrate synthase of Escherichia coli is an example of a Type II citrate synthase, a hexamer that is subject to allosteric inhibition by NADH. In previous crystallographic work, we defined the NADH binding sites, identifying nine amino acids whose side chains were proposed to make hydrogen bonds with the NADH molecule. Here, we describe the functional properties of nine sequence variants, in which these have been replaced by nonbonding residues. All of the variants show some changes in NADH binding and inhibition and small but significant changes in kinetic parameters for catalysis. In three cases, Y145A, R163L, and K167A, NADH inhibition has become extremely weak. We have used nanospray/time-of-flight mass spectrometry, under non-denaturing conditions, to show that two of these, R163L and K167A, do not form hexamers in response to NADH binding, unlike the wild type enzyme. One variant, R109L, shows tighter NADH binding. We have crystallized this variant and determined its structure, with and without bound NADH. Unexpectedly, the greatest structural changes in the R109L variant are in two regions outside the NADH binding site, both of which, in wild type citrate synthase, have unusually high mobilities as measured by crystallographic thermal factors. In the R109L variant, both regions (residues 260 -311 and 316-342) are much less mobile and have rearranged significantly. We argue that these two regions are elements in the path of communication between the NADH binding sites and the active sites and are centrally involved in the regulatory conformational change in E. coli citrate synthase.


Subject(s)
Citrate (si)-Synthase/chemistry , Citrate (si)-Synthase/metabolism , Escherichia coli/enzymology , NAD/metabolism , Amino Acid Substitution , Binding Sites , Crystallography, X-Ray , Genetic Variation , Hydrogen Bonding , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Static Electricity , Thermodynamics
18.
Biochemistry ; 42(19): 5555-65, 2003 May 20.
Article in English | MEDLINE | ID: mdl-12741811

ABSTRACT

Study of the hexameric and allosterically regulated citrate synthases (type II CS) provides a rare opportunity to gain not only an understanding of a novel allosteric mechanism but also insight into how such properties can evolve from an unregulated structural platform (the dimeric type I CS). To address both of these issues, we have determined the structure of the complex of NADH (a negative allosteric effector) with the F383A variant of type II Escherichia coli CS. This variant was chosen because its kinetics indicate it is primarily in the T or inactive allosteric conformation, the state that strongly binds to NADH. Our structural analyses show that the six NADH binding sites in the hexameric CS complex are located at the interfaces between dimer units such that most of each site is formed by one subunit, but a number of key residues are drawn from the adjacent dimer. This arrangement of interactions serves to explain why NADH allosteric regulation is a feature only of hexameric type II CS. Surprisingly, in both the wild-type enzyme and the NADH complex, the two subunits of each dimer within the hexameric conformation are similar but not identical in structure, and therefore, while the general characteristics of NADH binding interactions are similar in each subunit, the details of these are somewhat different between subunits. Detailed examination of the observed NADH binding sites indicates that both direct charged interactions and the overall cationic nature of the sites are likely responsible for the ability of these sites to discriminate between NADH and NAD(+). A particularly novel characteristic of the complex is the horseshoe conformation assumed by NADH, which is strikingly different from the extended conformation found in its complexes with most proteins. Sequence homology studies suggest that this approach to binding NADH may arise out of the evolutionary need to add an allosteric regulatory function to the base CS structure. Comparisons of the amino acid sequences of known type II CS enzymes, from different Gram-negative bacteria taxonomic groups, show that the NADH-binding residues identified in our structure are strongly conserved, while hexameric CS molecules that are insensitive to NADH have undergone key changes in the sequence of this part of the protein.


Subject(s)
Citrate (si)-Synthase/chemistry , Citrate (si)-Synthase/metabolism , NAD/metabolism , Allosteric Regulation , Allosteric Site/genetics , Amino Acid Sequence , Citrate (si)-Synthase/classification , Citrate (si)-Synthase/genetics , Crystallography, X-Ray , Dimerization , Escherichia coli/enzymology , Escherichia coli/genetics , Evolution, Molecular , Genetic Variation , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Quaternary , Sequence Homology, Amino Acid
19.
Proc Natl Acad Sci U S A ; 100(7): 3647-52, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12644706

ABSTRACT

A binding site for metal ions has been created on the surface of horse heart myoglobin (Mb) near the heme 6-propionate group by replacing K45 and K63 with glutamyl residues. One-dimensional (1)H NMR spectroscopy indicates that Mn(2+) binds in the vicinity of the heme 6-propionate as anticipated, and potentiometric titrations establish that the affinity of the new site for Mn(2+) is 1.28(4) x 10(4) M(-1) (pH 6.96, ionic strength I = 17.2 microM, 25 degrees C). In addition, these substitutions lower the reduction potential of the protein and increase the pK(a) for the water molecule coordinated to the heme iron of metmyoglobin. The peroxidase [2,2'-azinobis(3-ethylbenzthiazoline-6-sulfonic acid), ABTS, as substrate] and the Mn(2+)-peroxidase activity of the variant are both increased approximately 3-fold. In contrast to wild-type Mb, both the affinity for azide and the midpoint potential of the variant are significantly influenced by the addition of Mn(2+). The structure of the variant has been determined by x-ray crystallography to define the coordination environment of bound Mn(2+) and Cd(2+). Although slight differences are observed between the geometry of the binding of the two metal ions, both are hexacoordinate, and neither involves coordination by E63.


Subject(s)
Heme/metabolism , Metals/metabolism , Myoglobin/chemistry , Animals , Binding Sites , Chlorides/chemistry , Crystallography, X-Ray/methods , Electrochemistry/methods , Heme/chemistry , Horses , Kinetics , Manganese Compounds/chemistry , Models, Molecular , Mutagenesis , Myoglobin/genetics , Myoglobin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrophotometry/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...