Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 5: e1003, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24434511

RESUMEN

Cytolethal distending toxin (Cdt) is produced by Gram-negative bacteria of several species. It is composed of three subunits, CdtA, CdtB, and CdtC, with CdtB being the catalytic subunit. We fused CdtB from Haemophilus ducreyi to the N-terminal 255 amino acids of Bacillus anthracis toxin lethal factor (LFn) to design a novel, potentially potent antitumor drug. As a result of this fusion, CdtB was transported into the cytosol of targeted cells via the efficient delivery mechanism of anthrax toxin. The fusion protein efficiently killed various human tumor cell lines by first inducing a complete cell cycle arrest in the G2/M phase, followed by induction of apoptosis. The fusion protein showed very low toxicity in mouse experiments and impressive antitumor effects in a Lewis Lung carcinoma model, with a 90% cure rate. This study demonstrates that efficient drug delivery by a modified anthrax toxin system combined with the enzymatic activity of CdtB has great potential as anticancer treatment and should be considered for the development of novel anticancer drugs.


Asunto(s)
Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Neoplasias/tratamiento farmacológico , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Apoptosis/efectos de los fármacos , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/fisiopatología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología
2.
Infect Immun ; 71(11): 6591-606, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14573681

RESUMEN

Bacillus anthracis is nonhemolytic, even though it is closely related to the highly hemolytic Bacillus cereus. Hemolysis by B. cereus results largely from the action of phosphatidylcholine-specific phospholipase C (PC-PLC) and sphingomyelinase (SPH), encoded by the plc and sph genes, respectively. In B. cereus, these genes are organized in an operon regulated by the global regulator PlcR. B. anthracis contains a highly similar cereolysin operon, but it is transcriptionally silent because the B. anthracis PlcR is truncated at the C terminus. Here we report the cloning, expression, purification, and enzymatic characterization of PC-PLC and SPH from B. cereus and B. anthracis. We also investigated the effects of expressing PlcR on the expression of plc and sph. In B. cereus, PlcR was found to be a positive regulator of plc but a negative regulator of sph. Replacement of the B. cereus plcR gene by its truncated orthologue from B. anthracis eliminated the activities of both PC-PLC and SPH, whereas introduction into B. anthracis of the B. cereus plcR gene with its own promoter did not activate cereolysin expression. Hemolytic activity was detected in B. anthracis strains containing the B. cereus plcR gene on a multicopy plasmid under control of the strong B. anthracis protective antigen gene promoter or in a strain carrying a multicopy plasmid containing the entire B. cereus plc-sph operon. Slight hemolysis and PC-PLC activation were found when PlcR-producing B. anthracis strains were grown under anaerobic-plus-CO(2) or especially under aerobic-plus-CO(2) conditions. Unmodified parental B. anthracis strains did not demonstrate obvious hemolysis under the same conditions.


Asunto(s)
Bacillus cereus/enzimología , Esfingomielina Fosfodiesterasa/metabolismo , Transactivadores/fisiología , Fosfolipasas de Tipo C/metabolismo , Secuencia de Aminoácidos , Animales , Bacillus anthracis/enzimología , Bacillus anthracis/crecimiento & desarrollo , Bacillus cereus/genética , Proteínas Bacterianas/genética , Secuencia de Bases , Dióxido de Carbono/farmacología , Hemólisis , Sueros Inmunes/inmunología , Datos de Secuencia Molecular , Operón , Plásmidos , Regiones Promotoras Genéticas , Conejos , Ovinos , Esfingomielina Fosfodiesterasa/genética , Transactivadores/genética , Transactivadores/inmunología , Fosfolipasas de Tipo C/genética
3.
J Ind Microbiol Biotechnol ; 28(4): 232-8, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11986925

RESUMEN

The protective antigen (PA) is one of the three components of the anthrax toxin. It is a secreted nontoxic protein with a molecular weight of 83 kDa and is the major component of the currently licensed human vaccine for anthrax. Due to limitations found in the existing vaccine formulation, it has been proposed that genetically modified PA may be more effective as a vaccine. The expression and the stability of two recombinant PA (rPA) variants, PA-SNKE-deltaFF-E308D and PA-N657A, were studied. These proteins were expressed in the nonsporogenic avirulent strain BH445. Initial results indicated that PA-SNKE-deltaFF-E308D, which lacks two proteolysis-sensitive sites, is more stable than PA-N657A. Process development was conducted to establish an efficient production and purification process for PA-SNKE-deltaFF-E308D. pH, media composition, growth strategy and protease inhibitors composition were analyzed. The production process chosen was based on batch growth of B. anthracis using tryptone and yeast extract as the only source of carbon, pH control at 7.5, and antifoam 289. Optimal harvest time was 14-18 h after inoculation, and EDTA (5 mM) was added upon harvest for proteolysis control. Recovery of the rPA was performed by expanded-bed adsorption (EBA) on a hydrophobic interaction chromatography (HIC) resin, eliminating the need for centrifugation, microfiltration and diafiltration. The EBA step was followed by ion exchange and gel filtration. rPA yields before and after purification were 130 and 90 mg/l, respectively. The purified rPA, without further treatment, treated with small amounts of formalin or adsorbed on alum, induced, high levels of IgG anti-PA with neutralization activities.


Asunto(s)
Antígenos Bacterianos , Bacillus anthracis/inmunología , Bacillus anthracis/metabolismo , Toxinas Bacterianas/biosíntesis , Toxinas Bacterianas/inmunología , Animales , Bacillus anthracis/química , Bacillus anthracis/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/aislamiento & purificación , Bioensayo , Cromatografía en Agarosa , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Femenino , Fermentación , Concentración de Iones de Hidrógeno , Ratones , Inhibidores de Proteasas/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación
4.
Vaccine ; 20(5-6): 789-96, 2001 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-11738742

RESUMEN

As a novel and safe vaccine strategy, the anthrax toxin-mediated antigen delivery system composed of lethal factor (LF) fusion protein and protective antigen (PA) has been studied to prime hepatitis C virus (HCV) core-specific cytotoxic T lymphocytes (CTLs) in vivo. The core epitope fused to LF (LF-core) together with PA induces a negligible core-specific CTL response in mice, whereas core-specific CTL are effectively primed in mice by injecting dendritic cells (DCs) treated in vitro with LF-core and PA. These findings imply that LF fusion protein plus PA in combination with dendritic cells may be useful for a novel T cell vaccine against HCV infection.


Asunto(s)
Antígenos Bacterianos , Células Dendríticas/inmunología , Hepacivirus/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Presentación de Antígeno , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Línea Celular , Femenino , Hepacivirus/genética , Inmunización , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/inmunología , Vacunas contra Hepatitis Viral/administración & dosificación , Vacunas contra Hepatitis Viral/genética , Vacunas contra Hepatitis Viral/inmunología
5.
Nature ; 414(6860): 229-33, 2001 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-11700563

RESUMEN

Lethal factor (LF) is a protein (relative molecular mass 90,000) that is critical in the pathogenesis of anthrax. It is a highly specific protease that cleaves members of the mitogen-activated protein kinase kinase (MAPKK) family near to their amino termini, leading to the inhibition of one or more signalling pathways. Here we describe the crystal structure of LF and its complex with the N terminus of MAPKK-2. LF comprises four domains: domain I binds the membrane-translocating component of anthrax toxin, the protective antigen (PA); domains II, III and IV together create a long deep groove that holds the 16-residue N-terminal tail of MAPKK-2 before cleavage. Domain II resembles the ADP-ribosylating toxin from Bacillus cereus, but the active site has been mutated and recruited to augment substrate recognition. Domain III is inserted into domain II, and seems to have arisen from a repeated duplication of a structural element of domain II. Domain IV is distantly related to the zinc metalloprotease family, and contains the catalytic centre; it also resembles domain I. The structure thus reveals a protein that has evolved through a process of gene duplication, mutation and fusion, into an enzyme with high and unusual specificity.


Asunto(s)
Antígenos Bacterianos , Bacillus anthracis/química , Toxinas Bacterianas/química , Secuencia de Aminoácidos , Toxinas Bacterianas/metabolismo , Cristalografía por Rayos X , MAP Quinasa Quinasa 2 , Sustancias Macromoleculares , Quinasas de Proteína Quinasa Activadas por Mitógenos/química , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Zinc/química
7.
Infect Immun ; 69(7): 4509-15, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11401993

RESUMEN

The ability of genetic vaccination to protect against a lethal challenge of anthrax toxin was evaluated. BALB/c mice were immunized via gene gun inoculation with eucaryotic expression vector plasmids encoding either a fragment of the protective antigen (PA) or a fragment of lethal factor (LF). Plasmid pCLF4 contains the N-terminal region (amino acids [aa] 10 to 254) of Bacillus anthracis LF cloned into the pCI expression plasmid. Plasmid pCPA contains a biologically active portion (aa 175 to 764) of B. anthracis PA cloned into the pCI expression vector. One-micrometer-diameter gold particles were coated with plasmid pCLF4 or pCPA or a 1:1 mixture of both and injected into mice via gene gun (1 microg of plasmid DNA/injection) three times at 2-week intervals. Sera were collected and analyzed for antibody titer as well as antibody isotype. Significantly, titers of antibody to both PA and LF from mice immunized with the combination of pCPA and pCLF4 were four to five times greater than titers from mice immunized with either gene alone. Two weeks following the third and final plasmid DNA boost, all mice were challenged with 5 50% lethal doses of lethal toxin (PA plus LF) injected intravenously into the tail vein. All mice immunized with pCLF4, pCPA, or the combination of both survived the challenge, whereas all unimmunized mice did not survive. These results demonstrate that DNA-based immunization alone can provide protection against a lethal toxin challenge and that DNA immunization against the LF antigen alone provides complete protection.


Asunto(s)
Vacunas contra el Carbunco/inmunología , Carbunco/prevención & control , Antígenos Bacterianos/genética , Bacillus anthracis/genética , Toxinas Bacterianas/genética , ADN Bacteriano/inmunología , Plásmidos/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , Inmunización Secundaria , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Vacunación
8.
Proc Natl Acad Sci U S A ; 98(7): 4089-94, 2001 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-11259649

RESUMEN

Lethal factor is a protease, one component of Bacillus anthracis exotoxin, which cleaves many of the mitogen-activated protein kinase kinases (MEKs). Given the importance of MEK signaling in tumorigenesis, we assessed the effects of anthrax lethal toxin (LeTx) on tumor cells. LeTx was very effective in inhibiting mitogen-activated protein kinase activation in V12 H-ras-transformed NIH 3T3 cells. In vitro, treatment of transformed cells with LeTx caused them to revert to a nontransformed morphology, and inhibited their abilities to form colonies in soft agar and to invade Matrigel without markedly affecting cell proliferation. In vivo, LeTx inhibited growth of ras-transformed cells implanted in athymic nude mice (in some cases causing tumor regression) at concentrations that caused no apparent animal toxicity. Unexpectedly, LeTx also greatly decreased tumor neovascularization. These results demonstrate that LeTx potently inhibits ras-mediated tumor growth and is a potential antitumor therapeutic.


Asunto(s)
Antígenos Bacterianos , Toxinas Bacterianas/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neovascularización Fisiológica/efectos de los fármacos , Células 3T3 , Animales , Toxinas Bacterianas/uso terapéutico , Pruebas de Carcinogenicidad , División Celular/efectos de los fármacos , Línea Celular Transformada , Modelos Animales de Enfermedad , Ratones , Ratones Desnudos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Invasividad Neoplásica , Trasplante de Neoplasias , Neoplasias Experimentales/prevención & control , Péptido Hidrolasas/efectos de los fármacos , Péptido Hidrolasas/metabolismo , Proteínas ras/fisiología
9.
J Biol Chem ; 276(21): 17976-84, 2001 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-11278833

RESUMEN

Urokinase plasminogen activator receptor (uPAR) binds pro-urokinase plasminogen activator (pro-uPA) and thereby localizes it near plasminogen, causing the generation of active uPA and plasmin on the cell surface. uPAR and uPA are overexpressed in a variety of human tumors and tumor cell lines, and expression of uPAR and uPA is highly correlated to tumor invasion and metastasis. To exploit these characteristics in the design of tumor cell-selective cytotoxins, we constructed mutated anthrax toxin-protective antigen (PrAg) proteins in which the furin cleavage site is replaced by sequences cleaved specifically by uPA. These uPA-targeted PrAg proteins were activated selectively on the surface of uPAR-expressing tumor cells in the presence of pro-uPA and plasminogen. The activated PrAg proteins caused internalization of a recombinant cytotoxin, FP59, consisting of anthrax toxin lethal factor residues 1-254 fused to the ADP-ribosylation domain of Pseudomonas exotoxin A, thereby killing the uPAR-expressing tumor cells. The activation and cytotoxicity of these uPA-targeted PrAg proteins were strictly dependent on the integrity of the tumor cell surface-associated plasminogen activation system. We also constructed a mutated PrAg protein that selectively killed tissue plasminogen activator-expressing cells. These mutated PrAg proteins may be useful as new therapeutic agents for cancer treatment.


Asunto(s)
Antígenos Bacterianos , Toxinas Bacterianas/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Toxinas Bacterianas/genética , Toxinas Bacterianas/uso terapéutico , Toxinas Bacterianas/toxicidad , Muerte Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Humanos , Ratones , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Plasminógeno/metabolismo , Proteínas Recombinantes/metabolismo , Células Tumorales Cultivadas
10.
Cancer Res ; 60(21): 6061-7, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11085528

RESUMEN

Matrix metalloproteinases (MMPs) are overexpressed in a variety of tumor tissues and cell lines, and their expression is highly correlated to tumor invasion and metastasis. To exploit these characteristics in the design of tumor cell-selective cytotoxins, we constructed two mutated anthrax toxin protective antigen (PA) proteins in which the furin protease cleavage site is replaced by sequences selectively cleaved by MMPs. These MMP-targeted PA proteins were activated rapidly and selectively on the surface of MMP-overexpressing tumor cells. The activated PA proteins caused internalization of a recombinant cytotoxin, FP59, consisting of anthrax toxin lethal factor residues 1-254 fused to the ADP-ribosylation domain of Pseudomonas exotoxin A. The toxicity of the mutated PA proteins for MMP-overexpressing cells was blocked by hydroxamate inhibitors of MMPs, including BB94, and by a tissue inhibitor of matrix metalloproteinases (TIMP-2). The mutated PA proteins killed MMP-overexpressing tumor cells while sparing nontumorigenic normal cells when these were grown together in a coculture model, indicating that PA activation occurred on the tumor cell surface and not in the supernatant. This method of achieving cell-type specificity is conceptually distinct from, and potentially synergistic with, the more common strategy of retargeting a protein toxin by fusion to a growth factor, cytokine, or antibody.


Asunto(s)
Antígenos Bacterianos , Toxinas Bacterianas/farmacocinética , Toxinas Bacterianas/toxicidad , Metaloproteinasas de la Matriz/metabolismo , Secuencia de Aminoácidos , Animales , Toxinas Bacterianas/genética , Sitios de Unión , Biotransformación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Células COS/enzimología , Chlorocebus aethiops , Técnicas de Cocultivo , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/enzimología , Humanos , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/biosíntesis , Metaloproteinasas de la Matriz Asociadas a la Membrana , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Metaloendopeptidasas/biosíntesis , Metaloendopeptidasas/metabolismo , Mutación , Células Tumorales Cultivadas , Células Vero/enzimología
11.
Protein Expr Purif ; 18(3): 293-302, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10733882

RESUMEN

Bacillus anthracis lethal factor (LF) is a 90-kDa zinc metalloprotease that plays an important role in the virulence of the organism. LF has previously been purified from Escherichia coli and Bacillus anthracis. The yields and purities of these preparations were inadequate for crystal structure determination. In this study, the genes encoding wild-type LF and a mutated, inactive LF (LF-E687C) were placed in an E. coli-Bacillus shuttle vector so that LF was produced with the protective antigen (PA) signal peptide at its N-terminus. The resulting vectors, pSJ115 and pSJ121, express wild-type and mutated LF fusion proteins, respectively. Expression of the LF genes is under the control of the PA promoter and, during secretion, the PA signal peptide is cleaved to release the 90-kDa LF proteins. The wild-type and mutated LF proteins were purified from the culture medium using three chromatographic steps (Phenyl-Sepharose, Q-Sepharose, and hydroxyapatite). The purified proteins were greater than 95% pure and yields (20-30 mg/L) were higher than those obtained in other expression systems (1-5 mg/L). These proteins have been crystallized and are being used to solve the crystal structure of LF. Their potential use in anthrax vaccines is also discussed.


Asunto(s)
Antígenos Bacterianos/aislamiento & purificación , Bacillus anthracis/química , Toxinas Bacterianas/aislamiento & purificación , Metaloendopeptidasas/aislamiento & purificación , Sustitución de Aminoácidos , Animales , Antígenos Bacterianos/química , Antígenos Bacterianos/metabolismo , Bacillus/genética , Bacillus anthracis/genética , Bacillus anthracis/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Cromatografía en Agarosa , Cromatografía por Intercambio Iónico , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Vectores Genéticos , Metaloendopeptidasas/química , Metaloendopeptidasas/metabolismo , Ratones , Regiones Promotoras Genéticas , Señales de Clasificación de Proteína/genética , Transformación Bacteriana
12.
J Biol Chem ; 274(38): 27274-80, 1999 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-10480947

RESUMEN

The alpha toxin produced by Clostridium septicum is a channel-forming protein that is an important contributor to the virulence of the organism. Chinese hamster ovary (CHO) cells are sensitive to low concentrations of the toxin, indicating that they contain toxin receptors. Using retroviral mutagenesis, a mutant CHO line (BAG15) was generated that is resistant to alpha toxin. FACS analysis showed that the mutant cells have lost the ability to bind the toxin, indicating that they lack an alpha toxin receptor. The mutant cells are also resistant to aerolysin, a channel-forming protein secreted by Aeromonas spp., which is structurally and functionally related to alpha toxin and which is known to bind to glycosylphosphatidylinositol (GPI)-anchored proteins, such as Thy-1. We obtained evidence that the BAG15 cells lack N-acetylglucosaminyl-phosphatidylinositol deacetylase-L, needed for the second step in GPI anchor biosynthesis. Several lymphocyte cell lines lacking GPI-anchored proteins were also shown to be less sensitive to alpha toxin. On the other hand, the sensitivity of CHO cells to alpha toxin was increased when the cells were transfected with the GPI-anchored folate receptor. We conclude that alpha toxin, like aerolysin, binds to GPI-anchored protein receptors. Evidence is also presented that the two toxins bind to different subsets of GPI-anchored proteins.


Asunto(s)
Clostridium/metabolismo , Glicosilfosfatidilinositoles/metabolismo , Receptores de Superficie Celular , Fosfolipasas de Tipo C/metabolismo , Animales , Toxinas Bacterianas/metabolismo , Células CHO , Proteínas Portadoras/metabolismo , Cricetinae , Receptores de Folato Anclados a GPI , Modelos Químicos , Mutagénesis , Fosfatidilinositol Diacilglicerol-Liasa , Proteínas Citotóxicas Formadoras de Poros , Retroviridae
13.
Infect Immun ; 67(6): 3055-60, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10338520

RESUMEN

Anthrax lethal toxin (LeTx), consisting of protective antigen (PA) and lethal factor (LF), rapidly kills primary mouse macrophages and macrophage-like cell lines such as RAW 264.7. LF is translocated by PA into the cytosol of target cells, where it acts as a metalloprotease to cleave mitogen-activated protein kinase kinase 1 (MEK1) and possibly other proteins. In this study, we show that proteasome inhibitors such as acetyl-Leu-Leu-norleucinal, MG132, and lactacystin efficiently block LeTx cytotoxicity, whereas other protease inhibitors do not. The inhibitor concentrations that block LF cytotoxicity are similar to those that inhibit the proteasome-dependent IkappaB-alpha degradation induced by lipopolysaccharide. The inhibitors did not interfere with the proteolytic cleavage of MEK1 in LeTx-treated cells, indicating that they do not directly block the proteolytic activity of LF. However, the proteasome inhibitors did prevent ATP depletion, an early effect of LeTx. No overall activation of the proteasome by LeTx was detected, as shown by the cleavage of fluorogenic substrates of the proteasome. All of these results suggest that the proteasome mediates a toxic process initiated by LF in the cell cytosol. This process probably involves degradation of unidentified molecules that are essential for macrophage homeostasis. Moreover, this proteasome-dependent process is an early step in LeTx intoxication, but it is downstream of the cleavage by LF of MEK1 or other putative substrates.


Asunto(s)
Antígenos Bacterianos , Toxinas Bacterianas/toxicidad , Cisteína Endopeptidasas/metabolismo , Macrófagos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos , Complejos Multienzimáticos/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Inhibidores de Cisteína Proteinasa/farmacología , Relación Dosis-Respuesta a Droga , MAP Quinasa Quinasa 1 , Macrófagos/citología , Ratones , Complejo de la Endopetidasa Proteasomal , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo
14.
Infect Immun ; 67(4): 1853-9, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10085027

RESUMEN

The protective antigen (PA) protein of anthrax toxin binds to a cellular receptor and is cleaved by cell surface furin to produce a 63-kDa fragment (PA63). The receptor-bound PA63 oligomerizes to a heptamer and acts to translocate the catalytic moieties of the toxin, lethal factor (LF) and edema factor (EF), from endosomes to the cytosol. In this report, we used nondenaturing gel electrophoresis to show that each PA63 subunit in the heptamer can bind one LF molecule. Studies using PA immobilized on a plastic surface showed that monomeric PA63 is also able to bind LF. The internalization of PA and LF by cells was studied with radiolabeled and biotinylated proteins. Uptake was relatively slow, with a half-time of 30 min. The number of moles of LF internalized was nearly equal to the number of moles of PA subunit internalized. The essential role of PA oligomerization in LF translocation was shown with PA protein cleaved at residues 313-314. The oligomers formed by these proteins during uptake into cells were not as stable when subjected to heat and detergent as were those formed by native PA. The results show that the structure of the toxin proteins and the kinetics of proteolytic activation, LF binding, and internalization are balanced in a way that allows each PA63 subunit to internalize an LF molecule. This set of proteins has evolved to achieve highly efficient internalization and membrane translocation of the catalytic components, LF and EF.


Asunto(s)
Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Endocitosis/inmunología , Animales , Transporte Biológico , Línea Celular , Mamíferos , Oligopéptidos/metabolismo , Receptores de Péptidos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Soluciones
15.
Infect Immun ; 67(4): 1860-5, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10085028

RESUMEN

Anthrax toxin from Bacillus anthracis is a three-component toxin consisting of lethal factor (LF), edema factor (EF), and protective antigen (PA). LF and EF are the catalytic components of the toxin, whereas PA is the receptor-binding component. To identify residues of PA that are involved in interaction with the cellular receptor, two solvent-exposed loops of domain 4 of PA (amino acids [aa] 679 to 693 and 704 to 723) were mutagenized, and the altered proteins purified and tested for toxicity in the presence of LF. In addition to the intended substitutions, novel mutations were introduced by errors that occurred during PCR. Substitutions within the large loop (aa 704 to 723) had no effect on PA activity. A mutated protein, LST-35, with three substitutions in the small loop (aa 679 to 693), bound weakly to the receptor and was nontoxic. A mutated protein, LST-8, with changes in three separate regions did not bind to receptor and was nontoxic. Toxicity was greatly decreased by truncation of the C-terminal 3 to 5 aa, but not by their substitution with nonnative residues or the extension of the terminus with nonnative sequences. Comparison of the 28 mutant proteins described here showed that the large loop (aa 704 to 722) is not involved in receptor binding, whereas residues in and near the small loop (aa 679 to 693) play an important role in receptor interaction. Other regions of domain 4, in particular residues at the extreme C terminus, appear to play a role in stabilizing a conformation needed for receptor-binding activity.


Asunto(s)
Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Receptores de Péptidos/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Bacillus anthracis/química , Bacillus anthracis/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Citotoxicidad Inmunológica , Ratones , Datos de Secuencia Molecular , Mutagénesis , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
16.
Vaccine ; 17(4): 340-4, 1999 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-9987172

RESUMEN

A DNA vaccine encoding the immunogenic and biologically active portion of anthrax protective antigen (PA) was constructed. Spleen cells from BALB/c mice immunized intramuscularly with this vaccine were stimulated to secrete IFN gamma and IL-4 when exposed to PA in vitro. Immunized mice also mounted a humoral immune response dominated by IgG1 anti-PA antibody production, the subclass previously shown to confer protection against anthrax toxin. A 1:100 dilution of serum from these animals protected cells in vitro against cytotoxic concentrations of PA. Moreover, 7/8 mice immunized three times with the PA DNA vaccine were protected against lethal challenge with a combination of anthrax protective antigen plus lethal factor.


Asunto(s)
Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , ADN/genética , Plásmidos/genética , Vacunación , Animales , Anticuerpos Antibacterianos/biosíntesis , Antígenos Bacterianos/genética , Femenino , Código Genético , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Dosificación Letal Mediana , Ratones , Ratones Endogámicos BALB C
17.
J Biol Chem ; 273(49): 32656-61, 1998 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-9830006

RESUMEN

Aerolysin is secreted as an inactive dimeric precursor by the bacterium Aeromonas hydrophila. Proteolytic cleavage within a mobile loop near the C terminus of the protoxin is required for oligomerization and channel formation. This loop contains the sequence KVRRAR432, which should be recognized by mammalian proprotein convertases such as furin, PACE4, and PC5/6A. Here we show that these three proteases cleave proaerolysin after Arg-432 in vitro, yielding active toxin. We also investigated the potential role of these enzymes in the in vivo activation of the protoxin. We found that Chinese hamster ovary cells were able to convert the protoxin to aerolysin in the absence of exogenous proteases and that activation did not require internalization of the toxin. The furin inhibitor alpha1-antitrypsin Portland reduced the rate of proaerolysin activation in vivo, and proaerolysin processing was even further reduced in furin-deficient FD11 Chinese hamster ovary cells. The cells were also less sensitive to proaerolysin than wild type cells; however, transient transfection of FD11 cells with the cDNA encoding furin conferred normal sensitivity to the protoxin. Together these findings argue that furin catalyzes the cell-surface activation of proaerolysin in vivo.


Asunto(s)
Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Subtilisinas/farmacología , Animales , Células CHO , Cricetinae , Furina , Calor , Proteínas Citotóxicas Formadoras de Poros , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/farmacología , Vacuolas , alfa 1-Antitripsina/farmacología
18.
Infect Immun ; 66(7): 3447-8, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9632621

RESUMEN

Protective antigen (PA) of anthrax toxin is the major component of human anthrax vaccine. Currently available human vaccines in the United States and Europe consist of alum-precipitated supernatant material from cultures of toxigenic, nonencapsulated strains of Bacillus anthracis. Immunization with these vaccines requires several boosters and occasionally causes local pain and edema. We previously described the biological activity of a nontoxic mutant of PA expressed in Bacillus subtilis. In the present study, we evaluated the efficacy of the purified mutant PA protein alone or in combination with the lethal factor and edema factor components of anthrax toxin to protect against anthrax. Both mutant and native PA preparations elicited high anti-PA titers in Hartley guinea pigs. Mutant PA alone and in combination with lethal factor and edema factor completely protected the guinea pigs from B. anthracis spore challenge. The results suggest that the mutant PA protein may be used to develop an effective recombinant vaccine against anthrax.


Asunto(s)
Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Vacunas Bacterianas/inmunología , Vacunas Sintéticas/inmunología , Animales , Carbunco/prevención & control , Femenino , Cobayas , Inmunización , Proteínas Recombinantes/inmunología
19.
Science ; 280(5364): 734-7, 1998 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-9563949

RESUMEN

Anthrax lethal toxin, produced by the bacterium Bacillus anthracis, is the major cause of death in animals infected with anthrax. One component of this toxin, lethal factor (LF), is suspected to be a metalloprotease, but no physiological substrates have been identified. Here it is shown that LF is a protease that cleaves the amino terminus of mitogen-activated protein kinase kinases 1 and 2 (MAPKK1 and MAPKK2) and that this cleavage inactivates MAPKK1 and inhibits the MAPK signal transduction pathway. The identification of a cleavage site for LF may facilitate the development of LF inhibitors.


Asunto(s)
Antígenos Bacterianos , Bacillus anthracis , Toxinas Bacterianas/toxicidad , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Animales , Bacillus anthracis/enzimología , Toxinas Bacterianas/metabolismo , Sitios de Unión , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular Transformada , Activación Enzimática , Inhibidores Enzimáticos/toxicidad , Humanos , MAP Quinasa Quinasa 1 , MAP Quinasa Quinasa 2 , Metaloendopeptidasas/metabolismo , Metaloendopeptidasas/toxicidad , Ratones , Proteína Básica de Mielina/metabolismo , Oocitos/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia , Transducción de Señal , Xenopus laevis
20.
Mol Med ; 4(2): 87-95, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9508786

RESUMEN

BACKGROUND: Anthrax toxin, secreted by Bacillus anthracis, consists of protective antigen (PA) and either lethal factor (LF) or edema factor (EF). PA, the receptor-binding component of the toxin, translocates LF or EF into the cytosol, where the latter proteins exert their toxic effects. We hypothesized that anthrax toxin fusion proteins could be used to kill virus-infected cells and tumor cells, if PA could be redirected to unique receptors found only on these cells. MATERIALS AND METHODS: To test this hypothesis in a model system, amino acids 410-419 of the human p62(c-myc) epitope were fused to the C-terminus of PA to redirect PA to the c-Myc-specific hybridoma cell line 9E10. RESULTS: The PA-c-Myc fusion protein killed both mouse macrophages and 9E10 hybridoma cells when administered with LF or an LF fusion protein (FP59), respectively. Similar results were obtained with PA, which suggests that PA-c-Myc used the endogenous PA receptor to enter the cells. By blocking the endogenous PA receptors on 9E10 cells with the competitive inhibitor PA SNKEDeltaFF, the PA-c-Myc was directed to an alternate receptor, i.e., the anti-c-Myc antibodies presented on the cell surface. The c-Myc IgG were proven to act as receptors because the addition of a synthetic peptide containing the c-Myc epitope along with PA SNKEDeltaFF further reduced the toxicity of PA-c-Myc + FP59. CONCLUSION: This study shows that PA can be redirected to alternate receptors by adding novel epitopes to the C-terminus of PA, enabling the creation of cell-directed toxins for therapeutic purposes.


Asunto(s)
Carbunco/inmunología , Antígenos Bacterianos , Toxinas Bacterianas/inmunología , Proteínas Proto-Oncogénicas c-myc/inmunología , Animales , Bacillus anthracis , Toxinas Bacterianas/genética , Células Cultivadas , Citotoxicidad Inmunológica , Epítopos/inmunología , Humanos , Hibridomas/inmunología , Hibridomas/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Recombinantes de Fusión/inmunología , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA