Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Toxins (Basel) ; 11(12)2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31816894

RESUMO

The subtilase cytotoxin (SubAB) of Shiga toxin-producing Escherichia coli (STEC) is a member of the AB5 toxin family. In the current study, we analyzed the formation of active homo- and hetero-complexes of SubAB variants in vitro to characterize the mode of assembly of the subunits. Recombinant SubA1-His, SubB1-His, SubA2-2-His, and SubB2-2-His subunits, and His-tag-free SubA2-2 were separately expressed, purified, and biochemically characterized by circular dichroism (CD) spectroscopy, size-exclusion chromatography (SEC), and analytical ultracentrifugation (aUC). To confirm their biological activity, cytotoxicity assays were performed with HeLa cells. The formation of AB5 complexes was investigated with aUC and isothermal titration calorimetry (ITC). Binding of SubAB2-2-His to HeLa cells was characterized with flow cytometry (FACS). Cytotoxicity experiments revealed that the analyzed recombinant subtilase subunits were biochemically functional and capable of intoxicating HeLa cells. Inhibition of cytotoxicity by Brefeldin A demonstrated that the cleavage is specific. All His-tagged subunits, as well as the non-tagged SubA2-2 subunit, showed the expected secondary structural compositions and oligomerization. Whereas SubAB1-His complexes could be reconstituted in solution, and revealed a Kd value of 3.9 ± 0.8 µmol/L in the lower micromolar range, only transient interactions were observed for the subunits of SubAB2-2-His in solution, which did not result in any binding constant when analyzed with ITC. Additional studies on the binding characteristics of SubAB2-2-His on HeLa cells revealed that the formation of transient complexes improved binding to the target cells. Conclusively, we hypothesize that SubAB variants exhibit different characteristics in their binding behavior to their target cells.


Assuntos
Proteínas de Escherichia coli , Subunidades Proteicas , Subtilisinas , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/toxicidade , Células HeLa , Humanos , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/toxicidade , Escherichia coli Shiga Toxigênica , Subtilisinas/química , Subtilisinas/genética , Subtilisinas/toxicidade
2.
Int J Biol Macromol ; 102: 308-314, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28392381

RESUMO

The light subunit of mushroom Agaricus bisporus tyrosinase (LSMT) is a protein of unknown function that was discovered serendipitously during the elucidation of the crystal structure of the enzyme. The protein is non-immunogenic and can penetrate the intestinal epithelial cell barrier, and thus, similar to its structural homologue HA-33 from Clostridium botulinum, may be potentially absorbable by the intestine. LSMT also shares high structural homology with the ricin-B-like lectin from the mushroom Clitocybe nebularis (CNL), which has been shown to display biological activity against leukemic cancer cells and dendritic cells. Therefore, we evaluated the biological activity of LSMT. An in vitro assay suggested that LSMT presentation to most of the cancer cell lines studied has a negligible effect on their proliferation. However, inhibition of cell growth and a slight stimulation of cell proliferation were observed with breast cancer and macrophage cells, respectively. LSMT appeared to be relatively resistant against proteolysis by trypsin and papain, but not bromelain. Challenges with gastric and intestinal juice suggested that the protein is resistant to gastrointestinal tract conditions. This is the first report on the biological characteristics and implication of LSMT.


Assuntos
Agaricus/enzimologia , Monofenol Mono-Oxigenase/química , Monofenol Mono-Oxigenase/farmacologia , Subunidades Proteicas/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Humanos , Células MCF-7 , Camundongos , Monofenol Mono-Oxigenase/toxicidade , Subunidades Proteicas/toxicidade , Células RAW 264.7
3.
J Neuroimmunol ; 303: 13-21, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28038891

RESUMO

Myasthenia gravis (MG) is usually caused by antibodies against the muscle acetylcholine receptor (AChR). Experimental autoimmune MG (EAMG) is the animal model of MG, typically induced by immunization of rodents with AChR isolated from the electric organ of Torpedo californica. We have successfully induced EAMG in Lewis rats by immunization with the extracellular domains (ECDs) of the human AChR subunits (α, ß, γ, δ and ε) expressed in yeast. Analysis of the antibody titers revealed a robust antigenic response against all the peptides, but a marked difference in their pathogenicity; the α subunit ECD was the most pathogenic, resulting in the highest percentage of affected animals. Measurements of antibody titers, electromyographic tests and quantitation of the muscle AChR content, offered further support to these findings. The EAMG models presented herein, could be used for studying subunit-specific pathogenic mechanisms, and, more importantly, as tools for the evaluation of antigen-specific therapeutic approaches, which rely on the human AChR.


Assuntos
Modelos Animais de Doenças , Miastenia Gravis Autoimune Experimental/induzido quimicamente , Miastenia Gravis Autoimune Experimental/imunologia , Receptores Colinérgicos/administração & dosagem , Receptores Colinérgicos/imunologia , Animais , Feminino , Humanos , Miastenia Gravis Autoimune Experimental/metabolismo , Domínios Proteicos/fisiologia , Subunidades Proteicas/imunologia , Subunidades Proteicas/toxicidade , Ratos , Ratos Endogâmicos Lew
4.
Toxins (Basel) ; 7(5): 1467-85, 2015 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-25938272

RESUMO

Shiga toxin producing Escherichia coli O157:H7 (STEC) is one of the leading causes of food-poisoning around the world. Some STEC strains produce Shiga toxin 1 (Stx1) and/or Shiga toxin 2 (Stx2) or variants of either toxin, which are critical for the development of hemorrhagic colitis (HC) or hemolytic uremic syndrome (HUS). Currently, there are no therapeutic treatments for HC or HUS. E. coli O157:H7 strains carrying Stx2 are more virulent and are more frequently associated with HUS, which is the most common cause of renal failure in children in the US. The basis for the increased potency of Stx2 is not fully understood. Shiga toxins belong to the AB5 family of protein toxins with an A subunit, which depurinates a universally conserved adenine residue in the α-sarcin/ricin loop (SRL) of the 28S rRNA and five copies of the B subunit responsible for binding to cellular receptors. Recent studies showed differences in the structure, receptor binding, dependence on ribosomal proteins and pathogenicity of Stx1 and Stx2 and supported a role for the B subunit in differential toxicity. However, the current data do not rule out a potential role for the A1 subunits in the differential toxicity of Stx1 and Stx2. This review highlights the recent progress in understanding the differences in the A1 subunits of Stx1 and Stx2 and their role in defining toxicity.


Assuntos
Subunidades Proteicas/toxicidade , Toxina Shiga I/toxicidade , Toxina Shiga II/toxicidade , Animais , Humanos , Conformação Proteica , Subunidades Proteicas/química , Ribossomos/metabolismo , Toxina Shiga I/química , Toxina Shiga II/química
5.
Nat Commun ; 6: 6198, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25652783

RESUMO

Pore-forming toxins are protein assemblies used by many organisms to disrupt the membranes of target cells. They are expressed as soluble monomers that assemble spontaneously into multimeric pores. However, owing to their complexity, the assembly processes have not been resolved in detail for any pore-forming toxin. To determine the assembly mechanism for the ring-shaped, homododecameric pore of the bacterial cytolytic toxin ClyA, we collected a diverse set of kinetic data using single-molecule spectroscopy and complementary techniques on timescales from milliseconds to hours, and from picomolar to micromolar ClyA concentrations. The entire range of experimental results can be explained quantitatively by a surprisingly simple mechanism. First, addition of the detergent n-dodecyl-ß-D-maltopyranoside to the soluble monomers triggers the formation of assembly-competent toxin subunits, accompanied by the transient formation of a molten-globule-like intermediate. Then, all sterically compatible oligomers contribute to assembly, which greatly enhances the efficiency of pore formation compared with simple monomer addition.


Assuntos
Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Proteínas Hemolisinas/química , Multimerização Proteica , Subunidades Proteicas/química , Animais , Células Cultivadas , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/patogenicidade , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/toxicidade , Transferência Ressonante de Energia de Fluorescência , Expressão Gênica , Proteínas Hemolisinas/biossíntese , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/toxicidade , Hemólise/efeitos dos fármacos , Cavalos , Cinética , Maltose/análogos & derivados , Maltose/química , Modelos Moleculares , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Subunidades Proteicas/biossíntese , Subunidades Proteicas/genética , Subunidades Proteicas/toxicidade , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/toxicidade , Termodinâmica
6.
Toxins (Basel) ; 6(11): 3098-116, 2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25365527

RESUMO

The cytolethal distending toxin (Cdt) is a heterotrimeric holotoxin produced by a diverse group of Gram-negative pathogenic bacteria. The Cdts expressed by the members of this group comprise a subclass of the AB toxin superfamily. Some AB toxins have hijacked the retrograde transport pathway, carried out by the Golgi apparatus and endoplasmic reticulum (ER), to translocate to cytosolic targets. Those toxins have been used as tools to decipher the roles of the Golgi and ER in intracellular transport and to develop medically useful delivery reagents. In comparison to the other AB toxins, the Cdt exhibits unique properties, such as translocation to the nucleus, that present specific challenges in understanding the precise molecular details of the trafficking pathway in mammalian cells. The purpose of this review is to present current information about the mechanisms of uptake and translocation of the Cdt in relation to standard concepts of endocytosis and retrograde transport. Studies of the Cdt intoxication process to date have led to the discovery of new translocation pathways and components and most likely will continue to reveal unknown features about the mechanisms by which bacterial proteins target the mammalian cell nucleus. Insight gained from these studies has the potential to contribute to the development of novel therapeutic strategies.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Núcleo Celular/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Modelos Biológicos , Aggregatibacter actinomycetemcomitans/metabolismo , Animais , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/biossíntese , Toxinas Bacterianas/toxicidade , Núcleo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Complexo de Golgi/efeitos dos fármacos , Humanos , Mamíferos , Subunidades Proteicas/metabolismo , Subunidades Proteicas/toxicidade , Transporte Proteico/efeitos dos fármacos , Especificidade da Espécie
7.
Biochem J ; 457(3): 485-96, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24200403

RESUMO

EDEM1 [ER (endoplasmic reticulum)-degradation-enhancing α-mannosidase I-like protein 1] and EDEM2 are crucial regulators of ERAD (ER-associated degradation) that extracts non-native glycoproteins from the calnexin chaperone system. Ricin is a potent plant cytotoxin composed of an A-chain (RTA) connected by a disulfide bond to a cell-binding lectin B-chain (RTB). After endocytic uptake, the toxin is transported retrogradely to the ER, where the enzymatically active RTA is translocated to the cytosol in a similar manner as misfolded ER proteins. This transport is promoted by EDEM1. In the present study we report that EDEM2 is also involved in ricin retrotranslocation out of the ER. However, the role of EDEM1 and EDEM2 in ricin transport to the cytosol seems to differ. EDEM2 promotes ricin retrotranslocation irrespectively of ER translocon accessibility; moreover, co-immunoprecipitation and pull-down studies revealed that more ricin can interact with EDEM2 in comparison with EDEM1. On the other hand, interactions of both lectins with RTA are dependent on the structure of the RTA. Thus our data display a newly discovered role for EDEM2. Moreover, analysis of the involvement of EDEM1 and EDEM2 in ricin retrotranslocation to the cytosol may provide crucial information about general mechanisms of the recognition of ERAD substrates in the ER.


Assuntos
Substâncias para a Guerra Química/toxicidade , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Lectinas/metabolismo , Proteínas de Membrana/metabolismo , Ricina/toxicidade , Substituição de Aminoácidos , Animais , Sobrevivência Celular/efeitos dos fármacos , Substâncias para a Guerra Química/química , Glicoproteínas , Células HEK293 , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Lectinas/biossíntese , Lectinas/genética , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Proteínas Mutantes/química , Proteínas Mutantes/toxicidade , Inibidores de Proteassoma/farmacologia , Estabilidade Proteica/efeitos dos fármacos , Subunidades Proteicas/química , Subunidades Proteicas/toxicidade , Transporte Proteico/efeitos dos fármacos , Desdobramento de Proteína/efeitos dos fármacos , Deficiências na Proteostase/induzido quimicamente , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Ricina/química , Ricina/genética , alfa-Manosidase
8.
J Endocrinol ; 219(3): 183-93, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24029729

RESUMO

Graves' disease (GD) is a common organ-specific autoimmune disease with the prevalence between 0.5 and 2% in women. Several lines of evidence indicate that the shed A-subunit rather than the full-length thyrotropin receptor (TSHR) is the autoantigen that triggers autoimmunity and leads to hyperthyroidism. We have for the first time induced GD in female rhesus monkeys, which exhibit greater similarity to patients with GD than previous rodent models. After final immunization, the monkeys injected with adenovirus expressing the A-subunit of TSHR (A-sub-Ad) showed some characteristics of GD. When compared with controls, all the test monkeys had significantly higher TSHR antibody levels, half of them had increased total thyroxine (T4) and free T4, and 50% developed goiter. To better understand the underlying mechanisms, quantitative studies on subpopulations of CD4+T helper cells were carried out. The data indicated that this GD model involved a mixed Th1 and Th2 response. Declined Treg proportions and increased Th17:Treg ratio are also observed. Our rhesus monkey model successfully mimicked GD in humans in many aspects. It would be a useful tool for furthering our understanding of the pathogenesis of GD and would potentially shorten the distance toward the prevention and treatment of this disease in human.


Assuntos
Modelos Animais de Doenças , Doença de Graves/fisiopatologia , Macaca mulatta , Glândula Tireoide/fisiopatologia , Animais , Antígenos/genética , Antígenos/toxicidade , Autoanticorpos/análise , Biomarcadores/sangue , Feminino , Técnicas de Transferência de Genes , Doença de Graves/etiologia , Doença de Graves/imunologia , Doença de Graves/patologia , Humanos , Imunotoxinas/genética , Imunotoxinas/toxicidade , Tamanho do Órgão , Subunidades Proteicas/genética , Subunidades Proteicas/toxicidade , Receptores da Tireotropina/administração & dosagem , Receptores da Tireotropina/genética , Proteínas Recombinantes/toxicidade , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Glândula Tireoide/imunologia , Glândula Tireoide/patologia , Tiroxina/sangue , Tiroxina/metabolismo
9.
Artigo em Inglês | MEDLINE | ID: mdl-23162799

RESUMO

Shiga toxin producing Escherichia coli (STEC) are a major cause of food-borne illness worldwide. However, a consensus regarding the role Shiga toxins play in the onset of diarrhea and hemorrhagic colitis (HC) is lacking. One of the obstacles to understanding the role of Shiga toxins to STEC-mediated intestinal pathology is a deficit in small animal models that perfectly mimic human disease. Infant rabbits have been previously used to study STEC and/or Shiga toxin-mediated intestinal inflammation and diarrhea. We demonstrate using infant rabbits that Shiga toxin-mediated intestinal damage requires A-subunit activity, and like the human colon, that of the infant rabbit expresses the Shiga toxin receptor Gb(3). We also demonstrate that Shiga toxin treatment of the infant rabbit results in apoptosis and activation of p38 within colonic tissues. Finally we demonstrate that the infant rabbit model may be used to test candidate therapeutics against Shiga toxin-mediated intestinal damage. While the p38 inhibitor SB203580 and the ZAK inhibitor DHP-2 were ineffective at preventing Shiga toxin-mediated damage to the colon, pretreatment of infant rabbits with the drug imatinib resulted in a decrease of Shiga toxin-mediated heterophil infiltration of the colon. Therefore, we propose that this model may be useful in elucidating mechanisms by which Shiga toxins could contribute to intestinal damage in the human.


Assuntos
Benzamidas/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/patologia , Piperazinas/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Proteínas Quinases/administração & dosagem , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/metabolismo , Toxina Shiga II/toxicidade , Animais , Animais Recém-Nascidos , Apoptose , Mesilato de Imatinib , MAP Quinase Quinase Quinases , Subunidades Proteicas/toxicidade , Coelhos , Escherichia coli Shiga Toxigênica/patogenicidade , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Toxicon ; 60(3): 254-64, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22538196

RESUMO

Venoms often target vital processes to cause paralysis or death, but many types of venom also elicit notoriously intense pain. While these pain-producing effects can result as a byproduct of generalized tissue trauma, there are now multiple examples of venom-derived toxins that target somatosensory nerve terminals in order to activate nociceptive (pain-sensing) neural pathways. Intriguingly, investigation of the venom components that are responsible for evoking pain has revealed novel roles and/or configurations of well-studied toxin motifs. This review serves to highlight pain-producing toxins that target the capsaicin receptor, TRPV1, or members of the acid-sensing ion channel family, and to discuss the utility of venom-derived multivalent and multimeric complexes.


Assuntos
Dor Aguda/induzido quimicamente , Proteínas do Tecido Nervoso/agonistas , Nociceptores/efeitos dos fármacos , Proteínas/toxicidade , Agonistas de Canais de Sódio , Canais de Cátion TRPV/agonistas , Peçonhas/toxicidade , Canais Iônicos Sensíveis a Ácido , Dor Aguda/etiologia , Dor Aguda/metabolismo , Animais , Mordeduras e Picadas/metabolismo , Mordeduras e Picadas/fisiopatologia , Gânglios Sensitivos/efeitos dos fármacos , Gânglios Sensitivos/metabolismo , Humanos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Nociceptores/metabolismo , Conformação Proteica , Subunidades Proteicas/análise , Subunidades Proteicas/química , Subunidades Proteicas/toxicidade , Proteínas/análise , Proteínas/química , Canais de Sódio/química , Canais de Sódio/metabolismo , Córtex Somatossensorial/efeitos dos fármacos , Córtex Somatossensorial/metabolismo , Canais de Cátion TRPV/química , Canais de Cátion TRPV/metabolismo , Peçonhas/química , Peçonhas/enzimologia
11.
J Cell Biol ; 195(5): 751-64, 2011 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-22123862

RESUMO

We describe a novel labeling strategy to site-specifically attach fluorophores, biotin, and proteins to the C terminus of the A1 subunit (CTA1) of cholera toxin (CTx) in an otherwise correctly assembled and active CTx complex. Using a biotinylated N-linked glycosylation reporter peptide attached to CTA1, we provide direct evidence that ~12% of the internalized CTA1 pool reaches the ER. We also explored the sortase labeling method to attach the catalytic subunit of diphtheria toxin as a toxic warhead to CTA1, thus converting CTx into a cytolethal toxin. This new toxin conjugate enabled us to conduct a genetic screen in human cells, which identified ST3GAL5, SLC35A2, B3GALT4, UGCG, and ELF4 as genes essential for CTx intoxication. The first four encode proteins involved in the synthesis of gangliosides, which are known receptors for CTx. Identification and isolation of the ST3GAL5 and SLC35A2 mutant clonal cells uncover a previously unappreciated differential contribution of gangliosides to intoxication by CTx.


Assuntos
Toxina da Cólera/toxicidade , Toxina Diftérica/química , Transporte Proteico/genética , Aminoaciltransferases/química , Aminoaciltransferases/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Células Cultivadas , Cólera/fisiopatologia , Toxina da Cólera/química , Toxina da Cólera/genética , Clonagem Molecular , Cisteína Endopeptidases/química , Cisteína Endopeptidases/genética , Gangliosídeos/metabolismo , Gangliosídeos/fisiologia , Engenharia Genética , Haploidia , Humanos , Dados de Sequência Molecular , Proteínas de Transporte de Monossacarídeos/química , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas de Transporte de Monossacarídeos/fisiologia , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/toxicidade , Análise de Sequência de Proteína , Sialiltransferases/química , Sialiltransferases/metabolismo , Sialiltransferases/fisiologia
12.
PLoS One ; 6(4): e18825, 2011 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-21526142

RESUMO

Cholera toxin (CT) travels as an intact AB(5) protein toxin from the cell surface to the endoplasmic reticulum (ER) of an intoxicated cell. In the ER, the catalytic A1 subunit dissociates from the rest of the toxin. Translocation of CTA1 from the ER to the cytosol is then facilitated by the quality control mechanism of ER-associated degradation (ERAD). Thermal instability in the isolated CTA1 subunit generates an unfolded toxin conformation that acts as the trigger for ERAD-mediated translocation to the cytosol. In this work, we show by circular dichroism and fluorescence spectroscopy that exposure to 4-phenylbutyric acid (PBA) inhibited the thermal unfolding of CTA1. This, in turn, blocked the ER-to-cytosol export of CTA1 and productive intoxication of either cultured cells or rat ileal loops. In cell culture studies PBA did not affect CT trafficking to the ER, CTA1 dissociation from the holotoxin, or functioning of the ERAD system. PBA is currently used as a therapeutic agent to treat urea cycle disorders. Our data suggest PBA could also be used in a new application to prevent or possibly treat cholera.


Assuntos
Toxina da Cólera/toxicidade , Cólera/tratamento farmacológico , Fenilbutiratos/farmacologia , Fenilbutiratos/uso terapêutico , Subunidades Proteicas/toxicidade , Desdobramento de Proteína/efeitos dos fármacos , Animais , Toxina da Cólera/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Dissulfetos/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células HeLa , Humanos , Íleo/efeitos dos fármacos , Íleo/metabolismo , Íleo/patologia , Ligação Proteica/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/metabolismo , Subunidades Proteicas/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Temperatura
13.
J Invertebr Pathol ; 106(2): 230-5, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20965197

RESUMO

Bacillus sphaericus produces a mosquitocidal binary toxin composed of two subunits, BinA (42 kDa) and BinB (51 kDa). Both components are required for maximum toxicity against mosquito larvae. BinB has been proposed to provide specificity by binding to the epithelial gut cell membrane, while BinA may be responsible for toxicity. To identify regions in BinB responsible for receptor binding and for interaction to BinA, we used six BinB shorter constructs derived from both the N-terminal and the C-terminal halves of the protein. All constructs expressed as inclusion bodies in Escherichia coli, similarly to the wild-type protein. A marked decrease in larvicidal activity was observed when BinA was used in combination with these BinB constructs, used either individually or in pairs from both N and C-halves of BinB. Nevertheless, immunohistochemistry analyses demonstrate that these constructs are able to bind to the epithelium gut cell membrane, and in vitro protein-protein interaction assays revealed that these constructs can bind to BinA. These results show that fragments corresponding to both halves of BinB are able to bind the receptor and to interact with BinA, but both halves are required by the toxin to exhibit full larvicidal activity.


Assuntos
Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Membrana Celular/metabolismo , Culicidae/citologia , Subunidades Proteicas/metabolismo , Subunidades Proteicas/toxicidade , Animais , Culicidae/efeitos dos fármacos , Culicidae/microbiologia , Trato Gastrointestinal/citologia , Inseticidas/farmacologia , Larva/citologia , Ligação Proteica/fisiologia , Receptores de Superfície Celular/metabolismo
14.
FEMS Microbiol Lett ; 303(1): 84-91, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20002193

RESUMO

Bacillus sphaericus produces a mosquito-larvicidal binary toxin composed of BinB and BinA subunits. BinA is important for toxicity, whereas BinB acts as a specific receptor-binding component. To study the functional significance of two regions that are only present in BinB, four block mutations and two single mutations were initially introduced: (111)YLD(113)-->(111)AAA(113), (115)NNH(117)-->(115)AAA(117), (143)GEQ(145)-->(143)AAA(145), (147)FQFY(150)-->(147)AAAA(150), N114A and F146A. Only the replacements at (147)FQFY(150) resulted in a total loss of toxicity to Culex quinquefasciatus larvae. Further single alanine substitutions in this region, F147A, Q148A, F149A and Y150A, were introduced to identify residues playing a critical role in mosquito-larvicidal activity. Larvicidal activity assays revealed that only F149A and Y150A mutants exhibited a total loss of toxicity. The in vitro interaction assays demonstrated that all BinB mutants are able to interact with BinA. Immunohistochemistry analysis revealed that only the Y150A mutant was unable to bind to the larval midgut, suggesting an important role of this residue in receptor binding of the BinB subunit. Conservative aromatic substitutions at F149 and Y150 resulted in full recovery of larvicidal activity, indicating that the aromaticity of F149 and Y150 is a key determinant of larvicidal activity, possibly playing a key role in the membrane interaction and receptor binding.


Assuntos
Bacillus/genética , Bacillus/patogenicidade , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Subunidades Proteicas/metabolismo , Subunidades Proteicas/toxicidade , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Toxinas Bacterianas/genética , Sítios de Ligação , Culex/efeitos dos fármacos , Trato Gastrointestinal/química , Imuno-Histoquímica , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Subunidades Proteicas/genética , Alinhamento de Sequência
15.
Biophys J ; 97(11): 3029-37, 2009 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-19948133

RESUMO

In Alzheimer's disease, calcium permeability through cellular membranes appears to underlie neuronal cell death. It is increasingly accepted that calcium permeability involves toxic ion channels. We modeled Alzheimer's disease ion channels of different sizes (12-mer to 36-mer) in the lipid bilayer using molecular dynamics simulations. Our Abeta channels consist of the solid-state NMR-based U-shaped beta-strand-turn-beta-strand motif. In the simulations we obtain ion-permeable channels whose subunit morphologies and shapes are consistent with electron microscopy/atomic force microscopy. In agreement with imaged channels, the simulations indicate that beta-sheet channels break into loosely associated mobile beta-sheet subunits. The preferred channel sizes (16- to 24-mer) are compatible with electron microscopy/atomic force microscopy-derived dimensions. Mobile subunits were also observed for beta-sheet channels formed by cytolytic PG-1 beta-hairpins. The emerging picture from our large-scale simulations is that toxic ion channels formed by beta-sheets spontaneously break into loosely interacting dynamic units that associate and dissociate leading to toxic ionic flux. This sharply contrasts intact conventional gated ion channels that consist of tightly interacting alpha-helices that robustly prevent ion leakage, rather than hydrogen-bonded beta-strands. The simulations suggest why conventional gated channels evolved to consist of interacting alpha-helices rather than hydrogen-bonded beta-strands that tend to break in fluidic bilayers. Nature designs folded channels but not misfolded toxic channels.


Assuntos
Amiloide/química , Amiloide/metabolismo , Canais Iônicos/química , Canais Iônicos/metabolismo , Dobramento de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Motivos de Aminoácidos , Amiloide/toxicidade , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/metabolismo , Morte Celular , Cloretos/metabolismo , Canais Iônicos/toxicidade , Transporte de Íons , Modelos Moleculares , Peso Molecular , Permeabilidade , Estrutura Secundária de Proteína , Subunidades Proteicas/toxicidade
16.
Toxicon ; 53(2): 262-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19073204

RESUMO

Beta-bungarotoxin (beta-Bgt) consists of A chain (a phospholipase A2 subunit) and B chain, cross-linked by an intersubunit disulfide bridge. In contrast to a marginal activity noted with beta-Bgt, recombinant A1 chain and B1 chain markedly induced release of calcein from phospholipid vesicles. Reduction of intersubunit disulfide bond by dithiothreitol or glutathione enhanced membrane-damaging activity of beta-Bgt. Moreover, phospholipid-binding capability of recombinant A1 and B1 chains was higher than that of beta-Bgt. In contrast to beta-Bgt, A1 and B1 chains preferably bound lipids with a preference for anionic over zwitterionic phospholipids. Removal of positively charged residues lying on the interface between A chain and B chain resulted in abolishment of membrane-permeabilizing activity of B chain. Taken together, our data indicate that both A and B chains possess the capability to induce vesicle leakage, and reduction of interchain disulfide bond markedly releases this ability from intact beta-Bgt molecule.


Assuntos
Bungarotoxinas/química , Bungarotoxinas/toxicidade , Fosfolipídeos/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/toxicidade , Animais , Bungarus/fisiologia , Venenos Elapídicos/química , Lipossomos/química , Lipossomos/metabolismo , Modelos Moleculares , Fosfolipases A2/metabolismo , Fosfolipídeos/química , Conformação Proteica
17.
J Immunol ; 181(6): 4019-26, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768857

RESUMO

Infectious agents, including bacteria and viruses, are thought to provide triggers for the development or exacerbation of autoimmune diseases such as systemic lupus erythematosus in the genetically predisposed individual. Molecular mimicry and engagement of TLRs have been assigned limited roles that link infection to autoimmunity, but additional mechanisms are suspected to be involved. In this study we show that T cells from lupus-prone mice display aggregated lipid rafts that harbor signaling, costimulatory, inflammatory, adhesion, and TLR molecules. The percentage of T cells with clustered lipid rafts increases with age and peaks before the development of lupus pathology. We show that cholera toxin B, a component of Vibrio cholerae, promotes autoantibody production and glomerulonephritis in lupus-prone mice by enhancing lipid raft aggregation in T cells. In contrast, disruption of lipid raft aggregation results in delay of disease pathology. Our results demonstrate that lipid rafts contribute significantly to the pathogenesis of lupus and provide a novel mechanism whereby aggregated lipid rafts represent a potential link between infection and autoimmunity.


Assuntos
Toxina da Cólera/toxicidade , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Microdomínios da Membrana/imunologia , Microdomínios da Membrana/patologia , Animais , Autoanticorpos/biossíntese , Agregação Celular/imunologia , Progressão da Doença , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Camundongos Endogâmicos NZB , Camundongos Knockout , Subunidades Proteicas/toxicidade , Linfócitos T/imunologia , Linfócitos T/microbiologia , Linfócitos T/patologia
18.
J Infect Dis ; 196(7): 1093-101, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17763334

RESUMO

Subtilase cytotoxin (SubAB) is the prototype of a recently discovered AB(5) cytotoxin family produced by certain strains of Shiga toxigenic Escherichia coli (STEC). The catalytic A subunit is a highly specific subtilase-like serine protease that cleaves the endoplasmic reticulum chaperone BiP. The toxin is lethal for mice, but the pathology it induces is poorly understood. Here, we show that intraperitoneal injection of SubAB causes microangiopathic hemolytic anemia, thrombocytopenia, and renal impairment in mice--characteristics typical of Shiga toxin-induced hemolytic uremic syndrome. SubAB caused extensive microvascular thrombosis and other histologic damage in the brain, kidneys, and liver, as well as dramatic splenic atrophy. Peripheral blood leukocyte levels were increased at 24 h; there was also significant neutrophil infiltration in the liver, kidneys, and spleen and toxin-induced apoptosis at these sites. These findings raise the possibility that SubAB directly contributes to pathology in humans infected with strains of STEC that produce both Shiga toxin and SubAB.


Assuntos
Citotoxinas/toxicidade , Retículo Endoplasmático/patologia , Escherichia coli/patogenicidade , Subunidades Proteicas/toxicidade , Toxinas Shiga/toxicidade , Anemia Hemolítica/etiologia , Anemia Hemolítica/patologia , Animais , Apoptose , Encéfalo/patologia , Citotoxinas/administração & dosagem , Escherichia coli/metabolismo , Proteínas de Escherichia coli/administração & dosagem , Proteínas de Escherichia coli/toxicidade , Humanos , Rim/citologia , Rim/patologia , Fígado/citologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Subunidades Proteicas/administração & dosagem , Toxinas Shiga/administração & dosagem , Baço/citologia , Baço/patologia , Trombocitopenia/etiologia , Trombocitopenia/patologia
19.
Proc Natl Acad Sci U S A ; 103(25): 9667-72, 2006 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-16766659

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) 0157:H7 is a food-borne pathogen that can cause bloody diarrhea and, occasionally, acute renal failure as a consequence of Shiga toxin (Stx) production by the organism. Stxs are potent cytotoxins that are lethal to animals at low doses. Thus, Stxs not only harm the host but, as reported here, also significantly enhance the capacity of EHEC O157:H7 to adhere to epithelial cells and to colonize the intestines of mice. Tissue culture experiments showed that this toxin-mediated increase in bacterial adherence correlated with an Stx-evoked increase in a eukaryotic receptor for the EHEC O157:H7 attachment factor intimin.


Assuntos
Escherichia coli O157/crescimento & desenvolvimento , Escherichia coli O157/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Toxina Shiga/metabolismo , Toxina Shiga/toxicidade , Animais , Aderência Bacteriana , Linhagem Celular Tumoral , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Escherichia coli O157/citologia , Escherichia coli O157/genética , Regulação Bacteriana da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Mutação/genética , Fosfoproteínas/metabolismo , Subunidades Proteicas/metabolismo , Subunidades Proteicas/toxicidade , Proteínas de Ligação a RNA/metabolismo , Toxina Shiga/genética , Nucleolina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...