Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
FASEB J ; 33(4): 5755-5771, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30699302

RESUMEN

The antibiotic bacitracin (Bac) inhibits cell wall synthesis of gram-positive bacteria. Here, we discovered a totally different activity of Bac: the neutralization of bacterial exotoxins. Bac prevented intoxication of mammalian cells with the binary enterotoxins Clostridium botulinum C2, C. perfringens ι, C. difficile transferase (CDT), and Bacillus anthracis lethal toxin. The transport (B) subunits of these toxins deliver their respective enzyme (A) subunits into cells. Following endocytosis, the B subunits form pores in membranes of endosomes, which mediate translocation of the A subunits into the cytosol. Bac inhibited formation of such B pores in lipid bilayers in vitro and in living cells, thereby preventing translocation of the A subunit into the cytosol. Bac preserved the epithelial integrity of toxin-treated CaCo-2 monolayers, a model for the human gut epithelium. In conclusion, Bac should be discussed as a therapeutic option against infections with medically relevant toxin-producing bacteria, including C. difficile and B. anthracis, because it inhibits bacterial growth and neutralizes the secreted toxins.-Schnell, L., Felix, I., Müller, B., Sadi, M., von Bank, F., Papatheodorou, P., Popoff, M. R., Aktories, K., Waltenberger, E., Benz, R., Weichbrodt, C., Fauler, M., Frick, M., Barth, H. Revisiting an old antibiotic: bacitracin neutralizes binary bacterial toxins and protects cells from intoxication.


Asunto(s)
Antibacterianos/farmacología , Bacitracina/farmacología , Toxinas Bacterianas/metabolismo , Sustancias Protectoras/farmacología , Animales , Antígenos Bacterianos/metabolismo , Bacillus anthracis/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Chlorocebus aethiops , Clostridioides difficile/efectos de los fármacos , Citosol/efectos de los fármacos , Citosol/metabolismo , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Exotoxinas/metabolismo , Células HeLa , Humanos , Membrana Dobles de Lípidos/metabolismo , Transporte de Proteínas/efectos de los fármacos , Células Vero
2.
Curr Top Microbiol Immunol ; 406: 163-198, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27197646

RESUMEN

Bacterial ADP-ribosylating toxins are the causative agents for several severe human and animal diseases such as diphtheria, cholera, or enteric diseases. They display an AB-type structure: The enzymatically active A-domain attaches to the binding/translocation B-domain which then binds to a receptor on the cell surface. After receptor-mediated endocytosis, the B-domain facilitates the membrane translocation of the unfolded A-domain into the host cell cytosol. Here, the A-domain transfers an ADP-ribose moiety onto its specific substrate which leads to characteristic cellular effects and thus to severe clinical symptoms. Since the A-domain has to reach the cytosol to achieve a cytotoxic effect, the membrane translocation represents a crucial step during toxin uptake. Host cell chaperones including Hsp90 and protein-folding helper enzymes of the peptidyl-prolyl cis/trans isomerase (PPIase) type facilitate this membrane translocation of the unfolded A-domain for ADP-ribosylating toxins but not for toxins with a different enzyme activity. This review summarizes the uptake mechanisms of the ADP-ribosylating clostridial binary toxins, diphtheria toxin (DT) and cholera toxin (CT), with a special focus on the interaction of these toxins with the chaperones Hsp90 and Hsp70 and PPIases of the cyclophilin and FK506-binding protein families during the membrane translocation of their ADP-ribosyltransferase domains into the host cell cytosol. Moreover, the medical implications of host cell chaperones and PPIases as new drug targets for the development of novel therapeutic strategies against diseases caused by bacterial ADP-ribosylating toxins are discussed.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , Toxinas Bacterianas/metabolismo , Membrana Celular/enzimología , Membrana Celular/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Animales , Humanos , Transporte de Proteínas
3.
Life Sci Alliance ; 6(11)2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37604583

RESUMEN

The ubiquitin-like modifier FAT10 is up-regulated in many different cell types by IFNγ and TNFα (TNF) and directly targets proteins for proteasomal degradation. FAT10 gets covalently conjugated to its conjugation substrates by the E1 activating enzyme UBA6, the E2 conjugating enzyme USE1, and E3 ligases including Parkin. To date, USE1 was supposed to be the only E2 enzyme for FAT10ylation, and we show here that a knockout of USE1 strongly diminished FAT10 conjugation. Remarkably, under inflammatory conditions in the presence of TNF, FAT10 conjugation appears to be independent of USE1. We report on the identification of additional E2 conjugating enzymes, which were previously not associated with FAT10. We confirm their capacity to be charged with FAT10 onto their active site cysteine, and to rescue FAT10 conjugation in the absence of USE1. This finding strongly widens the field of FAT10 research by pointing to multiple, so far unknown pathways for the conjugation of FAT10, disclosing novel possibilities for pharmacological interventions to regulate FAT10 conjugation under inflammatory conditions and/or viral infections.


Asunto(s)
Inflamación , Factor de Necrosis Tumoral alfa , Ubiquitina , Cisteína , Ubiquitina/metabolismo , Humanos , Inflamación/metabolismo
4.
Stem Cells Int ; 2019: 4149762, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31467562

RESUMEN

Bacitracin is an established antibiotic for local application and inhibits the cell wall synthesis of Gram-positive bacteria. Recently, we discovered a completely different mode of action of bacitracin and reported that this drug protects human cells from intoxication by a variety of medically relevant bacterial protein toxins including CDT, the binary actin ADP-ribosylating toxin of Clostridium (C.) difficile. Bacitracin prevents the transport of CDT into the cytosol of target cells, most likely by inhibiting the transport function of the binding subunit of this toxin. Here, we tested the effect of bacitracin towards TcdB, a major virulence factor of C. difficile contributing to severe C. difficile-associated diseases (CDAD) including pseudomembranous colitis. Bacitracin protected stem cell-derived human intestinal organoids as well as human gut epithelial cells from intoxication with TcdB. Moreover, it prevented the TcdB-induced disruption of epithelia formed by gut epithelium cells in vitro and maintained the barrier function as detected by measuring transepithelial electrical resistance (TEER). In the presence of bacitracin, TcdB was not able reach its substrate Rac1 in the cytosol of human epithelial cells, most likely because its pH-dependent transport across cell membranes into the cytosol is decreased by bacitracin. In conclusion, in addition to its direct antibiotic activity against C. difficile and its inhibitory effect towards the toxin CDT, bacitracin neutralizes the exotoxin TcdB of this important pathogenic bacterium.

5.
Adv Healthc Mater ; 8(17): e1900665, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31318180

RESUMEN

The targeted pharmacological modulation of polymorphonuclear leukocytes (PMNs) is of major medical interest. These innate immune cells play a central role in the defense against pathogenic microorganisms. However, their excessive chemotactic recruitment into tissues after traumatic injury is detrimental due to local and systemic inflammation. Rho-GTPases, being the master regulators of the actin cytoskeleton, regulate migration and chemotaxis of PMNs, are attractive pharmacological targets. Herein, supramolecular protein complexes are assembled in a "mix-and-match" approach containing the specific Rho-inhibiting clostridial C3 enzyme and three PMN-binding peptides using an avidin platform. Selective delivery of the C3 Rho-inhibitor with these complexes into the cytosol of human neutrophil-like NB-4 cells and primary human PMNs ex vivo is demonstrated, where they catalyze the adenosine diphosphate (ADP) ribosylation of Rho and induce a characteristic change in cell morphology. Notably, the complexes do not deliver C3 enzyme into human lung epithelial cells, A549 lung cancer cells, and immortalized human alveolar epithelial cells (hAELVi), demonstrating their cell type-selectivity. The supramolecular complexes represent attractive molecular tools to decipher the role of PMNs in infection and inflammation or for the development of novel therapeutic approaches for diseases that are associated with hyperactivity and reactivity of PMNs such as post-traumatic injury.


Asunto(s)
Neutrófilos/metabolismo , Toxinas Biológicas/farmacología , ADP Ribosa Transferasas/metabolismo , Avidina/metabolismo , Biotinilación , Toxinas Botulínicas/metabolismo , Línea Celular , Citosol/metabolismo , Endocitosis/efectos de los fármacos , Humanos , Neutrófilos/efectos de los fármacos , Péptidos/síntesis química , Péptidos/química
6.
Sci Rep ; 7(1): 613, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28377614

RESUMEN

Diphtheria toxin kills human cells because it delivers its enzyme domain DTA into their cytosol where it inhibits protein synthesis. After receptor-mediated uptake of the toxin, DTA translocates from acidic endosomes into the cytosol, which might be assisted by host cell factors. Here we investigated the role of Hsp90 and its co-chaperones during the uptake of native diphtheria toxin into human cells and identified the components of the Hsp90 machinery including Hsp90, Hsp70, Cyp40 and the FK506 binding proteins FKBP51 and FKBP52 as DTA binding partners. Moreover, pharmacological inhibition of the chaperone activity of Hsp90 and Hsp70 and of the peptidyl-prolyl cis/trans isomerase (PPIase) activity of Cyps and FKBPs protected cells from intoxication with diphtheria toxin and inhibited the pH-dependent trans-membrane transport of DTA into the cytosol. In conclusion, these host cell factors facilitate toxin uptake into human cells, which might lead to development of novel therapeutic strategies against diphtheria.


Asunto(s)
Toxina Diftérica/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Animales , Células CHO , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Cricetulus , Citosol/metabolismo , Toxina Diftérica/toxicidad , Activación Enzimática/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Concentración de Iones de Hidrógeno , Modelos Biológicos , Chaperonas Moleculares/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Unión Proteica , Transporte de Proteínas , Proteolisis
7.
Toxicon ; 116: 23-8, 2016 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25911959

RESUMEN

During cellular uptake, diphtheria toxin delivers its catalytic domain DTA from acidified endosomes into the cytosol, which requires reduction of the disulfide linking DTA to the transport domain. In vitro, thioredoxin reduces this disulfide and thioredoxin reductase (TrxR) is part of a cytosolic complex facilitating DTA-translocation. We found that the TrxR-specific inhibitor auranofin prevented DTA delivery into the cytosol and intoxication of HeLa cells with diphtheria toxin, offering perspectives for novel pharmacological strategies against diphtheria.


Asunto(s)
Auranofina/farmacología , Transporte Biológico/efectos de los fármacos , Toxina Diftérica/metabolismo , Sustancias Protectoras/farmacología , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Citosol/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno
8.
Toxins (Basel) ; 8(7)2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27428999

RESUMEN

Diphtheria toxin is a single-chain protein toxin that invades human cells by receptor-mediated endocytosis. In acidic endosomes, its translocation domain inserts into endosomal membranes and facilitates the transport of the catalytic domain (DTA) from endosomal lumen into the host cell cytosol. Here, DTA ADP-ribosylates elongation factor 2 inhibits protein synthesis and leads to cell death. The compound 4-bromobenzaldehyde N-(2,6-dimethylphenyl)semicarbazone (EGA) has been previously shown to protect cells from various bacterial protein toxins which deliver their enzymatic subunits from acidic endosomes to the cytosol, including Bacillus anthracis lethal toxin and the binary clostridial actin ADP-ribosylating toxins C2, iota and Clostridium difficile binary toxin (CDT). Here, we demonstrate that EGA also protects human cells from diphtheria toxin by inhibiting the pH-dependent translocation of DTA across cell membranes. The results suggest that EGA might serve for treatment and/or prevention of the severe disease diphtheria.


Asunto(s)
Membrana Celular/efectos de los fármacos , Toxina Diftérica/toxicidad , Sustancias Protectoras/farmacología , Semicarbazonas/farmacología , Transporte Biológico , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Citoprotección , Toxina Diftérica/metabolismo , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Factores de Tiempo
9.
Toxins (Basel) ; 8(4): 101, 2016 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27043629

RESUMEN

The pathogenic bacteria Clostridium difficile, Clostridium perfringens and Clostridium botulinum produce the binary actin ADP-ribosylating toxins CDT, iota and C2, respectively. These toxins are composed of a transport component (B) and a separate enzyme component (A). When both components assemble on the surface of mammalian target cells, the B components mediate the entry of the A components via endosomes into the cytosol. Here, the A components ADP-ribosylate G-actin, resulting in depolymerization of F-actin, cell-rounding and eventually death. In the present study, we demonstrate that 4-bromobenzaldehyde N-(2,6-dimethylphenyl)semicarbazone (EGA), a compound that protects cells from multiple toxins and viruses, also protects different mammalian epithelial cells from all three binary actin ADP-ribosylating toxins. In contrast, EGA did not inhibit the intoxication of cells with Clostridium difficile toxins A and B, indicating a possible different entry route for this toxin. EGA does not affect either the binding of the C2 toxin to the cells surface or the enzyme activity of the A components of CDT, iota and C2, suggesting that this compound interferes with cellular uptake of the toxins. Moreover, for C2 toxin, we demonstrated that EGA inhibits the pH-dependent transport of the A component across cell membranes. EGA is not cytotoxic, and therefore, we propose it as a lead compound for the development of novel pharmacological inhibitors against clostridial binary actin ADP-ribosylating toxins.


Asunto(s)
ADP Ribosa Transferasas/toxicidad , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Toxinas Botulínicas/toxicidad , Semicarbazonas/farmacología , Actinas/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Animales , Membrana Celular/metabolismo , Chlorocebus aethiops , Células HeLa , Humanos , Transporte de Proteínas/efectos de los fármacos , Células Vero
10.
PLoS One ; 7(12): e51356, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23236484

RESUMEN

Various pathogenic clostridia produce binary protein toxins associated with enteric diseases of humans and animals. Separate binding/translocation (B) components bind to a protein receptor on the cell surface, assemble with enzymatic (A) component(s), and mediate endocytosis of the toxin complex. Ultimately there is translocation of A component(s) from acidified endosomes into the cytosol, leading to destruction of the actin cytoskeleton. Our results revealed that CD44, a multifunctional surface protein of mammalian cells, facilitates intoxication by the iota family of clostridial binary toxins. Specific antibody against CD44 inhibited cytotoxicity of the prototypical Clostridium perfringens iota toxin. Versus CD44(+) melanoma cells, those lacking CD44 bound less toxin and were dose-dependently resistant to C. perfringens iota, as well as Clostridium difficile and Clostridium spiroforme iota-like, toxins. Purified CD44 specifically interacted in vitro with iota and iota-like, but not related Clostridium botulinum C2, toxins. Furthermore, CD44 knockout mice were resistant to iota toxin lethality. Collective data reveal an important role for CD44 during intoxication by a family of clostridial binary toxins.


Asunto(s)
ADP Ribosa Transferasas/toxicidad , Toxinas Bacterianas/toxicidad , Endocitosis/fisiología , Receptores de Hialuranos/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Chlorocebus aethiops , Ditiotreitol/farmacología , Relación Dosis-Respuesta a Droga , Receptores de Hialuranos/genética , Inmunoprecipitación , Ratones , Ratones Noqueados , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA