Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 299(1): 102795, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36528064

RESUMO

Shiga toxin 2a (Stx2a) is the virulence factor of enterohemorrhagic Escherichia coli. The catalytic A1 subunit of Stx2a (Stx2A1) interacts with the ribosomal P-stalk for loading onto the ribosome and depurination of the sarcin-ricin loop, which halts protein synthesis. Because of the intrinsic flexibility of the P-stalk, a structure of the Stx2a-P-stalk complex is currently unknown. We demonstrated that the native P-stalk pentamer binds to Stx2a with nanomolar affinity, and we employed cryo-EM to determine a structure of the 72 kDa Stx2a complexed with the P-stalk. The structure identifies Stx2A1 residues involved in binding and reveals that Stx2a is anchored to the P-stalk via only the last six amino acids from the C-terminal domain of a single P-protein. For the first time, the cryo-EM structure shows the loop connecting Stx2A1 and Stx2A2, which is critical for activation of the toxin. Our principal component analysis of the cryo-EM data reveals the intrinsic dynamics of the Stx2a-P-stalk interaction, including conformational changes in the P-stalk binding site occurring upon complex formation. Our computational analysis unveils the propensity for structural rearrangements within the C-terminal domain, with its C-terminal six amino acids transitioning from a random coil to an α-helix upon binding to Stx2a. In conclusion, our cryo-EM structure sheds new light into the dynamics of the Stx2a-P-stalk interaction and indicates that the binding interface between Stx2a and the P-stalk is the potential target for drug discovery.


Assuntos
Escherichia coli O157 , Ribossomos , Toxina Shiga II , Aminoácidos/metabolismo , Microscopia Crioeletrônica , Ribossomos/metabolismo , Toxina Shiga II/química , Toxina Shiga II/metabolismo , Escherichia coli O157/química
2.
Anal Biochem ; 692: 115580, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38825159

RESUMO

Ricin is one of the most toxic substances known and a type B biothreat agent. Shiga toxins (Stxs) produced by E. coli (STEC) and Shigella dysenteriae are foodborne pathogens. There is no effective therapy against ricin or STEC and there is an urgent need for inhibitors. Ricin toxin A subunit (RTA) and A1 subunit of Stx2a (Stx2A1) bind to the C-terminal domain (CTD) of the ribosomal P-stalk proteins to depurinate the sarcin/ricin loop. Modulation of toxin-ribosome interactions has not been explored as a strategy for inhibition. Therefore, development of assays that detect inhibitors targeting toxin-ribosome interactions remains a critical need. Here we describe a fluorescence anisotropy (FA)-based competitive binding assay using a BODIPY-TMR labeled 11-mer peptide (P11) derived from the P-stalk CTD to measure the binding affinity of peptides ranging from 3 to 11 amino acids for the P-stalk pocket of RTA and Stx2A1. Comparison of the affinity with the surface plasmon resonance (SPR) assay indicated that although the rank order was the same by both methods, the FA assay could differentiate better between peptides that show nonspecific interactions by SPR. The FA assay detects only interactions that compete with the labeled P11 and can validate inhibitor specificity and mechanism of action.


Assuntos
Polarização de Fluorescência , Ribossomos , Ricina , Ricina/antagonistas & inibidores , Ricina/metabolismo , Ricina/química , Polarização de Fluorescência/métodos , Ribossomos/metabolismo , Ressonância de Plasmônio de Superfície , Toxina Shiga/antagonistas & inibidores , Toxina Shiga/metabolismo , Toxina Shiga/química , Ligação Competitiva , Ligação Proteica , Toxina Shiga II/antagonistas & inibidores , Toxina Shiga II/metabolismo , Toxina Shiga II/química
3.
J Biol Chem ; 296: 100299, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33460651

RESUMO

The human Gb3/CD77 synthase, encoded by the A4GALT gene, is an unusually promiscuous glycosyltransferase. It synthesizes the Galα1→4Gal linkage on two different glycosphingolipids (GSLs), producing globotriaosylceramide (Gb3, CD77, Pk) and the P1 antigen. Gb3 is the major receptor for Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli. A single amino acid substitution (p.Q211E) ramps up the enzyme's promiscuity, rendering it able to attach Gal both to another Gal residue and to GalNAc, giving rise to NOR1 and NOR2 GSLs. Human Gb3/CD77 synthase was long believed to transfer Gal only to GSL acceptors, therefore its GSL products were, by default, considered the only human Stx receptors. Here, using soluble, recombinant human Gb3/CD77 synthase and p.Q211E mutein, we demonstrate that both enzymes can synthesize the P1 glycotope (terminal Galα1→4Galß1→4GlcNAc-R) on a complex type N-glycan and a synthetic N-glycoprotein (saposin D). Moreover, by transfection of CHO-Lec2 cells with vectors encoding human Gb3/CD77 synthase and its p.Q211E mutein, we demonstrate that both enzymes produce P1 glycotopes on N-glycoproteins, with the mutein exhibiting elevated activity. These P1-terminated N-glycoproteins are recognized by Stx1 but not Stx2 B subunits. Finally, cytotoxicity assays show that Stx1 can use P1 N-glycoproteins produced in CHO-Lec2 cells as functional receptors. We conclude that Stx1 can recognize and use P1 N-glycoproteins in addition to its canonical GSL receptors to enter and kill the cells, while Stx2 can use GSLs only. Collectively, these results may have important implications for our understanding of the Shiga toxin pathology.


Assuntos
Galactosiltransferases/química , Globosídeos/química , Toxina Shiga I/química , Triexosilceramidas/química , Acetilgalactosamina/química , Acetilgalactosamina/metabolismo , Acetilglucosamina/química , Acetilglucosamina/metabolismo , Animais , Sítios de Ligação , Células CHO , Sequência de Carboidratos , Cricetulus , Escherichia coli Êntero-Hemorrágica/química , Escherichia coli Êntero-Hemorrágica/patogenicidade , Galactose/química , Galactose/metabolismo , Galactosiltransferases/genética , Galactosiltransferases/metabolismo , Expressão Gênica , Globosídeos/biossíntese , Globosídeos/metabolismo , Glucose/química , Glucose/metabolismo , Humanos , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Toxina Shiga I/metabolismo , Toxina Shiga II/química , Toxina Shiga II/metabolismo , Triexosilceramidas/biossíntese
4.
J Biol Chem ; 295(46): 15588-15596, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-32878986

RESUMO

The principal virulence factor of human pathogenic enterohemorrhagic Escherichia coli is Shiga toxin (Stx). Shiga toxin 2a (Stx2a) is the subtype most commonly associated with severe disease outcomes such as hemorrhagic colitis and hemolytic uremic syndrome. The catalytic A1 subunit (Stx2A1) binds to the conserved elongation factor binding C-terminal domain (CTD) of ribosomal P stalk proteins to inhibit translation. Stx2a holotoxin also binds to the CTD of P stalk proteins because the ribosome-binding site is exposed. We show here that Stx2a binds to an 11-mer peptide (P11) mimicking the CTD of P stalk proteins with low micromolar affinity. We cocrystallized Stx2a with P11 and defined their interactions by X-ray crystallography. We found that the last six residues of P11 inserted into a shallow pocket on Stx2A1 and interacted with Arg-172, Arg-176, and Arg-179, which were previously shown to be critical for binding of Stx2A1 to the ribosome. Stx2a formed a distinct P11-binding mode within a different surface pocket relative to ricin toxin A subunit and trichosanthin, suggesting different ribosome recognition mechanisms for each ribosome inactivating protein (RIP). The binding mode of Stx2a to P11 is also conserved among the different Stx subtypes. Furthermore, the P stalk protein CTD is flexible and adopts distinct orientations and interaction modes depending on the structural differences between the RIPs. Structural characterization of the Stx2a-ribosome complex is important for understanding the role of the stalk in toxin recruitment to the sarcin/ricin loop and may provide a new target for inhibitor discovery.


Assuntos
Peptídeos/metabolismo , Proteínas Ribossômicas/química , Toxina Shiga II/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Humanos , Simulação de Dinâmica Molecular , Peptídeos/química , Ligação Proteica , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo , Ricina/química , Ricina/metabolismo , Toxina Shiga II/química , Tricosantina/química , Tricosantina/metabolismo
5.
Biochem Biophys Res Commun ; 557: 247-253, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33894410

RESUMO

Accumulation of amyloid-ß peptide (Aß) in neuronal cells and in the extracellular regions in the brain is a major cause of Alzheimer's disease (AD); therefore, inhibition of Aß accumulation offers a promising approach for therapeutic strategies against AD. Aß is produced by sequential proteolysis of amyloid precursor protein (APP) in late/recycling endosomes after endocytosis of APP located in the plasma membrane. Aß is then released from cells in a free form or in an exosome-bound form. Shiga toxin (Stx) is a major virulence factor of enterohemorrhagic Escherichia coli. Recently, we found that one of the Stx subtypes, Stx2a, has a unique intracellular transport route after endocytosis through its receptor-binding B-subunit. A part of Stx2a can be transported to late/recycling endosomes and then degraded in a lysosomal acidic compartment, although in general Stx is transported to the Golgi and then to the endoplasmic reticulum in a retrograde manner. In this study, we found that treatment of APP-expressing cells with a mutant Stx2a (mStx2a), lacking cytotoxic activity because of mutations in the catalytic A-subunit, stimulated the transport of APP to the acidic compartment, which led to degradation of APP and a reduction in the amount of Aß. mStx2a-treatment also inhibited the extracellular release of Aß. Therefore, mStx2a may provide a new strategy to inhibit the production of Aß by modulating the intracellular transport of APP.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Membrana Celular/efeitos dos fármacos , Endossomos/metabolismo , Lisossomos/metabolismo , Transporte Proteico/efeitos dos fármacos , Toxina Shiga II/farmacologia , Animais , Células CHO , Domínio Catalítico/genética , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Cricetulus , Globosídeos/química , Humanos , Mutação , Fosfatidilcolinas/química , Proteínas Recombinantes , Toxina Shiga II/química , Toxina Shiga II/genética , Triexosilceramidas/química
6.
Cell Microbiol ; 21(5): e13000, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30578712

RESUMO

Hemolytic uremic syndrome (eHUS) is a severe complication of human infections with Shiga toxins (Stxs)-producing Escherichia coli. A key step in the pathogenesis of eHUS is the interaction of Stxs with blood components before the targeting of renal endothelial cells. Here, we show that a single proteolytic cleavage in the Stx2a A-subunit, resulting into two fragments (A1 and A2) linked by a disulfide bridge (cleaved Stx2a), dictates different binding abilities. Uncleaved Stx2a was confirmed to bind to human neutrophils and to trigger leukocyte/platelet aggregate formation, whereas cleaved Stx2a was ineffective. Conversely, binding of complement factor H was confirmed for cleaved Stx2a and not for uncleaved Stx2a. It is worth noting that uncleaved and cleaved Stx2a showed no differences in cytotoxicity for Vero cells or Raji cells, structural conformation, and contaminating endotoxin. These results have been obtained by comparing two Stx2a batches, purified in different laboratories by using different protocols, termed Stx2a(cl; cleaved toxin, Innsbruck) and Stx2a(uncl; uncleaved toxin, Bologna). Stx2a(uncl) behaved as Stx2a(cl) after mild trypsin treatment. In this light, previous controversial results obtained with purified Stx2a has to be critically re-evaluated; furthermore, characterisation of the structure of circulating Stx2a is mandatory to understand eHUS-pathogenesis and to develop therapeutic approaches.


Assuntos
Escherichia coli/química , Toxina Shiga II/química , Toxina Shiga II/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Chlorocebus aethiops , Dicroísmo Circular , Fator H do Complemento/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fluorescência , Humanos , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Ligação Proteica , Conformação Proteica , Toxina Shiga II/genética , Triexosilceramidas/metabolismo , Tripsina , Células Vero
7.
Soft Matter ; 16(4): 907-913, 2020 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-31854427

RESUMO

Biological systems have evolved sophisticated molecular assemblies capable of exquisite molecular recognition across length scales ranging from angstroms to microns. For instance, the self-organization of glycolipids and glycoproteins on cell membranes allows for molecular recognition of a diversity of ligands ranging from small molecules and proteins to viruses and whole cells. A distinguishing feature of these 2D surfaces is they achieve exceptional binding selectivity and avidity by exploiting multivalent binding interactions. Here we develop a 2D ligand display platform based on peptoid nanosheets that mimics the structure and function of the cell membrane. A variety of small-molecule lipid-conjugates were co-assembled with the peptoid chains to create a diversity of functionalized nanosheet bilayers with varying display densities. The functional heads of the lipids were shown to be surface-exposed, and the carbon tails immobilized into the hydrophobic interior. We demonstrate that saccharide-functionalized nanosheets (e.g., made from globotriaosylsphingosine or 1,2-dipalmitoyl-sn-glycero-3-phospho((ethyl-1',2',3'-triazole)triethyleneglycolmannose)) can have very diverse binding properties, exhibiting specific binding to multivalent proteins as well as to intact bacterial cells. Analysis of sugar display densities revealed that Shiga toxin 1 subunit B (a pentameric protein) and FimH-expressing Escherichia coli (E. coli) bind through the cooperative binding behavior of multiple carbohydrates. The ability to readily incorporate and display a wide variety of lipidated cargo on the surface of peptoid nanosheets makes this a convenient route to soluble, cell-surface mimetic materials. These materials hold great promise for drug screening, biosensing, bioremediation, and as a means to combat pathogens by direct physical binding through a well-defined, multivalent 2D material.


Assuntos
Carboidratos/química , Lipídeos/química , Nanoestruturas/química , Peptoides/química , Adesinas de Escherichia coli/química , Adesinas de Escherichia coli/genética , Biomimética , Escherichia coli/genética , Proteínas de Fímbrias/química , Proteínas de Fímbrias/genética , Interações Hidrofóbicas e Hidrofílicas , Toxina Shiga II/química , Toxina Shiga II/genética , Açúcares/química
8.
Int J Med Microbiol ; 308(8): 1073-1084, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30224239

RESUMO

Shiga toxin (Stx)-mediated injury of the kidneys and the brain represent the major extraintestinal complications in humans upon infection by enterohemorrhagic Escherichia coli (EHEC). Damage of renal and cerebral endothelial cells is the key event in the pathogenesis of the life-threatening hemolytic uremic syndrome (HUS). Stxs are AB5 toxins and the B-pentamers of the two clinically important Stx subtypes Stx1a and Stx2a preferentially bind to the glycosphingolipid globotriaosylceramide (Gb3Cer, Galα4Galß4Glcß1Cer) and to less extent to globotetraosylceramide (Gb4Cer, GalNAcß3Galα4Galß4Glcß1), which are expected to reside in lipid rafts in the plasma membrane of the human endothelium. This review summarizes the current knowledge on the Stx glycosphingolipid receptors and their lipid membrane ensemble in primary human brain microvascular endothelial cells (pHBMECs) and primary human renal glomerular endothelial cells (pHRGECs). Increasing knowledge on the precise initial molecular mechanisms by which Stxs interact with cellular targets will help to develop specific therapeutics and/or preventive measures to combat EHEC-caused diseases.


Assuntos
Escherichia coli Êntero-Hemorrágica/fisiologia , Infecções por Escherichia coli/metabolismo , Globosídeos/metabolismo , Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Triexosilceramidas/metabolismo , Encéfalo/citologia , Células Endoteliais/citologia , Escherichia coli Êntero-Hemorrágica/patogenicidade , Infecções por Escherichia coli/microbiologia , Globosídeos/química , Síndrome Hemolítico-Urêmica/metabolismo , Síndrome Hemolítico-Urêmica/microbiologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Rim/citologia , Cultura Primária de Células , Toxina Shiga I/química , Toxina Shiga II/química , Triexosilceramidas/química
9.
Traffic ; 16(12): 1270-87, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26420131

RESUMO

Shiga toxin-producing Escherichia coli (STEC) produce two types of Shiga toxin (STx): STx1 and STx2. The toxin A-subunits block protein synthesis, while the B-subunits mediate retrograde trafficking. STEC infections do not have definitive treatments, and there is growing interest in generating toxin transport inhibitors for therapy. However, a comprehensive understanding of the mechanisms of toxin trafficking is essential for drug development. While STx2 is more toxic in vivo, prior studies focused on STx1 B-subunit (STx1B) trafficking. Here, we show that, compared with STx1B, trafficking of the B-subunit of STx2 (STx2B) to the Golgi occurs with slower kinetics. Despite this difference, similar to STx1B, endosome-to-Golgi transport of STx2B does not involve transit through degradative late endosomes and is dependent on dynamin II, epsinR, retromer and syntaxin5. Importantly, additional experiments show that a surface-exposed loop in STx2B (ß4-ß5 loop) is required for its endosome-to-Golgi trafficking. We previously demonstrated that residues in the corresponding ß4-ß5 loop of STx1B are required for interaction with GPP130, the STx1B-specific endosomal receptor, and for endosome-to-Golgi transport. Overall, STx1B and STx2B share a common pathway and use a similar structural motif to traffic to the Golgi, suggesting that the underlying mechanisms of endosomal sorting may be evolutionarily conserved.


Assuntos
Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Escherichia coli Shiga Toxigênica/metabolismo , Motivos de Aminoácidos , Sequência Consenso , Sequência Conservada , Endossomos/metabolismo , Evolução Molecular , Galactosiltransferases/genética , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Cinética , Ligação Proteica , Subunidades Proteicas , Transporte Proteico , Toxina Shiga I/química , Toxina Shiga I/genética , Toxina Shiga II/química , Toxina Shiga II/genética , Proteínas de Transporte Vesicular/metabolismo
10.
Glycobiology ; 27(1): 99-109, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27558838

RESUMO

Shiga toxin (Stx)-mediated injury to microvascular endothelial cells in the brain significantly contributes to the pathogenesis of the hemolytic-uremic syndrome caused by enterohemorrhagic Escherichia coli (EHEC). Stxs are AB5 toxins and the B-pentamers of the two major Stx subtypes Stx1a and Stx2a preferentially bind to the glycosphingolipid (GSL) globotriaosylceramide (Gb3Cer) expressed by human endothelial cells. Here we report on comprehensive structural analysis of the different lipoforms of Gb3Cer (Galα4Galß4Glcß1Cer) and globotetraosylceramide (Gb4Cer, GalNAcß3Galα4Galß4Glcß1Cer, the less effective Stx receptor) of primary human brain microvascular endothelial cells and their association with lipid rafts. Detergent-resistant membranes (DRMs), obtained by sucrose density gradient ultracentrifugation, were used as lipid raft-analogous microdomains of the liquid-ordered phase and nonDRM fractions were employed as equivalents for the liquid-disordered phase of cell membranes. Structures of the prevalent lipoforms of Gb3Cer and Gb4Cer were those with Cer (d18:1, C16:0), Cer (d18:1, C22:0) and Cer (d18:1, C24:1/C24:0) determined by electrospray ionization mass spectrometry that was combined with thin-layer chromatography immunodetection using anti-Gb3Cer and anti-Gb4Cer antibodies as well as Stx1a and Stx2a subtypes. Association of Stx receptor GSLs was determined by co-localization with lipid raft-specific membrane protein flotillin-2 and canonical lipid raft marker sphingomyelin with Cer (d18:1, C16:0) and Cer (d18:1, C24:1/C24:0) in the liquid-ordered phase, whereas lyso-phosphatidylcholine was detectable exclusively in the liquid-disordered phase. Defining the precise microdomain structures of primary endothelial cells may help to unravel the initial mechanisms by which Stxs interact with their target cells and will help to develop novel preventive and therapeutic measures for EHEC-mediated diseases.


Assuntos
Globosídeos/química , Receptores de Superfície Celular/química , Toxina Shiga I/química , Toxina Shiga II/química , Triexosilceramidas/química , Anticorpos/química , Barreira Hematoencefálica/química , Barreira Hematoencefálica/metabolismo , Cromatografia em Camada Fina , Células Endoteliais/química , Escherichia coli/patogenicidade , Globosídeos/genética , Glicoesfingolipídeos/química , Glicoesfingolipídeos/genética , Humanos , Microdomínios da Membrana/química , Microdomínios da Membrana/genética , Receptores de Superfície Celular/genética , Toxina Shiga I/genética , Toxina Shiga II/genética , Triexosilceramidas/genética
11.
Biophys J ; 108(12): 2775-8, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26083916

RESUMO

Shiga toxin subunit B (STxB) binding to its cellular receptor Gb3 leads to the formation of protein-lipid clusters and bending of the membrane. A newly developed synthetic route allowed synthesizing the biologically most relevant Gb3-C24:1 2OH species with both, the natural (Gb3-R) as well as the unnatural (Gb3-S) configuration of the 2OH group. The derivatives bind STxB with identical nanomolar affinity, while the propensity to induce membrane tubules in giant unilamellar vesicles is more pronounced for Gb3-S. Fluorescence and atomic force microscopy images of phase-separated supported membranes revealed differences in the lateral organization of the protein on the membrane. Gb3-R favorably induces large and tightly packed protein clusters, while a lower protein density is found on Gb3-S doped membranes.


Assuntos
Membrana Celular/ultraestrutura , Ácidos Graxos/metabolismo , Hidroxiácidos/metabolismo , Toxina Shiga II/metabolismo , Triexosilceramidas/metabolismo , Membrana Celular/metabolismo , Ácidos Graxos/química , Hidroxiácidos/química , Ligação Proteica , Toxina Shiga II/química , Triexosilceramidas/química , Lipossomas Unilamelares/química , Lipossomas Unilamelares/metabolismo
12.
J Biol Chem ; 289(2): 885-94, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24225957

RESUMO

Shiga toxin type 2 (Stx2a) is clinically most closely associated with enterohemorrhagic E. coli O157:H7-mediated hemorrhagic colitis that sometimes progresses to hemolytic-uremic syndrome. The ability to express the toxin has been acquired by other Escherichia coli strains, and outbreaks of food poisoning have caused significant mortality rates as, for example, in the 2011 outbreak in northern Germany. Stx2a, an AB5 toxin, gains entry into human cells via the glycosphingolipid receptor Gb3. We have determined the first crystal structure of a disaccharide analog of Gb3 bound to the B5 pentamer of Stx2a holotoxin. In this Gb3 analog,-GalNAc replaces the terminal-Gal residue. This co-crystal structure confirms previous inferences that two of the primary binding sites identified in theB5 pentamer of Stx1 are also functional in Stx2a. This knowledge provides a rationale for the synthesis and evaluation of heterobifunctional antagonists for E. coli toxins that target Stx2a. Incorporation of GalNAc Gb3 trisaccharide in a heterobifunctional ligand with an attached pyruvate acetal, a ligand for human amyloid P component, and conjugation to poly[acrylamide-co-(3-azidopropylmethacrylamide)] produced a polymer that neutralized Stx2a in a mouse model of Shigatoxemia.


Assuntos
Dissacarídeos/química , Desenho de Fármacos , Inibidores Enzimáticos/química , Toxina Shiga II/química , Animais , Configuração de Carboidratos , Sequência de Carboidratos , Cristalografia por Raios X , Dissacarídeos/metabolismo , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Toxina Shiga II/antagonistas & inibidores , Toxina Shiga II/metabolismo , Análise de Sobrevida , Toxemia/prevenção & controle
13.
J Biol Chem ; 289(36): 25374-81, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25053417

RESUMO

Shiga toxin Stx2e is the major known agent that causes edema disease in newly weaned pigs. This severe disease is characterized by neurological disorders, hemorrhagic lesions, and frequent fatal outcomes. Stx2e consists of an enzymatically active A subunit and five B subunits that bind to a specific glycolipid receptor on host cells. It is evident that antibodies binding to the A subunit or the B subunits of Shiga toxin variants may have the capability to inhibit their cytotoxicity. Here, we report the discovery and characterization of a VHH single domain antibody (nanobody) isolated from a llama phage display library that confers potent neutralizing capacity against Stx2e toxin. We further present the crystal structure of the complex formed between the nanobody (NbStx2e1) and the Stx2e toxoid, determined at 2.8 Å resolution. Structural analysis revealed that for each B subunit of Stx2e, one NbStx2e1 is interacting in a head-to-head orientation and directly competing with the glycolipid receptor binding site on the surface of the B subunit. The neutralizing NbStx2e1 can in the future be used to prevent or treat edema disease.


Assuntos
Anticorpos Neutralizantes/química , Estrutura Terciária de Proteína , Toxina Shiga II/química , Anticorpos de Domínio Único/química , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/imunologia , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Ligação Competitiva/imunologia , Camelídeos Americanos/imunologia , Cristalografia por Raios X , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica/imunologia , Subunidades Proteicas/química , Subunidades Proteicas/imunologia , Subunidades Proteicas/metabolismo , Receptores de Superfície Celular/metabolismo , Homologia de Sequência de Aminoácidos , Toxina Shiga II/imunologia , Toxina Shiga II/metabolismo , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/imunologia
14.
Glycobiology ; 25(8): 845-54, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25941008

RESUMO

The affinities of the most abundant oligosaccharides found in human milk for four bacterial exotoxins (from Vibrio cholerae and pathogenic Escherichia coli) were quantified for the first time. Association constants (Ka) for a library of 20 human milk oligosaccharides (HMOs) binding to Shiga toxin type 2 holotoxin (Stx2) and the B subunit homopentamers of cholera toxin, heat-labile toxin and Shiga toxin type 1 (CTB5, HLTB5 and Stx1B5) were measured at 25°C and pH 7 using the direct electrospray ionization mass spectrometry assay. Notably, all four bacterial toxins bind to a majority of the HMOs tested and five of the HMOs (2'-fucosyllactose, lacto-N-tetraose, lacto-N-fucopentaose I, lacto-N-fucopentaose II and lacto-N-fucopentaose III) are ligands for all four toxins. These five HMOs are also reported to bind to other bacterial toxins (e.g. toxin A and toxin B of Clostridium difficile). In all cases, the HMO affinities (apparent Ka) are relatively modest (≤15,000 M(-1)). However, at the high concentrations of HMOs typically ingested by infants, a significant fraction of these toxins, if present, is expected to be bound to HMOs. Binding measurements carried out with 2'-fucosyllactose or lacto-N-fucopentaose I, together with a high-affinity ligand based on the native carbohydrate receptor, revealed that all four toxins possess HMO-binding sites that are distinct from those of the native receptors, although evidence of competitive binding was found for lacto-N-fucopentaose I with Stx2 and 2'-fucosyllactose and lacto-N-fucopentaose I with HLTB5. Taken together, the results of this study suggest that, while HMOs are expected to bind extensively to these bacterial toxins, it is unlikely that HMO binding will effectively inhibit their interactions with their cellular receptors.


Assuntos
Clostridioides difficile/química , Escherichia coli Enteropatogênica/química , Leite Humano/química , Vibrio cholerae/química , Amino Açúcares/química , Amino Açúcares/isolamento & purificação , Proteínas de Bactérias/química , Proteínas de Bactérias/isolamento & purificação , Toxinas Bacterianas/química , Toxinas Bacterianas/isolamento & purificação , Sítios de Ligação , Sequência de Carboidratos , Toxina da Cólera/química , Toxina da Cólera/isolamento & purificação , Enterotoxinas/química , Enterotoxinas/isolamento & purificação , Humanos , Concentração de Íons de Hidrogênio , Dados de Sequência Molecular , Oligossacarídeos/química , Oligossacarídeos/isolamento & purificação , Polissacarídeos/química , Polissacarídeos/isolamento & purificação , Ligação Proteica , Toxina Shiga I/química , Toxina Shiga I/isolamento & purificação , Toxina Shiga II/química , Toxina Shiga II/isolamento & purificação , Espectrometria de Massas por Ionização por Electrospray , Trissacarídeos/química , Trissacarídeos/isolamento & purificação
15.
Antimicrob Agents Chemother ; 59(11): 7054-60, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26349825

RESUMO

Shiga-like toxins (Stxs), produced by pathogenic Escherichia coli, are a major virulence factor involved in severe diseases in human and animals. These toxins are ribosome-inactivating proteins, and treatment for diseases caused by them is not available. Therefore, there is an urgent need for agents capable of effectively targeting this lethal toxin. In this study, we identified baicalin, a flavonoid compound used in Chinese traditional medicine, as a compound against Shiga-like toxin 2 (Stx2). We found that baicalin significantly improves renal function and reduces Stx2-induced lethality in mice. Further experiments revealed that baicalin induces the formation of oligomers by the toxin by direct binding. We also identified the residues important for such interactions and analyzed their roles in binding baicalin by biophysical and biochemical analyses. Our results establish baicalin as a candidate compound for the development of therapeutics against diseases caused by Stxs.


Assuntos
Flavonoides/uso terapêutico , Toxina Shiga II/metabolismo , Toxina Shiga II/toxicidade , Animais , Flavonoides/química , Células HeLa , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Camundongos , Toxina Shiga II/química
16.
Biochem Biophys Res Commun ; 464(4): 1282-1289, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26220340

RESUMO

The cell-specific cytosolic delivery of functional macromolecules with high efficiency is of great significance in molecular medicine and biotechnology. Herein, we present a Shiga-like toxin II-based high-efficiency and receptor-specific intracellular delivery system. We designed and constructed the Shiga-like toxin-based carrier (STC) to comprise the targeting and translocation domains, and used it for delivering a protein cargo. The STC was shown to deliver a protein cargo into the cytosol with high efficiency in a receptor-specific manner, exhibiting much higher efficiency than the most widely used cell-penetrating peptide. The general utility of the STC was demonstrated by modulating the targeting domain. The present delivery platform can be widely used for the intracellular delivery of diverse biomolecules in a receptor-specific and genetically encodable manner.


Assuntos
Membrana Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Fluorescência Verde/administração & dosagem , Proteínas de Fluorescência Verde/farmacocinética , Nanocápsulas/química , Toxina Shiga II/farmacocinética , Animais , Proteínas de Fluorescência Verde/genética , Humanos , Nanocápsulas/ultraestrutura , Toxina Shiga II/química , Toxina Shiga II/genética
17.
J Immunol ; 191(5): 2403-11, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23918978

RESUMO

The striking feature of enterohemorrhagic Escherichia coli (EHEC) infections is the production of Shiga toxins (Stx) implicated in the development of the life-threatening hemolytic uremic syndrome. Despite the magnitude of the social impact of EHEC infections, no licensed vaccine or effective therapy is available for human use. One of the biggest challenges is to develop an effective and safe immunogen to ensure nontoxicity, as well as a strong input to the immune system to induce long-lasting, high-affinity Abs with anti-Stx-neutralizing capacity. The enzyme lumazine synthase from Brucella spp. (BLS) is a highly stable dimer of pentamers and a scaffold with enormous plasticity on which to display foreign Ags. Taking into account the advantages of BLS and the potential capacity of the B subunit of Stx2 to induce Abs that prevent Stx2 toxicity by blocking its entrance into the host cells, we engineered a new immunogen by inserting the B subunit of Stx2 at the amino termini of BLS. The resulting chimera demonstrated a strong capacity to induce a long-lasting humoral immune response in mice. The chimera induced Abs with high neutralizing capacity for Stx2 and its variants. Moreover, immunized mice were completely protected against i.v. Stx2 challenge, and weaned mice receiving an oral challenge with EHEC were completely protected by the transference of immune sera. We conclude that this novel immunogen represents a promising candidate for vaccine or Ab development with preventive or therapeutic ends, for use in hemolytic uremic syndrome-endemic areas or during future outbreaks caused by pathogenic strains of Stx-producing E. coli.


Assuntos
Síndrome Hemolítico-Urêmica/prevenção & controle , Complexos Multienzimáticos/imunologia , Toxina Shiga II/imunologia , Vacinas contra Shigella/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Neutralizantes/sangue , Brucella , Modelos Animais de Doenças , Escherichia coli Êntero-Hemorrágica , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Complexos Multienzimáticos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Toxina Shiga II/química
18.
Glycobiology ; 24(1): 26-38, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24082034

RESUMO

The binding of Shiga-like toxin 1 (Stx1) and Shiga-like toxin 2 (Stx2) to a mucin-like fusion protein, P-selectin glycoprotein ligand-1/mouse IgG2b (PSGL-1/mIgG2b), carrying multiple copies of the blood group P1 determinant on O-glycans was investigated with western blot and the biosensor Biacore. Chinese hamster ovary K-1 (CHO-K1) cells were stably transfected with linearized plasmids encoding the PSGL-1/mIgG2b fusion protein, the pigeon α1,4-galactosyltransferase (α4Gal-T) and the core 2 ß1,6-N-acetylglucosaminyltransferase (C2GnT-I). Western blot analyses of purified PSGL-1/mIgG2b and liquid chromatography-mass spectrometry (LC-MS) of released O-glycans confirmed the presence of the P1 determinant. Western blot analysis indicated strong binding of Stx1, but not Stx2, to PSGL-1/mIgG2b. In a Biacore assay, Stx1 and Stx2 were immobilized on a dextran chip and the binding of purified PSGL-1/mIgG2b and a P(k)-albumin neoglycoprotein was analyzed. Stx1 and Stx2 bound with high avidity to both PSGL-1/mIgG2b and P(k)-albumin, while the Stx1 binding was the strongest. In summary, we have shown that the pigeon α4Gal-T can be aberrantly expressed in CHO cells together with the core 2 enzyme to generate multiple, O-linked P1 determinants on a simultaneously expressed mucin-type fusion protein. P1-decorated PSGL-1/mIgG2b bound with high avidity to both Stx1 and Stx2, and as such constitutes a potential therapeutic inhibitor of these toxins.


Assuntos
Globosídeos/química , Polissacarídeos/química , Toxina Shiga I/química , Toxina Shiga II/química , Animais , Células CHO , Columbidae , Cricetinae , Cricetulus , Globosídeos/genética , Globosídeos/metabolismo , Humanos , Imunoglobulina G/química , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , N-Acetilglucosaminiltransferases/química , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Polissacarídeos/genética , Polissacarídeos/metabolismo , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Toxina Shiga I/genética , Toxina Shiga I/metabolismo , Toxina Shiga II/genética , Toxina Shiga II/metabolismo , Escherichia coli Shiga Toxigênica/química , Escherichia coli Shiga Toxigênica/genética , Escherichia coli Shiga Toxigênica/metabolismo
19.
Appl Environ Microbiol ; 80(9): 2928-40, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24584253

RESUMO

We have analyzed 26 Shiga toxin-producing Escherichia coli (STEC) strains for Shiga toxin 2 (Stx2) production using matrix-assisted laser desorption ionization (MALDI)-tandem time of flight (TOF-TOF) tandem mass spectrometry (MS/MS) and top-down proteomic analysis. STEC strains were induced to overexpress Stx2 by overnight culturing on solid agar supplemented with either ciprofloxacin or mitomycin C. Harvested cells were lysed by bead beating, and unfractionated bacterial cell lysates were ionized by MALDI. The A2 fragment of the A subunit and the mature B subunit of Stx2 were analyzed by MS/MS. Sequence-specific fragment ions were used to identify amino acid subtypes of Stx2 using top-down proteomic analysis using software developed in-house at the U.S. Department of Agriculture (USDA). Stx2 subtypes (a, c, d, f, and g) were identified on the basis of the mass of the A2 fragment and the B subunit as well as from their sequence-specific fragment ions by MS/MS (postsource decay). Top-down proteomic identification was in agreement with DNA sequencing of the full Stx2 operon (stx2) for all strains. Top-down results were also compared to a bioassay using a Vero-d2EGFP cell line. Our results suggest that top-down proteomic identification is a rapid, highly specific technique for distinguishing Stx2 subtypes.


Assuntos
Proteínas de Escherichia coli/química , Proteômica/métodos , Toxina Shiga II/química , Escherichia coli Shiga Toxigênica/isolamento & purificação , Espectrometria de Massas em Tandem/métodos , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Dados de Sequência Molecular , Estrutura Molecular , Toxina Shiga II/biossíntese , Escherichia coli Shiga Toxigênica/química , Escherichia coli Shiga Toxigênica/classificação , Escherichia coli Shiga Toxigênica/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos
20.
Semin Thromb Hemost ; 40(4): 503-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24799304

RESUMO

Hemolytic uremic syndrome (HUS), the most common cause of acute renal failure in childhood, is mainly caused by infection with Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC). Besides its cytotoxic activity, Stx has been shown to interact with the complement system. Complement breakdown products have been found in serum of HUS patients suggesting complement activation and in vitro studies have demonstrated that Stx2 directly activates complement leading to formation of terminal complement complex. Furthermore, Stx2 has been found to bind to factor H (FH) resulting in a reduced cofactor activity on the cell surface. Binding of Stx2 has also been shown for other members of the FH family, namely FH-like protein 1 and FH-related protein 1. Both proteins also compete with FH for Stx binding, so that in the presence of FHR-1 less FH is bound to Stx and therefore more is available for endothelial cell protection. In addition, Stx2 has been demonstrated to downregulate the membrane-bound regulator CD59 on the surface of glomerular endothelial and tubulus epithelial cells on protein and at the mRNA level. In conclusion, Stx modulates complement regulator proteins leading to an impaired control and thus to enhanced complement activation. Its implication in the pathogenesis of EHEC-induced HUS in vivo and whether complement blockage might be a therapeutic option still has to be elucidated.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Escherichia coli Êntero-Hemorrágica , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/imunologia , Toxina Shiga II/química , Toxina Shiga/química , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígenos CD59/metabolismo , Membrana Celular/metabolismo , Ativação do Complemento , Fator H do Complemento/fisiologia , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Glomérulos Renais/imunologia , Glomérulos Renais/metabolismo , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA