Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Biochem J ; 475(1): 225-245, 2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29208763

RESUMO

Scabin was previously identified as a novel DNA-targeting mono-ADP-ribosyltransferase (mART) toxin from the plant pathogen 87.22 strain of Streptomyces scabies Scabin is a member of the Pierisin-like subgroup of mART toxins, since it targets DNA. An in-depth characterization of both the glycohydrolase and transferase enzymatic activities of Scabin was conducted. Several protein variants were developed based on an initial Scabin·DNA molecular model. Consequently, three residues were deemed important for DNA-binding and transferase activity. Trp128 and Trp155 are important for binding the DNA substrate and participate in the reaction mechanism, whereas Tyr129 was shown to be important only for DNA binding, but was not involved in the reaction mechanism. Trp128 and Trp155 are both conserved within the Pierisin-like toxins, whereas Tyr129 is a unique substitution within the group. Scabin showed substrate specificity toward double-stranded DNA containing a single-base overhang, as a model for single-stranded nicked DNA. The crystal structure of Scabin bound to NADH - a competitive inhibitor of Scabin - was determined, providing important insights into the active-site structure and Michaelis-Menten complex of the enzyme. Based on these results, a novel DNA-binding motif is proposed for Scabin with substrate and the key residues that may participate in the Scabin·NAD(+) complex are highlighted.


Assuntos
ADP Ribose Transferases/química , Proteínas de Bactérias/química , Toxinas Bacterianas/química , DNA Bacteriano/química , Glicosídeo Hidrolases/química , Streptomyces/enzimologia , ADP Ribose Transferases/genética , ADP Ribose Transferases/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Ligação Competitiva , Domínio Catalítico , Cristalografia por Raios X , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Expressão Gênica , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Proteínas de Insetos/química , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Cinética , Modelos Moleculares , NAD/química , NAD/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Streptomyces/genética , Streptomyces/patogenicidade , Especificidade por Substrato , Termodinâmica
2.
J Biol Chem ; 291(21): 11198-215, 2016 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-27002155

RESUMO

A bioinformatics strategy was used to identify Scabin, a novel DNA-targeting enzyme from the plant pathogen 87.22 strain of Streptomyces scabies Scabin shares nearly 40% sequence identity with the Pierisin family of mono-ADP-ribosyltransferase toxins. Scabin was purified to homogeneity as a 22-kDa single-domain enzyme and was shown to possess high NAD(+)-glycohydrolase (Km (NAD) = 68 ± 3 µm; kcat = 94 ± 2 min(-1)) activity with an RSQXE motif; it was also shown to target deoxyguanosine and showed sigmoidal enzyme kinetics (K0.5(deoxyguanosine) = 302 ± 12 µm; kcat = 14 min(-1)). Mass spectrometry analysis revealed that Scabin labels the exocyclic amino group on guanine bases in either single-stranded or double-stranded DNA. Several small molecule inhibitors were identified, and the most potent compounds were found to inhibit the enzyme activity with Ki values ranging from 3 to 24 µm PJ34, a well known inhibitor of poly-ADP-ribosyltransferases, was shown to be the most potent inhibitor of Scabin. Scabin was crystallized, representing the first structure of a DNA-targeting mono-ADP-ribosyltransferase enzyme; the structures of the apo-form (1.45 Å) and with two inhibitors (P6-E, 1.4 Å; PJ34, 1.6 Å) were solved. These x-ray structures are also the first high resolution structures of the Pierisin subgroup of the mono-ADP-ribosyltransferase toxin family. A model of Scabin with its DNA substrate is also proposed.


Assuntos
ADP Ribose Transferases/metabolismo , Proteínas de Bactérias/metabolismo , Streptomyces/enzimologia , ADP Ribose Transferases/antagonistas & inibidores , ADP Ribose Transferases/química , Sequência de Aminoácidos , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Toxinas Bacterianas/química , Cristalografia por Raios X , DNA Bacteriano/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Cinética , Simulação de Dinâmica Molecular , Homologia de Sequência de Aminoácidos , Streptomyces/genética , Streptomyces/patogenicidade , Especificidade por Substrato
3.
Environ Microbiol ; 19(12): 5100-5116, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29124866

RESUMO

The toxin Plx2A is an important virulence factor of Paenibacillus larvae, the etiological agent of American Foulbrood, the most destructive bacterial disease of honey bees. Biochemical and functional analyses as well as the crystal structure of Plx2A revealed that it belongs to the C3 mono-ADP-ribosylating toxin subgroup. RhoA was identified as the cellular target of Plx2A activity. The kinetic parameters (KM , kcat ) were established for both the transferase and glycohydrolase reactions. When expressed in yeast, Plx2A was cytotoxic for eukaryotic cells and catalytic variants confirmed that the cytotoxicity of Plx2A depends on its enzymatic activity. The crystal structure of Plx2A was solved to 1.65 Å and confirmed that it is a C3-like toxin, although with a new molecular twist, it has a B-domain. A molecular model of the 'active' enzyme conformation in complex with NAD+ was produced by computational methods based on the recent structure of C3bot1 with RhoA. In murine macrophages, Plx2A induced actin cytoskeleton reorganization while in insect cells, vacuolization and the occurrence of bi-nucleated cells was observed. The latter is indicative of an inhibition of cytokinesis. All these cellular effects are consistent with Plx2A inhibiting the activity of RhoA by covalent modification.


Assuntos
ADP Ribose Transferases/metabolismo , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Abelhas/microbiologia , Macrófagos/patologia , Paenibacillus larvae/patogenicidade , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Animais , Catálise , Linhagem Celular , Camundongos , Modelos Moleculares , Estrutura Secundária de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Fatores de Virulência/metabolismo
4.
J Biol Chem ; 290(3): 1639-53, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25477523

RESUMO

C3larvin toxin was identified by a bioinformatic strategy as a putative mono-ADP-ribosyltransferase and a possible virulence factor from Paenibacillus larvae, which is the causative agent of American Foulbrood in honey bees. C3larvin targets RhoA as a substrate for its transferase reaction, and kinetics for both the NAD(+) (Km = 34 ± 12 µm) and RhoA (Km = 17 ± 3 µm) substrates were characterized for this enzyme from the mono-ADP-ribosyltransferase C3 toxin subgroup. C3larvin is toxic to yeast when expressed in the cytoplasm, and catalytic variants of the enzyme lost the ability to kill the yeast host, indicating that the toxin exerts its lethality through its enzyme activity. A small molecule inhibitor of C3larvin enzymatic activity was discovered called M3 (Ki = 11 ± 2 µm), and to our knowledge, is the first inhibitor of transferase activity of the C3 toxin family. C3larvin was crystallized, and its crystal structure (apoenzyme) was solved to 2.3 Å resolution. C3larvin was also shown to have a different mechanism of cell entry from other C3 toxins.


Assuntos
ADP Ribose Transferases/química , Proteínas de Bactérias/química , Paenibacillus/enzimologia , Proteína rhoA de Ligação ao GTP/química , Sequência de Aminoácidos , Toxinas Botulínicas/química , Catálise , Dicroísmo Circular , Biologia Computacional , Cristalização , Cristalografia por Raios X , Escherichia coli/metabolismo , Guanosina Trifosfato/química , Concentração Inibidora 50 , Dados de Sequência Molecular , NAD/química , Ligação Proteica , Conformação Proteica , Homologia de Sequência de Aminoácidos , Toxinas Biológicas/química , Fatores de Virulência
5.
Arch Biochem Biophys ; 608: 52-73, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27596846

RESUMO

Current evidence suggests that the closed-state membrane model for the channel-forming domain of colicin E1 involves eight amphipathic α-helices (helices I-VII and X) that adopt a two-dimensional arrangement on the membrane surface. Two central hydrophobic α-helices in colicin E1 (VIII and IX) adopt a transmembrane location-the umbrella model. Helices I and II have been shown to participate in the channel by forming a transmembrane segment (TM1) in the voltage-induced open channel state. Consequently, it is paramount to determine the relative location and orientation of helix I in the two-dimensional arrangement of the membrane. A new, low-resolution, three-dimensional model of the closed state of the colicin E1 channel was constructed based on FRET measurements between three naturally occurring Trp residues and three sites in helix I, in addition to previously reported FRET distances for the channel domain. Furthermore, a new mechanism for the channel integration process involving the transition of the soluble to membrane-bound form is presented based on a plethora of kinetic data for this process.


Assuntos
Colicinas/química , Proteínas de Escherichia coli/química , Escherichia coli/química , Transferência Ressonante de Energia de Fluorescência , Anisotropia , Compostos Bicíclicos com Pontes/química , Membrana Celular/química , Cisteína/química , Concentração de Íons de Hidrogênio , Cinética , Domínios Proteicos , Estrutura Secundária de Proteína , Triptofano/química
6.
Biochemistry ; 54(38): 5920-36, 2015 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-26352925

RESUMO

Vis toxin was identified by a bioinformatics strategy as a putative virulence factor produced by Vibrio splendidus with mono-ADP-ribosyltransferase activity. Vis was purified to homogeneity as a 28 kDa single-domain enzyme and was shown to possess NAD(+)-glycohydrolase [KM(NAD(+)) = 276 ± 12 µM] activity and with an R-S-E-X-E motif; it targets arginine-related compounds [KM(agmatine) = 272 ± 18 mM]. Mass spectrometry analysis revealed that Vis labels l-arginine with ADP-ribose from the NAD(+) substrate at the amino nitrogen of the guanidinium side chain. Vis is toxic to yeast when expressed in the cytoplasm under control of the CUP1 promotor, and catalytic variants lost the ability to kill the yeast host, indicating that the toxin exerts its lethality through its enzyme activity. Several small molecule inhibitors were identified from a virtual screen, and the most potent compounds were found to inhibit the transferase activity of the enzyme with Ki values ranging from 25 to 134 µM. Inhibitor compound M6 bears the necessary attributes of a solid candidate as a lead compound for therapeutic development. Vis toxin was crystallized, and the structures of the apoenzyme (1.4 Å) and the enzyme bound with NAD(+) (1.8 Å) and with the M6 inhibitor (1.5 Å) were determined. The structures revealed that Vis represents a new subgroup within the mono-ADP-ribosyltransferase toxin family.


Assuntos
ADP Ribose Transferases/química , Toxinas Bacterianas/química , Vibrio/enzimologia , ADP Ribose Transferases/metabolismo , Adenosina Difosfato Ribose/metabolismo , Sequência de Aminoácidos , Toxinas Bacterianas/metabolismo , Cristalografia por Raios X , Guanidina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , NAD+ Nucleosidase/química , NAD+ Nucleosidase/metabolismo , Conformação Proteica , Alinhamento de Sequência , Vibrio/química , Vibrio/metabolismo
7.
J Mol Recognit ; 28(9): 539-52, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25756608

RESUMO

Cholix toxin from Vibrio cholerae is a novel mono-ADP-ribosyltransferase (mART) toxin that shares structural and functional properties with Pseudomonas aeruginosa exotoxin A and Corynebacterium diphtheriae diphtheria toxin. Herein, we have used the high-resolution X-ray structure of full-length cholix toxin in the apo form, NAD(+) bound, and 10 structures of the cholix catalytic domain (C-domain) complexed with several strong inhibitors of toxin enzyme activity (NAP, PJ34, and the P-series) to study the binding mode of the ligands. A pharmacophore model based on the active pose of NAD(+) was compared with the active conformation of the inhibitors, which revealed a cationic feature in the side chain of the inhibitors that may determine the active pose. Moreover, a conformational search was conducted for the missing coordinates of one of the main active-site loops (R-loop). The resulting structural models were used to evaluate the interaction energies and for 3D-QSAR modeling. Implications for a rational drug design approach for mART toxins were derived.


Assuntos
Fatores de Ribosilação do ADP/antagonistas & inibidores , Fatores de Ribosilação do ADP/química , Toxinas Bacterianas/antagonistas & inibidores , Toxinas Bacterianas/química , Inibidores Enzimáticos/farmacologia , Relação Estrutura-Atividade , Vibrio cholerae/metabolismo , Fatores de Ribosilação do ADP/metabolismo , Toxinas Bacterianas/metabolismo , Domínio Catalítico , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Modelos Moleculares , NAD/química , NAD/metabolismo
8.
J Biol Chem ; 288(7): 5136-48, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23264635

RESUMO

The pre-channel state of helices 6, 7, and 10 (Val(447)-Gly(475) and Ile(508)-Ile(522)) of colicin E1 was investigated by a site-directed fluorescence labeling technique. A total of 44 cysteine variants were purified and covalently labeled with monobromobimane fluorescent probe. A variety of fluorescence properties of the bimane fluorophore were measured for both the soluble and membrane-bound states of the channel peptide, including the fluorescence emission maximum, fluorescence anisotropy, and membrane bilayer penetration depth. Using site-directed fluorescence labeling combined with our novel helical periodicity analysis method, the data revealed that helices 6, 7, and 10 are separate amphipathic α-helices with a calculated periodicity of T = 3.34 ± 0.08 for helix 6, T = 3.56 ± 0.03 for helix 7, and T = 2.99 ± 0.12 for helix 10 in the soluble state. In the membrane-bound state, the helical periodicity was determined to be T = 3.00 ± 0.15 for helix 6, T = 3.68 ± 0.03 for helix 7, and T = 3.47 ± 0.04 for helix 10. Dual fluorescence quencher analysis showed that both helices 6 and 7 adopt a tilted topology that correlates well with the analysis based on the fluorescence anisotropy profile. These data provide further support for the umbrella model of the colicin E1 channel domain.


Assuntos
Colicinas/química , Escherichia coli/metabolismo , Sequência de Aminoácidos , Anisotropia , Bacteriocinas/química , Cisteína/química , Citoplasma/metabolismo , Dissulfetos , Transferência Ressonante de Energia de Fluorescência/métodos , Microscopia de Fluorescência/métodos , Conformação Molecular , Dados de Sequência Molecular , Mutagênese , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Temperatura
9.
J Biol Chem ; 287(25): 21176-88, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22535961

RESUMO

Certain Vibrio cholerae strains produce cholix, a potent protein toxin that has diphthamide-specific ADP-ribosyltransferase activity against eukaryotic elongation factor 2. Here we present a 1.8 Å crystal structure of cholix in complex with its natural substrate, nicotinamide adenine dinucleotide (NAD(+)). We also substituted hallmark catalytic residues by site-directed mutagenesis and analyzed both NAD(+) binding and ADP-ribosyltransferase activity using a fluorescence-based assay. These data are the basis for a new kinetic model of cholix toxin activity. Further, the new structural data serve as a reference for continuing inhibitor development for this toxin class.


Assuntos
Fatores de Ribosilação do ADP , Toxinas Bacterianas , Modelos Biológicos , Modelos Moleculares , NAD , Vibrio cholerae/enzimologia , Fatores de Ribosilação do ADP/química , Fatores de Ribosilação do ADP/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Cristalografia por Raios X , NAD/química , NAD/metabolismo , Estrutura Terciária de Proteína
10.
Biochemistry ; 50(22): 4830-42, 2011 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-21528912

RESUMO

The membrane topology of the colicin E1 channel domain was studied by fluorescence resonance energy transfer (FRET). The FRET involved a genetically encoded fluorescent amino acid (coumarin) as the donor and a selectively labeled cysteine residue tethered with DABMI (4-(dimethylamino)phenylazophenyl-4'-maleimide) as the FRET acceptor. The fluorescent coumarin residue was incorporated into the protein via an orthogonal tRNA/aminoacyl-tRNA synthetase pair that allowed selective incorporation into any site within the colicin channel domain. Each variant harbored a stop (TAG) mutation for coumarin incorporation and a cysteine (TGT) mutation for DABMI attachment. Six interhelical distances within helices 1-6 were determined using FRET analysis for both the soluble and membrane-bound states. The FRET data showed large changes in the interhelical distances among helices 3-6 upon membrane association providing new insight into the membrane-bound structure of the channel domain. In general, the coumarin-DABMI FRET interhelical efficiencies decreased upon membrane binding, building upon the umbrella model for the colicin channel. A tentative model for the closed state of the channel domain was developed based on current and previously published FRET data. The model suggests circular arrangement of helices 1-7 in a clockwise direction from the extracellular side and membrane interfacial association of helices 1, 6, 7, and 10 around the central transmembrane hairpin formed by helices 8 and 9.


Assuntos
Colicinas/química , Aminoacil-tRNA Sintetases , Colicinas/genética , Colicinas/metabolismo , Cumarínicos/química , Cisteína/metabolismo , Escherichia coli/metabolismo , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/metabolismo , Bicamadas Lipídicas/química , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Estrutura Secundária de Proteína , p-Dimetilaminoazobenzeno/análogos & derivados , p-Dimetilaminoazobenzeno/química , p-Dimetilaminoazobenzeno/metabolismo
11.
Sci Rep ; 11(1): 7429, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33795718

RESUMO

The 2019 novel coronavirus pandemic caused by SARS-CoV-2 remains a serious health threat to humans and there is an urgent need to develop therapeutics against this deadly virus. Recent scientific evidences have suggested that the main protease (Mpro) enzyme in SARS-CoV-2 can be an ideal drug target due to its crucial role in the viral replication and transcription processes. Therefore, there are ongoing research efforts to identify drug candidates against SARS-CoV-2 Mpro that resulted in hundreds of X-ray crystal structures of ligand-bound Mpro complexes in the Protein Data Bank (PDB) describing the interactions of different fragment chemotypes within different sites of the Mpro. In this work, we performed rigorous molecular dynamics (MD) simulation of 62 reversible ligand-Mpro complexes in the PDB to gain mechanistic insights about their interactions at the atomic level. Using a total of over 3 µs long MD trajectories, we characterized different pockets in the apo Mpro structure, and analyzed the dynamic interactions and binding affinity of ligands within those pockets. Our results identified the key residues that stabilize the ligands in the catalytic sites and other pockets of Mpro. Our analyses unraveled the role of a lateral pocket in the catalytic site in Mpro that is critical for enhancing the ligand binding to the enzyme. We also highlighted the important contribution from HIS163 in the lateral pocket towards ligand binding and affinity against Mpro through computational mutation analyses. Further, we revealed the effects of explicit water molecules and Mpro dimerization in the ligand association with the target. Thus, comprehensive molecular-level insights gained from this work can be useful to identify or design potent small molecule inhibitors against SARS-CoV-2 Mpro.


Assuntos
Simulação de Dinâmica Molecular , Inibidores de Proteases/química , SARS-CoV-2/metabolismo , Proteínas da Matriz Viral/antagonistas & inibidores , Sítios de Ligação , COVID-19/patologia , COVID-19/virologia , Domínio Catalítico , Bases de Dados de Proteínas , Humanos , Ligantes , Mutagênese Sítio-Dirigida , Análise de Componente Principal , Inibidores de Proteases/metabolismo , SARS-CoV-2/isolamento & purificação , Termodinâmica , Proteínas da Matriz Viral/metabolismo
12.
Toxins (Basel) ; 13(1)2020 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-33375750

RESUMO

Mono-ADP-ribosyltransferase toxins are often key virulence factors produced by pathogenic bacteria as tools to compromise the target host cell. These toxins are enzymes that use host cellular NAD+ as the substrate to modify a critical macromolecule target in the host cell machinery. This post-translational modification of the target macromolecule (usually protein or DNA) acts like a switch to turn the target activity on or off resulting in impairment of a critical process or pathway in the host. One approach to stymie bacterial pathogens is to curtail the toxic action of these factors by designing small molecules that bind tightly to the enzyme active site and prevent catalytic function. The inactivation of these toxins/enzymes is targeted for the site of action within the host cell and small molecule therapeutics can function as anti-virulence agents by disarming the pathogen. This represents an alternative strategy to antibiotic therapy with the potential as a paradigm shift that may circumvent multi-drug resistance in the offending microbe. In this review, work that has been accomplished during the past two decades on this approach to develop anti-virulence compounds against mono-ADP-ribosyltransferase toxins will be discussed.


Assuntos
ADP Ribose Transferases/toxicidade , Antídotos , Toxinas Bacterianas , Fatores de Virulência/antagonistas & inibidores , Animais , Humanos , Fatores de Virulência/metabolismo , Fatores de Virulência/toxicidade
13.
Biosci Rep ; 40(1)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31844879

RESUMO

C3larvinA is a putative virulence factor produced by Paenibacillus larvae enterobacterial-repetitive-intergenic-consensus (ERIC) III/IV (strain 11-8051). Biochemical, functional and structural analyses of C3larvinA revealed that it belongs to the C3-like mono-ADP-ribosylating toxin subgroup. Mammalian RhoA was the target substrate for its transferase activity suggesting that it may be the biological target of C3larvinA. The kinetic parameters of the NAD+ substrate for the transferase (KM = 75 ± 10 µM) and glycohydrolase (GH) (KM = 107 ± 20 µM) reactions were typical for a C3-like bacterial toxin, including the Plx2A virulence factor from Paenibacillus larvae ERIC I. Upon cytoplasmic expression in yeast, C3larvinA caused a growth-defective phenotype indicating that it is an active C3-like toxin and is cytotoxic to eukaryotic cells. The catalytic variant of the Q187-X-E189 motif in C3larvinA showed no cytotoxicity toward yeast confirming that the cytotoxicity of this factor depends on its enzymatic activity. A homology consensus model of C3larvinA with NAD+ substrate was built on the structure of Plx2A, provided additional confirmation that C3larvinA is a member of the C3-like mono-ADP-ribosylating toxin subgroup. A homology model of C3larvinA with NADH and RhoA was built on the structure of the C3cer-NADH-RhoA complex which provided further evidence that C3larvinA is a C3-like toxin that shares an identical catalytic mechanism with C3cer from Bacillus cereus. C3larvinA induced actin cytoskeleton reorganization in murine macrophages, whereas in insect cells, vacuolization and bi-nucleated cells were observed. These cellular effects are consistent with C3larvinA disrupting RhoA function by covalent modification that is shared among C3-like bacterial toxins.


Assuntos
ADP Ribose Transferases/metabolismo , Toxinas Bacterianas/metabolismo , Abelhas/microbiologia , Paenibacillus larvae/enzimologia , Fatores de Virulência/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , ADP Ribose Transferases/química , ADP Ribose Transferases/genética , Citoesqueleto de Actina/enzimologia , Animais , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Interações Hospedeiro-Patógeno , Cinética , Macrófagos/enzimologia , Mutação , Paenibacillus larvae/genética , Paenibacillus larvae/patogenicidade , Conformação Proteica , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Células Sf9 , Spodoptera , Relação Estrutura-Atividade , Especificidade por Substrato , Virulência , Fatores de Virulência/química , Fatores de Virulência/genética , Proteína rhoA de Ligação ao GTP/química
14.
Arch Biochem Biophys ; 492(1-2): 17-28, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19818729

RESUMO

P-glycoprotein (ABCB1) is an ATP-driven efflux pump which binds drugs within a large flexible binding pocket. Intrinsic Trp fluorescence was used to probe the interactions of LDS-751 (2-[4-(4-[dimethylamino]phenyl)-1,3-butadienyl]-3-ethylbenzo-thiazolium perchlorate) with purified P-glycoprotein, using steady-state/lifetime measurements and collisional quenching. The fast decay component of P-glycoprotein intrinsic fluorescence (tau(1)=0.97 ns) was unaffected by LDS-751 binding, while the slow decay component (tau(2)=4.02 ns) was quenched by dynamic and static mechanisms. Both the wavelength-dependence of the decay kinetics, and the time-resolved emission spectra, suggested the existence of excited-state relaxation processes within the protein matrix on the nanosecond time-scale, which were altered by LDS-751 binding. The fast decay component, which is more solvent-exposed, can be attributed to cytosolic/extracellular Trp residues, while the slow decay component likely arises from more buried transmembrane Trp residues. Interaction of a drug with the binding pocket of P-glycoprotein thus affects its molecular structure and fast dynamics.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/química , Corantes Fluorescentes/química , Triptofano/química , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Citosol/química , Interações Medicamentosas , Espaço Extracelular/química , Corantes Fluorescentes/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Camundongos , Compostos Orgânicos/química , Compostos Orgânicos/metabolismo , Ligação Proteica , Espectrometria de Fluorescência , Relação Estrutura-Atividade
15.
Arch Biochem Biophys ; 489(1-2): 55-61, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19631607

RESUMO

The plasma membrane Ca(2+)-ATPase (PMCA) plays a key role in the regulation of the intracellular Ca(2+) concentration. Ethanol stimulates this Ca(2+) pump in an isoform-specific manner. On search for a physiological molecule that could mimic the effect of ethanol, we have previously demonstrated that some sphingolipids containing free "hydroxyl" groups, like ceramide, are able to stimulate the PMCA. Since diacylglycerol (DAG) structurally shares some characteristics with ceramide, we evaluate its effect on the PMCA. We demonstrated that DAG is a potent stimulator of this enzyme. The activation induced is additive to that produced by calmodulin, protein-kinase C and ethanol, which implies that DAG interacts with the PMCA through a different mechanism. Additionally, by different fluorescent approaches, we demonstrated a direct binding between PMCA and DAG. The results obtained in this work strongly suggest that DAG is a novel effector of the PMCA, acting by a direct interaction.


Assuntos
Cálcio/química , Diglicerídeos/química , Membrana Eritrocítica/enzimologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/química , Cálcio/metabolismo , Calmodulina/química , Calmodulina/metabolismo , Ceramidas/química , Ceramidas/metabolismo , Diglicerídeos/metabolismo , Ativação Enzimática/fisiologia , Etanol/química , Etanol/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Ligação Proteica/fisiologia , Proteína Quinase C/química , Proteína Quinase C/metabolismo
16.
Toxins (Basel) ; 11(6)2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31234283

RESUMO

The C3-like toxins are single-domain proteins that represent a minimal mono-ADP-ribosyl transferase (mART) enzyme with a simple model scaffold for the entire cholera toxin (CT)-group. These proteins possess a single (A-domain) that modifies Rho proteins. In contrast, C2-like toxins require a binding/translocation partner (B-component) for intoxication. These are A-only toxins that contain the E-x-E motif, modify G-actin, but are two-domains with a C-domain possessing enzymatic activity. The N-domain of the C2-like toxins is unstructured, and its function is currently unknown. A sequence-structure-function comparison was performed on the N-terminal region of the mART domain of the enzymatic component of the CT toxin group in the CATCH fold (3.90.210.10). Special consideration was given to the N-domain distal segment, the α-lobe (α1-α4), and its different roles in these toxin sub-groups. These results show that the role of the N-terminal α-lobe is to provide a suitable configuration (i) of the α2-α3 helices to feature the α3-motif that has a role in NAD+ substrate binding and possibly in the interaction with the protein target; (ii) the α3-α4 helices to provide the α3/4-loop with protein-protein interaction capability; and (iii) the α1-Ntail that features specialized motif(s) according to the toxin type (A-only or A-B toxins) exhibiting an effect on the catalytic activity via the ARTT-loop, with a role in the inter-domain stability, and with a function in the binding and/or translocation steps during the internalization process.


Assuntos
ADP Ribose Transferases/química , Sequência de Aminoácidos , Toxinas Bacterianas , Simulação por Computador , Modelos Moleculares , Conformação Proteica , Alinhamento de Sequência
17.
PLoS One ; 13(3): e0194425, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29543870

RESUMO

Scabin is a mono-ADP-ribosyltransferase enzyme and is a putative virulence factor produced by the plant pathogen, Streptomyces scabies. Previously, crystal structures of Scabin were solved in the presence and absence of substrate analogues and inhibitors. Herein, experimental (hydrogen-deuterium exchange), simulated (molecular dynamics), and theoretical (Gaussian Network Modeling) approaches were systematically applied to study the dynamics of apo-Scabin in the context of a Scabin·NAD+·DNA model. MD simulations revealed that the apo-Scabin solution conformation correlates well with the X-ray crystal structure, beyond the conformation of the exposed, mobile regions. In turn, the MD fluctuations correspond with the crystallographic B-factors, with the fluctuations derived from a Gaussian network model, and with the experimental H/D exchange rates. An Essential Dynamics Analysis identified the dynamic aspects of the toxin as a crab-claw-like mechanism of two topological domains, along with coupled deformations of exposed motifs. The "crab-claw" movement resembles the motion of C3-like toxins and emerges as a property of the central ß scaffold of catalytic single domain toxins. The exposure and high mobility of the cis side motifs in the Scabin ß-core suggest involvement in DNA substrate binding. A ternary Scabin·NAD+·DNA model was produced via an independent docking methodology, where the intermolecular interactions correspond to the region of high mobility identified by dynamics analyses and agree with binding and kinetic data reported for wild-type and Scabin variants. Based on data for the Pierisin-like toxin group, the sequence motif Rß1-RLa-NLc-STTß2-WPN-WARTT-(QxE)ARTT emerges as a catalytic signature involved in the enzymatic activity of these DNA-acting toxins. However, these results also show that Scabin possesses a unique DNA-binding motif within the Pierisin-like toxin group.


Assuntos
ADP Ribose Transferases/metabolismo , Toxinas Bacterianas/metabolismo , DNA/metabolismo , Simulação de Dinâmica Molecular , Streptomyces/metabolismo , ADP Ribose Transferases/química , ADP Ribose Transferases/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Sítios de Ligação/genética , Domínio Catalítico , Cristalografia por Raios X , DNA/química , DNA/genética , Cinética , NAD/química , NAD/metabolismo , Ligação Proteica , Streptomyces/genética , Especificidade por Substrato
18.
J Biomol Struct Dyn ; 34(12): 2537-2560, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26610041

RESUMO

C3larvin toxin is a new member of the C3 class of the mono-ADP-ribosyltransferase toxin family. The C3 toxins are known to covalently modify small G-proteins, e.g. RhoA, impairing their function, and serving as virulence factors for an offending pathogen. A full-length X-ray structure of C3larvin (2.3 Å) revealed that the characteristic mixed α/ß fold consists of a central ß-core flanked by two helical regions. Topologically, the protein can be separated into N and C lobes, each formed by a ß-sheet and an α-motif, and connected by exposed loops involved in the recognition, binding, and catalysis of the toxin/enzyme, i.e. the ADP-ribosylation turn-turn and phosphate-nicotinamide PN loops. Herein, we provide two new C3larvin X-ray structures and present a systematic study of the toxin dynamics by first analyzing the experimental variability of the X-ray data-set followed by contrasting those results with theoretical predictions based on Elastic Network Models (GNM and ANM). We identify residues that participate in the stability of the N-lobe, putative hinges at loop residues, and energy-favored deformation vectors compatible with conformational changes of the key loops and 3D-subdomains (N/C-lobes), among the X-ray structures. We analyze a larger ensemble of known C3bot1 conformations and conclude that the characteristic 'crab-claw' movement may be driven by the main intrinsic modes of motion. Finally, via computational simulations, we identify harmonic and anharmonic fluctuations that might define the C3larvin 'native state.' Implications for docking protocols are derived.


Assuntos
ADP Ribose Transferases/química , Toxinas Bacterianas/química , Modelos Moleculares , Dobramento de Proteína , Estrutura Secundária de Proteína , Sequência de Aminoácidos , Cristalografia por Raios X , Simulação de Dinâmica Molecular , Relação Estrutura-Atividade
19.
Toxins (Basel) ; 7(8): 2757-72, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26213968

RESUMO

The cholix toxin gene (chxA) was first identified in V. cholerae strains in 2007, and the protein was identified by bioinformatics analysis in 2008. It was identified as the third member of the diphtheria toxin group of mono-ADP-ribosyltransferase toxins along with P. aeruginosa exotoxin A and C. diphtheriae diphtheria toxin. Our group determined the structure of the full-length, three-domain cholix toxin at 2.1 Å and its C-terminal catalytic domain (cholixc) at 1.25 Å resolution. We showed that cholix toxin is specific for elongation factor 2 (diphthamide residue), similar to exotoxin A and diphtheria toxin. Cholix toxin possesses molecular features required for infection of eukaryotes by receptor-mediated endocytosis, translocation to the host cytoplasm and inhibition of protein synthesis. More recently, we also solved the structure of full-length cholix toxin in complex with NAD+ and proposed a new kinetic model for cholix enzyme activity. In addition, we have taken a computational approach that revealed some important properties of the NAD+-binding pocket at the residue level, including the role of crystallographic water molecules in the NAD+ substrate interaction. We developed a pharmacophore model of cholix toxin, which revealed a cationic feature in the side chain of cholix toxin active-site inhibitors that may determine the active pose. Notably, several recent reports have been published on the role of cholix toxin as a major virulence factor in V. cholerae (non-O1/O139 strains). Additionally, FitzGerald and coworkers prepared an immunotoxin constructed from domains II and III as a cancer treatment strategy to complement successful immunotoxins derived from P. aeruginosa exotoxin A.


Assuntos
Fatores de Ribosilação do ADP , Toxinas Bacterianas , Fatores de Virulência , ADP Ribose Transferases , Fatores de Ribosilação do ADP/química , Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Fatores de Ribosilação do ADP/farmacologia , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Imunotoxinas/química , Imunotoxinas/genética , Imunotoxinas/metabolismo , Imunotoxinas/farmacologia , NAD/metabolismo , Fatores de Virulência/química , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Fatores de Virulência/farmacologia
20.
J Biomol Struct Dyn ; 33(11): 2452-68, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25559625

RESUMO

Cholix toxin from Vibrio cholerae is the third member of the diphtheria toxin (DT) group of mono-ADP-ribosyltransferase (mART) bacterial toxins. It shares structural and functional properties with Pseudomonas aeruginosa exotoxin A and Corynebacterium diphtheriae DT. Cholix toxin is an important model for the development of antivirulence approaches and therapeutics against these toxins from pathogenic bacteria. Herein, we have used the high-resolution X-ray structure of full-length cholix complexed with NAD(+) to describe the properties of the NAD(+)-binding pocket at the residue level, including the role of crystallographic water molecules in the NAD(+) substrate interaction. The full-length apo cholix structure is used to describe the putative NAD(+)-binding site(s) and to correlate biochemical with crystallographic data to study the stoichiometry and orientation of bound NAD(+) molecules. We quantitatively describe the NAD(+) substrate interactions on a residue basis for the main 22 pocket residues in cholixf, a glycerol and 5 contact water molecules as part of the recognition surface by the substrate according to the conditions of crystallization. In addition, the dynamic properties of an in silico version of the catalytic domain were investigated in order to understand the lack of electronic density for one of the main flexible loops (R-loop) in the pocket of X-ray complexes. Implications for a rational drug design approach for mART toxins are derived.


Assuntos
Fatores de Ribosilação do ADP/química , Toxinas Bacterianas/química , Domínio Catalítico , Modelos Moleculares , Fatores de Ribosilação do ADP/metabolismo , Toxinas Bacterianas/metabolismo , Sítios de Ligação , Ligação de Hidrogênio , Ligantes , Simulação de Dinâmica Molecular , NAD/química , NAD/metabolismo , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Relação Quantitativa Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA