Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Leukoc Biol ; 108(3): 773-786, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32421904

RESUMEN

Anthrax lethal toxin (LT) is a protease that activates the NLRP1b inflammasome sensor in certain rodent strains. Unlike better-studied sensors, relatively little is known about the priming requirements for NLRP1b. In this study, we investigate the rapid and striking priming-independent LT-induced release of IL-1ß in mice within hours of toxin challenge. We find IL-1ß release to be a NLRP1b- and caspase-1-dependent, NLRP3 and caspase-11-independent event that requires both neutrophils and peptidyl arginine deiminiase-4 (PAD4) activity. The simultaneous LT-induced IL-18 response is neutrophil-independent. Bone marrow reconstitution experiments in mice show toxin-induced IL-1ß originates from hematopoietic cells. LT treatment of neutrophils in vitro did not induce IL-1ß, neutrophil extracellular traps (NETs), or pyroptosis. Although platelets interact closely with neutrophils and are also a potential source of IL-1ß, they were unable to bind or endocytose LT and did not secrete IL-1ß in response to the toxin. LT-treated mice had higher levels of cell-free DNA and HMGB1 in circulation than PBS-treated controls, and treatment of mice with recombinant DNase reduced the neutrophil- and NLRP1-dependent IL-1ß release. DNA sensor AIM2 deficiency, however, did not impact IL-1ß release. These data, in combination with the findings on PAD4, suggest a possible role for in vivo NETs or cell-free DNA in cytokine induction in response to LT challenge. Our findings suggest a complex interaction of events and/or mediators in LT-treated mice with the neutrophil as a central player in induction of a profound and rapid inflammatory response to toxin.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Antígenos Bacterianos/toxicidad , Proteínas Reguladoras de la Apoptosis/fisiología , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/toxicidad , Trampas Extracelulares/fisiología , Interleucina-1beta/metabolismo , Neutrófilos/metabolismo , Arginina Deiminasa Proteína-Tipo 4/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Carbunco/inmunología , Antígenos Bacterianos/farmacología , Proteínas Reguladoras de la Apoptosis/deficiencia , Bacillus anthracis/fisiología , Toxinas Bacterianas/farmacología , Inflamasomas/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Congénicos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Monocitos/efectos de los fármacos , Monocitos/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Neutrófilos/efectos de los fármacos , Arginina Deiminasa Proteína-Tipo 4/deficiencia , Piroptosis/efectos de los fármacos , Quimera por Radiación , Especificidad de la Especie , Esporas Bacterianas
2.
Proc Natl Acad Sci U S A ; 115(4): E733-E742, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311317

RESUMEN

Protein-based drugs are very active in treating cancer, but their efficacy can be limited by the formation of neutralizing antidrug antibodies (ADAs). Recombinant immunotoxins are proteins that are very effective in patients with leukemia, where immunity is suppressed, but induce ADAs, which compromise their activity, in patients with intact immunity. Here we induced a specific, durable, and transferable immune tolerance to recombinant immunotoxins by combining them with nanoparticles containing rapamycin (SVP-R). SVP-R mitigated the formation of inhibitory ADAs in naïve and sensitized mice, resulting in restoration of antitumor activity. The immune tolerance is mediated by colocalization of the SVP-R and immunotoxin to dendritic cells and macrophages in the spleen and is abrogated by depletion of regulatory T cells. Tolerance induced by SVPs was not blocked by checkpoint inhibitors or costimulatory agonist monoclonal antibodies that by themselves enhance ADA formation.


Asunto(s)
Inmunomodulación , Inmunosupresores/administración & dosificación , Inmunotoxinas/administración & dosificación , Leucemia/terapia , Sirolimus/administración & dosificación , Animales , Anticuerpos Neutralizantes , Proteínas Ligadas a GPI/inmunología , Humanos , Inmunotoxinas/inmunología , Mesotelina , Nanopartículas , Factores de Tiempo
3.
Cell Mol Immunol ; 14(5): 432-442, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-26477977

RESUMEN

Antibodies against the toxin portion of recombinant immunotoxins (RIT) reduce their efficacy and pose a potential safety risk. To overcome this problem we mutated the very immunogenic immunotoxin SS1P to produce LMB-T20, a de-immunized RIT that has the eight human T-cell epitopes in SS1P modified or removed. To determine the effect of T-cell epitope removal in vivo we mapped the T-cell epitopes in immune-competent BALB/c mice and found that these mice recognize two epitopes. One corresponds to the human immunodominant T-cell epitope and the other to a human subdominant epitope; both were eliminated in LMB-T20. We found that mice immunized with LMB-T20 did not have T-cell activation and did not develop anti-drug antibodies (ADA), whereas mice immunized with SS1P, showed T-cell activation, and developed ADA detected by both ELISA and drug neutralizing assays. The ability of the mice treated with LMB-T20 to respond to other antigens was not compromised. We conclude that elimination of T-cell epitopes is sufficient to prevent formation of antibodies to an immunogenic foreign protein.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos/inmunología , Formación de Anticuerpos/inmunología , Epítopos de Linfocito T/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Femenino , Humanos , Inmunización , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C
4.
J Biol Chem ; 290(10): 6584-95, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25564615

RESUMEN

Anthrax disease is caused by a toxin consisting of protective antigen (PA), lethal factor, and edema factor. Antibodies against PA have been shown to be protective against the disease. Variable domains of camelid heavy chain-only antibodies (VHHs) with affinity for PA were obtained from immunized alpacas and screened for anthrax neutralizing activity in macrophage toxicity assays. Two classes of neutralizing VHHs were identified recognizing distinct, non-overlapping epitopes. One class recognizes domain 4 of PA at a well characterized neutralizing site through which PA binds to its cellular receptor. A second neutralizing VHH (JKH-C7) recognizes a novel epitope. This antibody inhibits conversion of the PA oligomer from "pre-pore" to its SDS and heat-resistant "pore" conformation while not preventing cleavage of full-length 83-kDa PA (PA83) by cell surface proteases to its oligomer-competent 63-kDa form (PA63). The antibody prevents endocytosis of the cell surface-generated PA63 subunit but not preformed PA63 oligomers formed in solution. JKH-C7 and the receptor-blocking VHH class (JIK-B8) were expressed as a heterodimeric VHH-based neutralizing agent (VNA2-PA). This VNA displayed improved neutralizing potency in cell assays and protected mice from anthrax toxin challenge with much better efficacy than the separate component VHHs. The VNA protected virtually all mice when separately administered at a 1:1 ratio to toxin and protected mice against Bacillus anthracis spore infection. Thus, our studies show the potential of VNAs as anthrax therapeutics. Due to their simple and stable nature, VNAs should be amenable to genetic delivery or administration via respiratory routes.


Asunto(s)
Carbunco/inmunología , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Animales , Carbunco/microbiología , Carbunco/patología , Carbunco/terapia , Anticuerpos Antibacterianos/administración & dosificación , Bacillus anthracis/inmunología , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/antagonistas & inhibidores , Camélidos del Nuevo Mundo/inmunología , Epítopos/inmunología , Humanos , Ratones , Esporas/inmunología , Esporas/patogenicidad
5.
PLoS Pathog ; 10(3): e1003927, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24626226

RESUMEN

Toxoplasma gondii is an intracellular parasite that infects a wide range of warm-blooded species. Rats vary in their susceptibility to this parasite. The Toxo1 locus conferring Toxoplasma resistance in rats was previously mapped to a region of chromosome 10 containing Nlrp1. This gene encodes an inflammasome sensor controlling macrophage sensitivity to anthrax lethal toxin (LT) induced rapid cell death (pyroptosis). We show here that rat strain differences in Toxoplasma infected macrophage sensitivity to pyroptosis, IL-1ß/IL-18 processing, and inhibition of parasite proliferation are perfectly correlated with NLRP1 sequence, while inversely correlated with sensitivity to anthrax LT-induced cell death. Using recombinant inbred rats, SNP analyses and whole transcriptome gene expression studies, we narrowed the candidate genes for control of Toxoplasma-mediated rat macrophage pyroptosis to four genes, one of which was Nlrp1. Knockdown of Nlrp1 in pyroptosis-sensitive macrophages resulted in higher parasite replication and protection from cell death. Reciprocally, overexpression of the NLRP1 variant from Toxoplasma-sensitive macrophages in pyroptosis-resistant cells led to sensitization of these resistant macrophages. Our findings reveal Toxoplasma as a novel activator of the NLRP1 inflammasome in rat macrophages.


Asunto(s)
Inflamasomas/inmunología , Macrófagos/parasitología , Proteínas del Tejido Nervioso/inmunología , Toxoplasmosis/inmunología , Animales , Western Blotting , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Predisposición Genética a la Enfermedad/genética , Inflamasomas/genética , Macrófagos/inmunología , Proteínas del Tejido Nervioso/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Ratas , Ratas Endogámicas , Toxoplasmosis/genética , Transcriptoma
6.
mBio ; 5(1)2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24549849

RESUMEN

UNLABELLED: Induction of immunity that limits Toxoplasma gondii infection in mice is critically dependent on the activation of the innate immune response. In this study, we investigated the role of cytoplasmic nucleotide-binding domain and leucine-rich repeat containing a pyrin domain (NLRP) inflammasome sensors during acute toxoplasmosis in mice. We show that in vitro Toxoplasma infection of murine bone marrow-derived macrophages activates the NLRP3 inflammasome, resulting in the rapid production and cleavage of interleukin-1ß (IL-1ß), with no measurable cleavage of IL-18 and no pyroptosis. Paradoxically, Toxoplasma-infected mice produced large quantities of IL-18 but had no measurable IL-1ß in their serum. Infection of mice deficient in NLRP3, caspase-1/11, IL-1R, or the inflammasome adaptor protein ASC led to decreased levels of circulating IL-18, increased parasite replication, and death. Interestingly, mice deficient in NLRP1 also displayed increased parasite loads and acute mortality. Using mice deficient in IL-18 and IL-18R, we show that this cytokine plays an important role in limiting parasite replication to promote murine survival. Our findings reveal T. gondii as a novel activator of the NLRP1 and NLRP3 inflammasomes in vivo and establish a role for these sensors in host resistance to toxoplasmosis. IMPORTANCE: Inflammasomes are multiprotein complexes that are a major component of the innate immune system. They contain "sensor" proteins that are responsible for detecting various microbial and environmental danger signals and function by activating caspase-1, an enzyme that mediates cleavage and release of the proinflammatory cytokines interleukin-1ß (IL-1ß) and IL-18. Toxoplasma gondii is a highly successful protozoan parasite capable of infecting a wide range of host species that have variable levels of resistance. We report here that T. gondii is a novel activator of the NLRP1 and NLRP3 inflammasomes in vivo and establish a role for these sensors in host resistance to toxoplasmosis. Using mice deficient in IL-18 and IL-18R, we show that the IL-18 cytokine plays a pivotal role by limiting parasite replication to promote murine survival.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas Portadoras/inmunología , Resistencia a la Enfermedad , Inflamasomas/inmunología , Toxoplasma/inmunología , Toxoplasmosis Animal/inmunología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Portadoras/genética , Femenino , Macrófagos/inmunología , Macrófagos/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Carga de Parásitos , Análisis de Supervivencia
7.
J Immunol ; 192(2): 763-70, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24337744

RESUMEN

Inflammasomes are large cytoplasmic multiprotein complexes that activate caspase-1 in response to diverse intracellular danger signals. Inflammasome components termed nucleotide-binding oligomerization domain-like receptor (NLR) proteins act as sensors for pathogen-associated molecular patterns, stress, or danger stimuli. We discovered that arsenicals, including arsenic trioxide and sodium arsenite, inhibited activation of the NLRP1, NLRP3, and NAIP5/NLRC4 inflammasomes by their respective activating signals, anthrax lethal toxin, nigericin, and flagellin. These compounds prevented the autoproteolytic activation of caspase-1 and the processing and secretion of IL-1ß from macrophages. Inhibition was independent of protein synthesis induction, proteasome-mediated protein breakdown, or kinase signaling pathways. Arsenic trioxide and sodium arsenite did not directly modify or inhibit the activity of preactivated recombinant caspase-1. Rather, they induced a cellular state inhibitory to both the autoproteolytic and substrate cleavage activities of caspase-1, which was reversed by the reactive oxygen species scavenger N-acetylcysteine but not by reducing agents or NO pathway inhibitors. Arsenicals provided protection against NLRP1-dependent anthrax lethal toxin-mediated cell death and prevented NLRP3-dependent neutrophil recruitment in a monosodium urate crystal inflammatory murine peritonitis model. These findings suggest a novel role in inhibition of the innate immune response for arsenical compounds that have been used as therapeutics for a few hundred years.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Arsenicales/farmacología , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/metabolismo , Inflamasomas/efectos de los fármacos , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Óxidos/farmacología , Animales , Antígenos Bacterianos/farmacología , Trióxido de Arsénico , Arsenitos/farmacología , Toxinas Bacterianas/farmacología , Caspasa 1/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Flagelina/farmacología , Inmunidad Innata/efectos de los fármacos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteína con Dominio Pirina 3 de la Familia NLR , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Nigericina/farmacología , Óxidos de Nitrógeno/metabolismo , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , Proteolisis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Compuestos de Sodio/farmacología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
8.
PLoS One ; 8(10): e76955, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204713

RESUMEN

Bacillus cereus is a spore-forming, Gram-positive bacterium commonly associated with outbreaks of food poisoning. It is also known as an opportunistic pathogen causing clinical infections such as bacteremia, meningitis, pneumonia, and gas gangrene-like cutaneous infections, mostly in immunocompromised patients. B. cereus secretes a plethora of toxins of which four are associated with the symptoms of food poisoning. Two of these, the non-hemolytic enterotoxin Nhe and the hemolysin BL (Hbl) toxin, are predicted to be structurally similar and are unique in that they require the combined action of three toxin proteins to induce cell lysis. Despite their dominant role in disease, the molecular mechanism of their toxic function is still poorly understood. We report here that B. cereus strain ATCC 10876 harbors not only genes encoding Nhe, but also two copies of the hbl genes. We identified Hbl as the major secreted toxin responsible for inducing rapid cell lysis both in cultured cells and in an intraperitoneal mouse toxicity model. Antibody neutralization and deletion of Hbl-encoding genes resulted in significant reductions of cytotoxic activity. Microscopy studies with Chinese Hamster Ovary cells furthermore showed that pore formation by both Hbl and Nhe occurs through a stepwise, sequential binding of toxin components to the cell surface and to each other. This begins with binding of Hbl-B or NheC to the eukaryotic membrane, and is followed by the recruitment of Hbl-L1 or NheB, respectively, followed by the corresponding third protein. Lastly, toxin component complementation studies indicate that although Hbl and Nhe can be expressed simultaneously and are predicted to be structurally similar, they are incompatible and cannot complement each other.


Asunto(s)
Bacillus cereus/metabolismo , Proteínas Bacterianas/metabolismo , Enterotoxinas/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/farmacología , Animales , Bacillus cereus/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/farmacología , Células CHO , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cricetinae , Cricetulus , Medios de Cultivo Condicionados/farmacología , Enterotoxinas/genética , Enterotoxinas/farmacología , Dosificación de Gen , Orden Génico , Prueba de Complementación Genética , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/farmacología , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Microscopía Confocal , Mutación , Unión Proteica
9.
PLoS One ; 8(8): e74474, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24015319

RESUMEN

Anthrax edema factor (EF) is a calmodulin-dependent adenylate cyclase that converts adenosine triphosphate (ATP) into 3'-5'-cyclic adenosine monophosphate (cAMP), contributing to the establishment of Bacillus anthracis infections and the resulting pathophysiology. We show that EF adenylate cyclase toxin activity is strongly mediated by the N-end rule, and thus is dependent on the identity of the N-terminal amino acid. EF variants having different N-terminal residues varied by more than 100-fold in potency in cultured cells and mice. EF variants having unfavorable, destabilizing N-terminal residues showed much greater activity in cells when the E1 ubiquitin ligase was inactivated or when proteasome inhibitors were present. Taken together, these results show that EF is uniquely affected by ubiquitination and/or proteasomal degradation.


Asunto(s)
Adenilil Ciclasas/metabolismo , Antígenos Bacterianos/metabolismo , Bacillus anthracis/enzimología , Toxinas Bacterianas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Adenilil Ciclasas/genética , Animales , Antígenos Bacterianos/genética , Bacillus anthracis/genética , Toxinas Bacterianas/genética , Línea Celular , Ratones
10.
Nature ; 501(7465): 63-8, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23995686

RESUMEN

Bacillus anthracis, the causative agent of anthrax disease, is lethal owing to the actions of two exotoxins: anthrax lethal toxin (LT) and oedema toxin (ET). The key tissue targets responsible for the lethal effects of these toxins are unknown. Here we generated cell-type-specific anthrax toxin receptor capillary morphogenesis protein-2 (CMG2)-null mice and cell-type-specific CMG2-expressing mice and challenged them with the toxins. Our results show that lethality induced by LT and ET occurs through damage to distinct cell types; whereas targeting cardiomyocytes and vascular smooth muscle cells is required for LT-induced mortality, ET-induced lethality occurs mainly through its action in hepatocytes. Notably, and in contradiction to what has been previously postulated, targeting of endothelial cells by either toxin does not seem to contribute significantly to lethality. Our findings demonstrate that B. anthracis has evolved to use LT and ET to induce host lethality by coordinately damaging two distinct vital systems.


Asunto(s)
Antígenos Bacterianos/toxicidad , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/toxicidad , Animales , Carbunco/genética , Carbunco/metabolismo , Carbunco/microbiología , Resistencia a la Enfermedad/genética , Edema/inducido químicamente , Células Endoteliales/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Intestinos/patología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Especificidad de Órganos/efectos de los fármacos , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Especificidad por Sustrato/efectos de los fármacos , Análisis de Supervivencia
11.
Antimicrob Agents Chemother ; 57(9): 4139-45, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23774434

RESUMEN

Bacillus anthracis, the causative agent of anthrax, manifests its pathogenesis through the action of two secreted toxins. The bipartite lethal and edema toxins, a combination of lethal factor or edema factor with the protein protective antigen, are important virulence factors for this bacterium. We previously developed small-molecule inhibitors of lethal factor proteolytic activity (LFIs) and demonstrated their in vivo efficacy in a rat lethal toxin challenge model. In this work, we show that these LFIs protect against lethality caused by anthrax infection in mice when combined with subprotective doses of either antibiotics or neutralizing monoclonal antibodies that target edema factor. Significantly, these inhibitors provided protection against lethal infection when administered as a monotherapy. As little as two doses (10 mg/kg) administered at 2 h and 8 h after spore infection was sufficient to provide a significant survival benefit in infected mice. Administration of LFIs early in the infection was found to inhibit dissemination of vegetative bacteria to the organs in the first 32 h following infection. In addition, neutralizing antibodies against edema factor also inhibited bacterial dissemination with similar efficacy. Together, our findings confirm the important roles that both anthrax toxins play in establishing anthrax infection and demonstrate the potential for small-molecule therapeutics targeting these proteins.


Asunto(s)
Carbunco/tratamiento farmacológico , Antibacterianos/farmacología , Anticuerpos Neutralizantes/farmacología , Bacillus anthracis/efectos de los fármacos , Toxinas Bacterianas/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Esporas Bacterianas/efectos de los fármacos , Animales , Carbunco/microbiología , Carbunco/mortalidad , Antibacterianos/farmacocinética , Antígenos Bacterianos , Bacillus anthracis/crecimiento & desarrollo , Esquema de Medicación , Femenino , Ratones , Ratones Endogámicos C57BL , Peso Molecular , Inhibidores de Proteasas/farmacocinética , Esporas Bacterianas/crecimiento & desarrollo , Análisis de Supervivencia , Factores de Tiempo
12.
BMC Genomics ; 14: 188, 2013 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-23506131

RESUMEN

BACKGROUND: Signals of danger and damage in the cytosol of cells are sensed by NOD-like receptors (NLRs), which are components of multiprotein complexes called inflammasomes. Inflammasomes activate caspase-1, resulting in IL-1-beta and IL-18 secretion and an inflammatory response. To date, the only known activator of rodent Nlrp1 is anthrax lethal toxin (LT), a protease secreted by the bacterial pathogen Bacillus anthracis. Although susceptibility of mouse macrophages to LT has been genetically linked to Nlrp1b, mice harbor two additional Nlrp1 paralogs in their genomes (Nlrp1a and Nlrp1c). However, little is known about their expression profile and sequence in different mouse strains. Furthermore, simultaneous expression of these paralogs may lead to competitional binding of Nlrp1b interaction partners needed for inflammasome activation, thus influencing macrophages susceptibility to LT. To more completely understand the role(s) of Nlrp1 paralogs in mice, we surveyed for their expression in a large set of LT-resistant and sensitive mouse macrophages. In addition, we provide sequence comparisons for Nlrp1a and report on previously unrecognized splice variants of Nlrp1b. RESULTS: Our results show that macrophages from some inbred mouse strains simultaneously express different splice variants of Nlrp1b. In contrast to the highly polymorphic Nlrp1b splice variants, sequencing of expressed Nlrp1a showed the protein to be highly conserved across all mouse strains. We found that Nlrp1a was expressed only in toxin-resistant macrophages, with the sole exception of expression in LT-sensitive CAST/EiJ macrophages. CONCLUSIONS: Our data present a complex picture of Nlrp1 protein variations and provide a basis for elucidating their roles in murine macrophage function. Furthermore, the high conservation of Nlrp1a implies that it might be an important inflammasome sensor in mice.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Ratones Endogámicos/genética , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Empalme Alternativo , Animales , Antígenos Bacterianos/farmacología , Proteínas Reguladoras de la Apoptosis/biosíntesis , Toxinas Bacterianas/farmacología , Inflamasomas/metabolismo , Ratones
13.
J Biol Chem ; 288(13): 9058-65, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23393143

RESUMEN

Anthrax toxin protective antigen (PA) delivers its effector proteins into the host cell cytosol through formation of an oligomeric pore, which can assume heptameric or octameric states. By screening a highly directed library of PA mutants, we identified variants that complement each other to exclusively form octamers. These PA variants were individually nontoxic and demonstrated toxicity only when combined with their complementary partner. We then engineered requirements for activation by matrix metalloproteases and urokinase plasminogen activator into two of these variants. The resulting therapeutic toxin specifically targeted cells expressing both tumor associated proteases and completely stopped tumor growth in mice when used at a dose far below that which caused toxicity. This scheme for obtaining intercomplementing subunits can be employed with other oligomeric proteins and potentially has wide application.


Asunto(s)
Antígenos Bacterianos/química , Toxinas Bacterianas/química , Neoplasias/tratamiento farmacológico , Animales , Bacillus anthracis/metabolismo , Línea Celular Tumoral , Femenino , Biblioteca de Genes , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Conformación Molecular , Mutación , Neoplasias/metabolismo , Plásmidos/metabolismo , Conformación Proteica , Ingeniería de Proteínas/métodos , Mapeo de Interacción de Proteínas/métodos , Estructura Terciaria de Proteína , Proteínas/química , Ultracentrifugación
14.
Proc Natl Acad Sci U S A ; 109(34): 13817-22, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22869748

RESUMEN

To study the role of the diphthamide modification on eukaryotic elongation factor 2 (eEF2), we generated an eEF2 Gly(717)Arg mutant mouse, in which the first step of diphthamide biosynthesis is prevented. Interestingly, the Gly(717)-to-Arg mutation partially compensates the eEF2 functional loss resulting from diphthamide deficiency, possibly because the added +1 charge compensates for the loss of the +1 charge on diphthamide. Therefore, in contrast to mouse embryonic fibroblasts (MEFs) from OVCA1(-/-) mice, eEF2(G717R/G717R) MEFs retain full activity in polypeptide elongation and have normal growth rates. Furthermore, eEF2(G717R/G717R) mice showed milder phenotypes than OVCA1(-/-) mice (which are 100% embryonic lethal) and a small fraction survived to adulthood without obvious abnormalities. Moreover, eEF2(G717R/G717R)/OVCA1(-/-) double mutant mice displayed the milder phenotypes of the eEF2(G717R/G717R) mice, suggesting that the embryonic lethality of OVCA1(-/-) mice is due to diphthamide deficiency. We confirmed that the diphthamide modification is essential for eEF2 to prevent -1 frameshifting during translation and show that the Gly(717)-to-Arg mutation cannot rescue this defect.


Asunto(s)
Histidina/análogos & derivados , Factor 2 de Elongación Peptídica/metabolismo , Biosíntesis de Proteínas , Adenosina Difosfato/química , Animales , Biotina/química , Células CHO , Cricetinae , Fibroblastos/citología , Eliminación de Gen , Histidina/farmacología , Ratones , Ratones Transgénicos , Antígenos de Histocompatibilidad Menor , Péptidos/química , Fenotipo , Proteínas Supresoras de Tumor/genética
15.
Bioorg Med Chem Lett ; 22(6): 2242-6, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22342144

RESUMEN

Four core structures capable of providing sub-nanomolar inhibitors of anthrax lethal factor (LF) were evaluated by comparing the potential for toxicity, physicochemical properties, in vitro ADME profiles, and relative efficacy in a rat lethal toxin (LT) model of LF intoxication. Poor efficacy in the rat LT model exhibited by the phenoxyacetic acid series (3) correlated with low rat microsome and plasma stability. Specific molecular interactions contributing to the high affinity of inhibitors with a secondary amine in the C2-side chain were revealed by X-ray crystallography.


Asunto(s)
Acetatos/síntesis química , Carbunco/tratamiento farmacológico , Antídotos/síntesis química , Bacillus anthracis/efectos de los fármacos , Toxinas Bacterianas/antagonistas & inhibidores , Acetatos/farmacocinética , Acetatos/farmacología , Animales , Antídotos/farmacocinética , Antídotos/farmacología , Antígenos Bacterianos , Bacillus anthracis/fisiología , Cristalografía por Rayos X , Citocromo P-450 CYP2D6/metabolismo , Inhibidores del Citocromo P-450 CYP2D6 , Microsomas Hepáticos/enzimología , Modelos Moleculares , Conejos , Ratas
16.
Mol Microbiol ; 83(1): 96-109, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22103323

RESUMEN

Bacterial lipoproteins play a crucial role in virulence in some gram-positive bacteria. However, the role of lipoprotein biosynthesis in Bacillus anthracis is unknown. We created a B. anthracis mutant strain altered in lipoproteins by deleting the lgt gene encoding the enzyme prolipoprotein diacylglyceryl transferase, which attaches the lipid anchor to prolipoproteins. (14)C-palmitate labelling confirmed that the mutant strain lacked lipoproteins, and hydrocarbon partitioning showed it to have decreased surface hydrophobicity. The anthrax toxin proteins were secreted from the mutant strain at nearly the same levels as from the wild-type strain. The TLR2-dependent TNF-α response of macrophages to heat-killed lgt mutant bacteria was reduced. Spores of the lgt mutant germinated inefficiently in vitro and in mouse skin. As a result, in a murine subcutaneous infection model, lgt mutant spores had markedly attenuated virulence. In contrast, vegetative cells of the lgt mutant were as virulent as those of the wild-type strain. Thus, lipoprotein biosynthesis in B. anthracis is required for full virulence in a murine infection model.


Asunto(s)
Carbunco/microbiología , Bacillus anthracis/enzimología , Bacillus anthracis/patogenicidad , Proteínas Bacterianas/metabolismo , Lipoproteínas/biosíntesis , Precursores de Proteínas/biosíntesis , Esporas Bacterianas/crecimiento & desarrollo , Transferasas/metabolismo , Animales , Bacillus anthracis/genética , Bacillus anthracis/metabolismo , Proteínas Bacterianas/genética , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Esporas Bacterianas/enzimología , Esporas Bacterianas/metabolismo , Transferasas/genética , Virulencia
17.
Infect Immun ; 80(2): 529-38, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22104108

RESUMEN

The anthrax edema toxin (ET) of Bacillus anthracis is composed of the receptor-binding component protective antigen (PA) and of the adenylyl cyclase catalytic moiety, edema factor (EF). Uptake of ET into cells raises intracellular concentrations of the secondary messenger cyclic AMP, thereby impairing or activating host cell functions. We report here on a new consequence of ET action in vivo. We show that in mouse models of toxemia and infection, serum PA concentrations were significantly higher in the presence of enzymatically active EF. These higher concentrations were not caused by ET-induced inhibition of PA endocytosis; on the contrary, ET induced increased PA binding and uptake of the PA oligomer in vitro and in vivo through upregulation of the PA receptors TEM8 and CMG2 in both myeloid and nonmyeloid cells. ET effects on protein clearance from circulation appeared to be global and were not limited to PA. ET also impaired the clearance of ovalbumin, green fluorescent protein, and EF itself, as well as the small molecule biotin when these molecules were coinjected with the toxin. Effects on injected protein levels were not a result of general increase in protein concentrations due to fluid loss. Functional markers for liver and kidney were altered in response to ET. Concomitantly, ET caused phosphorylation and activation of the aquaporin-2 water channel present in the principal cells of the collecting ducts of the kidneys that are responsible for fluid homeostasis. Our data suggest that in vivo, ET alters circulatory protein and small molecule pharmacokinetics by an as-yet-undefined mechanism, thereby potentially allowing a prolonged circulation of anthrax virulence factors such as EF during infection.


Asunto(s)
Carbunco/metabolismo , Antígenos Bacterianos/toxicidad , Bacillus anthracis/metabolismo , Toxinas Bacterianas/toxicidad , Animales , Carbunco/inmunología , Antígenos Bacterianos/sangre , Bacillus anthracis/inmunología , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/sangre , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Células CHO , Cricetinae , Femenino , Regulación de la Expresión Génica/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos , Receptores de Superficie Celular , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo
18.
Infect Immun ; 79(11): 4609-16, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21911463

RESUMEN

Bacillus anthracis is the causative agent of anthrax, and the tripartite anthrax toxin is an essential element of its pathogenesis. Edema factor (EF), a potent adenylyl cyclase, is one of the toxin components. In this work, anti-EF monoclonal antibodies (MAb) were produced following immunization of mice, and four of the antibodies were fully characterized. MAb 3F2 has an affinity of 388 pM, was most effective for EF detection, and appears to be the first antibody reported to neutralize EF by binding to the catalytic C(B) domain. MAb 7F10 shows potent neutralization of edema toxin activity in vitro and in vivo; it targets the N-terminal protective antigen binding domain. The four MAb react with three different domains of edema factor, and all were able to detect purified edema factor in Western blot analysis. None of the four MAb cross-reacted with the lethal factor toxin component. Three of the four MAb protected mice in both a systemic edema toxin challenge model and a subcutaneous spore-induced foreleg edema model. A combination of three of the MAb also significantly delayed the time to death in a third subcutaneous spore challenge model. This appears to be the first direct evidence that monoclonal antibody-mediated neutralization of EF alone is sufficient to delay anthrax disease progression.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Animales , Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/toxicidad , Línea Celular , Relación Dosis-Respuesta a Droga , Edema/inducido químicamente , Edema/prevención & control , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Hibridomas , Inmunización , Inmunoglobulina G , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C
19.
Bioorg Med Chem Lett ; 21(7): 2030-3, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21334206

RESUMEN

New anthrax lethal factor inhibitors (LFIs) were designed based upon previously identified potent inhibitors 1a and 2. Combining the new core structures with modifications to the C2-side chain yielded analogs with improved efficacy in the rat lethal toxin model.


Asunto(s)
Antídotos/uso terapéutico , Antígenos Bacterianos/envenenamiento , Toxinas Bacterianas/envenenamiento , Animales , Estructura Molecular , Intoxicación/tratamiento farmacológico , Ratas
20.
Infect Immun ; 79(1): 118-24, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20974827

RESUMEN

MyD88-deficient mice were previously shown to have increased susceptibility to Bacillus anthracis infection relative to wild-type animals. To determine the mechanism by which MyD88 protects against B. anthracis infection, knockout mice were challenged with nonencapsulated, toxigenic B. anthracis or with anthrax toxins. MyD88-deficient mice had increased susceptibility to B. anthracis and anthrax lethal toxin but not to edema toxin. Lethal toxin alone induced marked multifocal intestinal ulcers in the knockout animals, compromising the intestinal epithelial barrier. The resulting enteric bacterial leakage in the knockout animals led to peritonitis and septicemia. Focal ulcers and erosion were also found in MyD88-heterozygous control mice but with far lower incidence and severity. B. anthracis infection also induced a similar enteric bacterial septicemia in MyD88-deficient mice but not in heterozygous controls. We show that lethal toxin and B. anthracis challenge induce bacteremia as a result of intestinal damage in MyD88-deficient mice. These results suggest that loss of the intestinal epithelial barrier and enteric bacterial septicemia may contribute to sensitizing MyD88-deficient mice to B. anthracis and that MyD88 plays a protective role against lethal toxin-induced impairment of intestinal barrier.


Asunto(s)
Carbunco/patología , Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/toxicidad , Intestinos/efectos de los fármacos , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal/fisiología , Animales , Carbunco/microbiología , Bacteriemia/microbiología , Predisposición Genética a la Enfermedad , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Esporas Bacterianas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...