Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Chem Sci ; 9(25): 5585-5593, 2018 Jul 07.
Article in English | MEDLINE | ID: mdl-30061990

ABSTRACT

The bioconjugation of proteins with small molecules has proved an invaluable strategy for probing and perturbing biological mechanisms. The general use of chemical methods for protein functionalisation can be limited however by the requirement for complicated reaction partners to be present in large excess, and harsh conditions which are incompatible with many protein scaffolds. Herein we describe a site-selective organocatalyst-mediated protein aldol ligation (OPAL) that affords stable carbon-carbon linked bioconjugates at neutral pH. OPAL enables rapid modification of proteins using simple aldehyde probes in minimal excess, and is utilised here in the affinity tagging of proteins in cell lysate. Furthermore we demonstrate that the ß-hydroxy aldehyde OPAL product can be functionalised again at neutral pH in a tandem organocatalyst-mediated oxime ligation. This tandem strategy is showcased in the 'chemical mimicry' of a previously inaccessible natural dual post-translationally modified protein integral to the pathogenesis of the neglected tropical disease Leishmaniasis.

2.
J Med Chem ; 55(20): 8879-90, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-23035716

ABSTRACT

Design of inhibitors for N-myristoyltransferase (NMT), an enzyme responsible for protein trafficking in Plasmodium falciparum , the most lethal species of parasites that cause malaria, is described. Chemistry-driven optimization of compound 1 from a focused NMT inhibitor library led to the identification of two early lead compounds 4 and 25, which showed good enzyme and cellular potency and excellent selectivity over human NMT. These molecules provide a valuable starting point for further development.


Subject(s)
Acyltransferases/antagonists & inhibitors , Antimalarials/chemical synthesis , Benzofurans/chemical synthesis , Piperidines/chemical synthesis , Plasmodium falciparum/enzymology , Acyltransferases/genetics , Antimalarials/chemistry , Antimalarials/pharmacology , Benzofurans/chemistry , Benzofurans/pharmacology , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Piperidines/chemistry , Piperidines/pharmacology , Plasmodium falciparum/drug effects , Protein Conformation , Stereoisomerism , Structure-Activity Relationship
3.
J Biol Chem ; 286(42): 36968-77, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21880708

ABSTRACT

Apart from its role in insulin receptor (IR) activation, the C terminus of the B-chain of insulin is also responsible for the formation of insulin dimers. The dimerization of insulin plays an important role in the endogenous delivery of the hormone and in the administration of insulin to patients. Here, we investigated insulin analogues with selective N-methylations of peptide bond amides at positions B24, B25, or B26 to delineate their structural and functional contribution to the dimer interface. All N-methylated analogues showed impaired binding affinities to IR, which suggests a direct IR-interacting role for the respective amide hydrogens. The dimerization capabilities of analogues were investigated by isothermal microcalorimetry. Selective N-methylations of B24, B25, or B26 amides resulted in reduced dimerization abilities compared with native insulin (K(d) = 8.8 µM). Interestingly, although the N-methylation in [NMeTyrB26]-insulin or [NMePheB24]-insulin resulted in K(d) values of 142 and 587 µM, respectively, the [NMePheB25]-insulin did not form dimers even at high concentrations. This effect may be attributed to the loss of intramolecular hydrogen bonding between NHB25 and COA19, which connects the B-chain ß-strand to the core of the molecule. The release of the B-chain ß-strand from this hydrogen bond lock may result in its higher mobility, thereby shifting solution equilibrium toward the monomeric state of the hormone. The study was complemented by analyses of two novel analogue crystal structures. All examined analogues crystallized only in the most stable R(6) form of insulin oligomers (even if the dimer interface was totally disrupted), confirming the role of R(6)-specific intra/intermolecular interactions for hexamer stability.


Subject(s)
Insulin, Regular, Pork/chemistry , Protein Multimerization , Animals , Crystallography, X-Ray , Hydrogen Bonding , Methylation , Protein Stability , Protein Structure, Quaternary , Protein Structure, Secondary , Swine
4.
Proc Natl Acad Sci U S A ; 107(50): 21412-7, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21098297

ABSTRACT

The porins OmpF and OmpC are trimeric ß-barrel proteins with narrow channels running through each monomer that exclude molecules > 600 Da while mediating the passive diffusion of small nutrients and metabolites across the Gram-negative outer membrane (OM). Here, we elucidate the mechanism by which an entire soluble protein domain (> 6 kDa) is delivered through the lumen of such porins. Following high-affinity binding to the vitamin B(12) receptor in Escherichia coli, the bacteriocin ColE9 recruits OmpF or OmpC using an 83-residue intrinsically unstructured translocation domain (IUTD) to deliver a 16-residue TolB-binding epitope (TBE) in the center of the IUTD to the periplasm where it triggers toxin entry. We demonstrate that the IUTD houses two OmpF-binding sites, OBS1 (residues 2-18) and OBS2 (residues 54-63), which flank the TBE and bind with K(d)s of 2 and 24 µM, respectively, at pH 6.5 and 25 ºC. We show the two OBSs share the same binding site on OmpF and that the colicin must house at least one of them for antibiotic activity. Finally, we report the structure of the OmpF-OBS1 complex that shows the colicin bound within the porin lumen spanning the membrane bilayer. Our study explains how colicins exploit porins to deliver epitope signals to the bacterial periplasm and, more broadly, how the inherent flexibility and narrow cross-sectional area of an IUP domain can endow it with the ability to traverse a biological membrane via the constricted lumen of a ß-barrel membrane protein.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/metabolism , Epitopes , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Porins/metabolism , Bacterial Outer Membrane Proteins/chemistry , Colicins/chemistry , Colicins/genetics , Colicins/metabolism , Crystallography, X-Ray , Escherichia coli/cytology , Escherichia coli Proteins/chemistry , Models, Molecular , Porins/chemistry , Porins/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
6.
J Mol Biol ; 384(5): 1218-31, 2008 Dec 31.
Article in English | MEDLINE | ID: mdl-18951902

ABSTRACT

Monoamine oxidase from Aspergillus niger (MAO-N) is a flavoenzyme that catalyses the oxidative deamination of primary amines. MAO-N has been used as the starting model for a series of directed evolution experiments, resulting in mutants of improved activity and broader substrate specificity, suitable for application in the preparative deracemisation of primary, secondary and tertiary amines when used as part of a chemoenzymatic oxidation-reduction cycle. The structures of a three-point mutant (Asn336Ser/Met348Lys/Ile246Met or MAO-N-D3) and a five-point mutant (Asn336Ser/Met348Lys/Ile246Met/Thr384Asn/Asp385Ser or MAO-N-D5) have been obtained using a multiple-wavelength anomalous diffraction experiment on a selenomethionine derivative of the truncated MAO-N-D5 enzyme. MAO-N exists as a homotetramer with a large channel at its centre and shares some structural features with human MAO B (MAO-B). A hydrophobic cavity extends from the protein surface to the active site, where a non-covalently bound flavin adenine dinucleotide (FAD) sits at the base of an 'aromatic cage,' the sides of which are formed by Trp430 and Phe466. A molecule of l-proline was observed near the FAD, and this ligand superimposed well with isatin, a reversible inhibitor of MAO-B, when the structures of MAO-N proline and MAO-B-isatin were overlaid. Of the mutations that confer the ability to catalyse the oxidation of secondary amines in MAO-N-D3, Asn336Ser reduces steric bulk behind Trp430 of the aromatic cage and Ile246Met confers greater flexibility within the substrate binding site. The two additional mutations, Thr384Asn and Asp385Ser, that occur in the MAO-N-D5 variant, which is able to oxidise tertiary amines, appear to influence the active-site environment remotely through changes in tertiary structure that perturb the side chain of Phe382, again altering the steric and electronic character of the active site near FAD. The possible implications of the change in steric and electronic environment caused by relevant mutations are discussed with respect to the improved catalytic efficiency of the MAO-N variants described in the literature.


Subject(s)
Aspergillus niger/enzymology , Directed Molecular Evolution , Monoamine Oxidase/chemistry , Monoamine Oxidase/metabolism , Amino Acid Sequence , Aspergillus niger/genetics , Binding Sites , Biocatalysis , Catalytic Domain , Flavin-Adenine Dinucleotide/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Isomerism , Molecular Sequence Data , Monoamine Oxidase/genetics , Mutant Proteins/chemistry , Mutation/genetics , Oxidation-Reduction , Proline/metabolism , Protein Structure, Secondary , Sequence Alignment , Static Electricity , Substrate Specificity
7.
Biochem J ; 414(2): 281-9, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18479250

ABSTRACT

HCHL (hydroxycinnamoyl-CoA hydratase-lyase) catalyses the biotransformation of feruloyl-CoA to acetyl-CoA and the important flavour-fragrance compound vanillin (4-hydroxy-3-methoxybenzaldehyde) and is exploited in whole-cell systems for the bioconversion of ferulic acid into natural equivalent vanillin. The reaction catalysed by HCHL has been thought to proceed by a two-step process involving first the hydration of the double bond of feruloyl-CoA and then the cleavage of the resultant beta-hydroxy thioester by retro-aldol reaction to yield the products. Kinetic analysis of active-site residues identified using the crystal structure of HCHL revealed that while Glu-143 was essential for activity, Ser-123 played no major role in catalysis. However, mutation of Tyr-239 to Phe greatly increased the K(M) for the substrate ferulic acid, fulfilling its anticipated role as a factor in substrate binding. Structures of WT (wild-type) HCHL and of the S123A mutant, each of which had been co-crystallized with feruloyl-CoA, reveal a subtle helix movement upon ligand binding, the consequence of which is to bring the phenolic hydroxyl of Tyr-239 into close proximity to Tyr-75 from a neighbouring subunit in order to bind the phenolic hydroxyl of the product vanillin, for which electron density was observed. The active-site residues of ligand-bound HCHL display a remarkable three-dimensional overlap with those of a structurally unrelated enzyme, vanillyl alcohol oxidase, that also recognizes p-hydroxylated aromatic substrates related to vanillin. The data both explain the observed substrate specificity of HCHL for p-hydroxylated cinnamate derivatives and illustrate a remarkable convergence of the molecular determinants of ligand recognition between the two otherwise unrelated enzymes.


Subject(s)
Acetyl Coenzyme A/metabolism , Benzaldehydes/metabolism , Enoyl-CoA Hydratase/metabolism , Hydro-Lyases/metabolism , Acetyl Coenzyme A/chemistry , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Benzaldehydes/chemistry , Binding Sites , Crystallization , Crystallography, X-Ray , Enoyl-CoA Hydratase/chemistry , Enoyl-CoA Hydratase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Hydro-Lyases/chemistry , Hydro-Lyases/genetics , Kinetics , Magnetic Resonance Spectroscopy , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Pseudomonas fluorescens/enzymology , Pseudomonas fluorescens/genetics , Pseudomonas fluorescens/metabolism , Substrate Specificity
8.
Article in English | MEDLINE | ID: mdl-17620706

ABSTRACT

Benzaldehyde lyase (BAL; EC 4.1.2.38) is a thiamine diphosphate (ThDP) dependent enzyme that catalyses the enantioselective carboligation of two molecules of benzaldehyde to form (R)-benzoin. BAL has hence aroused interest for its potential in the industrial synthesis of optically active benzoins and derivatives. The structure of BAL was previously solved to a resolution of 2.6 A using MAD experiments on a selenomethionine derivative [Mosbacher et al. (2005), FEBS J. 272, 6067-6076]. In this communication of parallel studies, BAL was crystallized in an alternative space group (P2(1)2(1)2(1)) and its structure refined to a resolution of 1.65 A, allowing detailed observation of the water structure, active-site interactions with ThDP and also the electron density for the co-solvent 2-methyl-2,4-pentanediol (MPD) at hydrophobic patches of the enzyme surface.


Subject(s)
Aldehyde-Lyases/chemistry , Thiamine Pyrophosphate/chemistry , Aldehyde-Lyases/metabolism , Binding Sites , Crystallography, X-Ray , Protein Conformation , Protein Structure, Secondary , Thiamine Pyrophosphate/metabolism
9.
Biochemistry ; 46(1): 137-44, 2007 Jan 09.
Article in English | MEDLINE | ID: mdl-17198383

ABSTRACT

The gene alr4455 from the well-studied cyanobacterium Anabaena sp. PCC 7120 encodes a crotonase orthologue that displays beta-diketone hydrolase activity. Anabaena beta-diketone hydrolase (ABDH), in common with 6-oxocamphor hydrolase (OCH) from Rhodococcus sp. NCIMB 9784, catalyzes the desymmetrization of bicyclo[2.2.2]octane-2,6-dione to yield [(S)-3-oxocyclohexyl]acetic acid, a reaction unusual among the crotonase superfamily as the substrate is not an acyl-CoA thioester. The structure of ABDH has been determined to a resolution of 1.5 A in both native and ligand-bound forms. ABDH forms a hexamer similar to OCH and features one active site per enzyme monomer. The arrangement of side chains in the active site indicates that while the catalytic chemistry may be conserved in OCH orthologues, the structural determinants of substrate specificity are different. In the active site of ligand-bound forms that had been cocrystallized with the bicyclic diketone substrate bicyclo[2.2.2]octane-2,6-dione was found the product of the asymmetric enzymatic retro-Claisen reaction [(S)-3-oxocyclohexyl]acetic acid. The structures of ABDH in both native and ligand-bound forms reveal further details about structural variation and modes of coenzyme A-independent activity within the crotonases and provide further evidence of a wider suprafamily of enzymes that have recruited the crotonase fold for the catalysis of reactions other than those regularly attributed to canonical superfamily members.


Subject(s)
Anabaena/enzymology , Bacterial Proteins/chemistry , Hydrolases/chemistry , Amino Acid Sequence , Anabaena/classification , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Binding Sites , Coenzyme A/metabolism , Crystallography, X-Ray , Enoyl-CoA Hydratase/chemistry , Enoyl-CoA Hydratase/classification , Enoyl-CoA Hydratase/metabolism , Hydrolases/classification , Hydrolases/metabolism , Ketones/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Alignment
10.
Acta Crystallogr D Biol Crystallogr ; 62(Pt 12): 1494-501, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17139085

ABSTRACT

The crystal structure of hydroxycinnamoyl-CoA hydratase-lyase (HCHL) from Pseudomonas fluorescens AN103 has been solved to 1.8 A resolution. HCHL is a member of the crotonase superfamily and catalyses the hydration of the acyl-CoA thioester of ferulic acid [3-(4-hydroxy-3-methoxy-phenyl)prop-2-enoic acid] and the subsequent retro-aldol cleavage of the hydrated intermediate to yield vanillin (4-hydroxy-3-methoxy-benzaldehyde). The structure contains 12 molecules in the asymmetric unit, in which HCHL assumes a hexameric structure of two stacked trimers. The substrate, feruloyl-CoA, was modelled into the active site based on the structure of enoyl-CoA hydratase bound to the feruloyl-CoA-like substrate 4-(N,N-dimethylamino)-cinnamoyl-CoA (PDB code 1ey3). Feruloyl-CoA was bound in this model between helix 3 of the A subunit and helix 9 of the B subunit. A highly ordered structural water in the HCHL structure coincided with the thioester carbonyl of feruloyl-CoA in the model, suggesting that the oxyanion hole for stabilization of a thioester-derived enolate, characteristic of coenzyme-A dependent members of the crotonase superfamily, is conserved. The model also suggested that a strong hydrogen bond between the phenolic hydroxyl groups of feruloyl-CoA and BTyr239 may be an important determinant of the enzyme's ability to discriminate between the natural substrate and cinnamoyl-CoA, which is not a substrate.


Subject(s)
Acyl Coenzyme A/metabolism , Benzaldehydes/metabolism , Hydro-Lyases/chemistry , Hydro-Lyases/metabolism , Pseudomonas fluorescens/enzymology , Acyl Coenzyme A/chemistry , Benzaldehydes/chemistry , Binding Sites , Catalysis , Crystallography, X-Ray , Enoyl-CoA Hydratase/chemistry , Models, Molecular , Protein Folding , Protein Structure, Quaternary , Protein Structure, Secondary
11.
J Biol Chem ; 281(16): 10968-75, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16431911

ABSTRACT

The enzymatic degradation of plant cell wall xylan requires the concerted action of a diverse enzymatic syndicate. Among these enzymes are xylan esterases, which hydrolyze the O-acetyl substituents, primarily at the O-2 position of the xylan backbone. All acetylxylan esterase structures described previously display a alpha/beta hydrolase fold with a "Ser-His-Asp" catalytic triad. Here we report the structures of two distinct acetylxylan esterases, those from Streptomyces lividans and Clostridium thermocellum, in native and complex forms, with x-ray data to between 1.6 and 1.0 A resolution. We show, using a novel linked assay system with PNP-2-O-acetylxyloside and a beta-xylosidase, that the enzymes are sugar-specific and metal ion-dependent and possess a single metal center with a chemical preference for Co2+. Asp and His side chains complete the catalytic machinery. Different metal ion preferences for the two enzymes may reflect the surprising diversity with which the metal ion coordinates residues and ligands in the active center environment of the S. lividans and C. thermocellum enzymes. These "CE4" esterases involved in plant cell wall degradation are shown to be closely related to the de-N-acetylases involved in chitin and peptidoglycan degradation (Blair, D. E., Schuettelkopf, A. W., MacRae, J. I., and Aalten, D. M. (2005) Proc. Natl. Acad. Sci. U. S. A., 102, 15429-15434), which form the NodB deacetylase "superfamily."


Subject(s)
Acetylesterase/chemistry , Amidohydrolases/chemistry , Cell Wall/metabolism , Peptidoglycan/chemistry , Aspartic Acid/chemistry , Binding Sites , Carbohydrate Metabolism , Carbohydrates/chemistry , Catalysis , Chitin/chemistry , Clostridium thermocellum/metabolism , Cobalt/chemistry , Crystallography, X-Ray , Dimerization , Escherichia coli/metabolism , Histidine/chemistry , Ions , Ligands , Metals/chemistry , Models, Chemical , Models, Molecular , Models, Statistical , Nickel/chemistry , Plants/metabolism , Protein Conformation , Protein Structure, Tertiary , Streptomyces lividans/metabolism , Structure-Activity Relationship , Xylans/chemistry
12.
Acta Crystallogr D Biol Crystallogr ; 61(Pt 2): 190-3, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15681870

ABSTRACT

The enzymatic digestion of starch by alpha-amylases is one of the key biotechnological reactions of recent times. In the search for industrial biocatalysts, the family GH13 alpha-amylase BHA from Bacillus halmapalus has been cloned and expressed. The three-dimensional structure at 2.1 A resolution has been determined in complex with the (pseudo)tetrasaccharide inhibitor acarbose. Acarbose is found bound as a nonasaccharide transglycosylation product spanning the -6 to +3 subsites. Careful inspection of electron density suggests that the bound ligand could not have been formed through successive transglycosylations of acarbose and must also have featured maltose or maltooligosaccharides as an acceptor.


Subject(s)
Acarbose/chemistry , Bacillus/enzymology , Monosaccharides/chemistry , alpha-Amylases/chemistry , Carbohydrate Sequence , Models, Molecular , Molecular Sequence Data , Protein Conformation
13.
Chem Biol ; 10(7): 619-28, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12890535

ABSTRACT

The formation of glycoconjugates and oligosaccharides remains one of the most challenging chemical syntheses. Chemo-enzymatic routes using retaining glycosidases have been successfully harnessed but require tight kinetic or thermodynamic control. "Glycosynthases," specifically engineered glycosidases that catalyze the formation of glycosidic bonds from glycosyl donor and acceptor alcohol, are an emerging range of synthetic tools in which catalytic nucleophile mutants are harnessed together with glycosyl fluoride donors to generate powerful and versatile catalysts. Here we present the structural and kinetic dissection of the Humicola insolens Cel7B glycosynthases in which the nucleophile of the wild-type enzyme is mutated to alanine and serine (E197A and E197S). 3-D structures reveal the acceptor and donor subsites and the basis for substrate inhibition. Kinetic analysis shows that the E197S mutant is considerably more active than the corresponding alanine mutant due to a 40-fold increase in k(cat).


Subject(s)
Fungi/metabolism , Glycoside Hydrolases/metabolism , Glycosides/biosynthesis , Lactose/analogs & derivatives , Cellobiose/metabolism , Crystallization , Data Interpretation, Statistical , Fungi/enzymology , Indicators and Reagents , Kinetics , Models, Molecular , Mutagenesis/genetics , Oligosaccharides/metabolism
14.
Eur J Biochem ; 269(6): 1613-21, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11895431

ABSTRACT

The binding of Thermomyces lanuginosa lipase and its mutants [TLL(S146A), TLL(W89L), TLL(W117F, W221H, W260H)] to the mixed micelles of cis-parinaric acid/sodium taurodeoxycholate at pH 5.0 led to the quenching of the intrinsic tryptophan fluorescence emission (300-380 nm) and to a simultaneous increase in the cis-parinaric acid fluorescence emission (380-500 nm). These findings were used to characterize the Thermomyces lanuginosa lipase/cis-parinaric acid interactions occurring in the presence of sodium taurodeoxycholate. The fluorescence resonance energy transfer and Stern-Volmer quenching constant values obtained were correlated with the accessibility of the tryptophan residues to the cis-parinaric acid and with the lid opening ability of Thermomyces lanuginosa lipase (and its mutants). TLL(S146A) was found to have the highest fluorescence resonance energy transfer. In addition, a TLL(S146A)/oleic acid complex was crystallised and its three-dimensional structure was solved. Surprisingly, two possible binding modes (sn-1 and antisn1) were found to exist between oleic acid and the catalytic cleft of the open conformation of TLL(S146A). Both binding modes involved an interaction with tryptophan 89 of the lipase lid, in agreement with fluorescence resonance energy transfer experiments. As a consequence, we concluded that TLL(S146A) mutant is not an appropriate substitute for the wild-type Thermomyces lanuginosa lipase for mimicking the interaction between the wild-type enzyme and lipids.


Subject(s)
Ascomycota/enzymology , Fatty Acids, Unsaturated/metabolism , Lipase/metabolism , Micelles , Catalytic Domain , Crystallography, X-Ray , Energy Transfer , Fluorescence , Lipase/chemistry , Substrate Specificity
15.
Acta Crystallogr D Biol Crystallogr ; 58(Pt 3): 451-5, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11856830

ABSTRACT

Cathepsin S (EC 3.4.22.27), a cysteine proteinase of the papain superfamily, plays a critical role in the generation of a major histocompatibility complex (MHC) class II restricted T-cell response by antigen-presenting cells. Therefore, selective inhibition of this enzyme may be useful in modulating class II restricted T-cell responses in immune-related disorders such as rheumatoid arthritis, multiple sclerosis and extrinsic asthma. The three-dimensional structure at 2.2 A resolution of the active-site Cys25-->Ser mutant presented here in an unliganded state provides further insight useful for the design of selective enzyme inhibitors.


Subject(s)
Cathepsins/chemistry , Amino Acid Substitution , Cathepsins/genetics , Crystallization , Crystallography, X-Ray , Cysteine/genetics , Humans , Models, Molecular , Mutation , Protein Conformation , Recombinant Proteins/chemistry , Serine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...