Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34417313

RESUMO

When displayed on erythrocytes, peptides and proteins can drive antigen-specific immune tolerance. Here, we investigated a straightforward approach based on erythrocyte binding to promote antigen-specific tolerance to both peptides and proteins. We first identified a robust erythrocyte-binding ligand. A pool of one million fully d-chiral peptides was injected into mice, blood cells were isolated, and ligands enriched on these cells were identified using nano-liquid chromatography-tandem mass spectrometry. One round of selection yielded a murine erythrocyte-binding ligand with an 80 nM apparent dissociation constant, Kd We modified an 83-kDa bacterial protein and a peptide antigen derived from ovalbumin (OVA) with the identified erythrocyte-binding ligand. An administration of the engineered bacterial protein led to decreased protein-specific antibodies in mice. Similarly, mice given the engineered OVA-derived peptide had decreased inflammatory anti-OVA CD8+ T cell responses. These findings suggest that our tolerance-induction strategy is applicable to both peptide and protein antigens and that our in vivo selection strategy can be used for de novo discovery of robust erythrocyte-binding ligands.


Assuntos
Antígenos/genética , Antígenos/metabolismo , Eritrócitos/metabolismo , Engenharia de Proteínas/métodos , Animais , Antígenos/química , Linhagem Celular , Bases de Dados Factuais , Feminino , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Ligação Proteica
2.
Nature ; 521(7553): 545-9, 2015 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-25778700

RESUMO

Anthrax toxin, comprising protective antigen, lethal factor, and oedema factor, is the major virulence factor of Bacillus anthracis, an agent that causes high mortality in humans and animals. Protective antigen forms oligomeric prepores that undergo conversion to membrane-spanning pores by endosomal acidification, and these pores translocate the enzymes lethal factor and oedema factor into the cytosol of target cells. Protective antigen is not only a vaccine component and therapeutic target for anthrax infections but also an excellent model system for understanding the mechanism of protein translocation. On the basis of biochemical and electrophysiological results, researchers have proposed that a phi (Φ)-clamp composed of phenylalanine (Phe)427 residues of protective antigen catalyses protein translocation via a charge-state-dependent Brownian ratchet. Although atomic structures of protective antigen prepores are available, how protective antigen senses low pH, converts to active pore, and translocates lethal factor and oedema factor are not well defined without an atomic model of its pore. Here, by cryo-electron microscopy with direct electron counting, we determine the protective antigen pore structure at 2.9-Å resolution. The structure reveals the long-sought-after catalytic Φ-clamp and the membrane-spanning translocation channel, and supports the Brownian ratchet model for protein translocation. Comparisons of four structures reveal conformational changes in prepore to pore conversion that support a multi-step mechanism by which low pH is sensed and the membrane-spanning channel is formed.


Assuntos
Antígenos de Bactérias/metabolismo , Antígenos de Bactérias/ultraestrutura , Bacillus anthracis/química , Bacillus anthracis/ultraestrutura , Toxinas Bacterianas/metabolismo , Microscopia Crioeletrônica , Antígenos de Bactérias/química , Toxinas Bacterianas/química , Biocatálise , Concentração de Íons de Hidrogênio , Canais Iônicos/química , Canais Iônicos/metabolismo , Canais Iônicos/ultraestrutura , Modelos Moleculares , Fenilalanina/metabolismo , Conformação Proteica , Transporte Proteico , Relação Estrutura-Atividade
3.
Biochemistry ; 53(44): 6934-40, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25317832

RESUMO

The protective antigen (PA) moiety of anthrax toxin forms oligomeric pores in the endosomal membrane, which translocate the effector proteins of the toxin to the cytosol. Effector proteins bind to oligomeric PA via their respective N-terminal domains and undergo N- to C-terminal translocation through the pore. Earlier we reported that a tract of basic amino acids fused to the N-terminus of an unrelated effector protein (the catalytic domain diphtheria toxin, DTA) potentiated that protein to undergo weak PA-dependent translocation. In this study, we varied the location of the tract (N-terminal or C-terminal) and the length of a poly-Lys tract fused to DTA and examined the effects of these variations on PA-dependent translocation into cells and across planar bilayers in vitro. Entry into cells was most efficient with ∼12 Lys residues (K12) fused to the N-terminus but also occurred, albeit 10-100-fold less efficiently, with a C-terminal tract of the same length. Similarly, K12 tracts at either terminus occluded PA pores in planar bilayers, and occlusion was more efficient with the N-terminal tag. We used biotin-labeled K12 constructs in conjunction with streptavidin to show that a biotinyl-K12 tag at either terminus is transiently exposed to the trans compartment of planar bilayers at 20 mV; this partial translocation in vitro was more efficient with an N-terminal tag than a C-terminal tag. Significantly, our studies with polycationic tracts fused to the N- and C-termini of DTA suggest that PA-mediated translocation can occur not only in the N to C direction but also in the C to N direction.


Assuntos
Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Toxina Diftérica/metabolismo , Polilisina/metabolismo , Animais , Antígenos de Bactérias/química , Toxinas Bacterianas/química , Células CHO , Domínio Catalítico , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Toxina Diftérica/química , Humanos , Cinética , Bicamadas Lipídicas/química , Polilisina/química , Transporte Proteico
4.
Proc Natl Acad Sci U S A ; 108(5): 1868-73, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21262847

RESUMO

The protective antigen (PA) moiety of anthrax toxin forms oligomeric pores that translocate the enzymatic moieties of the toxin--lethal factor (LF) and edema factor (EF)--across the endosomal membrane of mammalian cells. Here we describe site-directed spin-labeling studies that identify interactions of LF with the prepore and pore conformations of PA. Our results reveal a direct interaction between the extreme N terminus of LF (residues 2-5) and the Φ-clamp, a structure within the lumen of the pore that catalyzes translocation. Also, consistent with a recent crystallographic model, we find that, upon binding of the translocation substrate to PA, LF helix α1 separates from helices α2 and α3 and binds in the α-clamp of PA. These interactions, together with the binding of the globular part of the N-terminal domain of LF to domain 1' of PA, indicate that LF interacts with the PA pore at three distinct sites. Our findings elucidate the state from which translocation of LF and EF proceeds through the PA pore.


Assuntos
Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Marcadores de Spin , Antígenos de Bactérias/química , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/química , Toxinas Bacterianas/imunologia , Cristalografia por Raios X , Bicamadas Lipídicas , Modelos Moleculares
5.
Proc Natl Acad Sci U S A ; 108(40): 16577-81, 2011 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-21949363

RESUMO

Many bacterial toxins form proteinaceous pores that facilitate the translocation of soluble effector proteins across cellular membranes. With anthrax toxin this process may be monitored in real time by electrophysiology, where fluctuations in ionic current through these pores inserted in model membranes are used to infer the translocation of individual protein molecules. However, detecting the minute quantities of translocated proteins has been a challenge. Here, we describe use of the droplet-interface bilayer system to follow the movement of proteins across a model membrane separating two submicroliter aqueous droplets. We report the capture and subsequent direct detection of as few as 100 protein molecules that have translocated through anthrax toxin pores. The droplet-interface bilayer system offers new avenues of approach to the study of protein translocation.


Assuntos
Antígenos de Bactérias/química , Toxinas Bacterianas/química , Proteínas de Transporte/metabolismo , Eletrofisiologia/métodos , Bicamadas Lipídicas/metabolismo , Proteínas Citotóxicas Formadoras de Poros/química , Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Transporte Proteico/fisiologia
6.
Biochemistry ; 52(37): 6335-47, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-23964683

RESUMO

Domain 2 of the anthrax protective antigen (PA) prepore heptamer unfolds and refolds during endosome acidification to generate an extended 100 Å ß barrel pore that inserts into the endosomal membrane. The PA pore facilitates the pH-dependent unfolding and translocation of bound toxin enzymic components, lethal factor (LF) and/or edema factor, from the endosome to the cytoplasm. We constructed immobilized complexes of the prepore with the PA-binding domain of LF (LFN) to monitor the real-time prepore to pore kinetic transition using surface plasmon resonance and biolayer interferometry (BLI). The kinetics of this transition increased as the solution pH was decreased from 7.5 to 5.0, mirroring acidification of the endosome. Once it had undergone the transition, the LFN-PA pore complex was removed from the BLI biosensor tip and deposited onto electron microscopy grids, where PA pore formation was confirmed by negative stain electron microscopy. When the soluble receptor domain (ANTRX2/CMG2) binds the immobilized PA prepore, the transition to the pore state was observed only after the pH was lowered to early (pH 5.5) or late (pH 5.0) endosomal pH conditions. Once the pore formed, the soluble receptor readily dissociated from the PA pore. Separate binding experiments with immobilized PA pores and the soluble receptor indicate that the receptor has a weakened propensity to bind to the transitioned pore. This immobilized anthrax toxin platform can be used to identify or validate potential antimicrobial lead compounds capable of regulating and/or inhibiting anthrax toxin complex formation or pore transitions.


Assuntos
Antígenos de Bactérias/química , Toxinas Bacterianas/química , Endossomos/metabolismo , Proteínas Imobilizadas/química , Membranas Intracelulares/metabolismo , Antígenos de Bactérias/ultraestrutura , Técnicas Biossensoriais , Humanos , Concentração de Íons de Hidrogênio , Interferometria , Cinética , Micelas , Microscopia Eletrônica , Dobramento de Proteína , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Receptores de Peptídeos/metabolismo , Ressonância de Plasmônio de Superfície
7.
Cell Microbiol ; 13(3): 359-73, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20946244

RESUMO

Bacillus anthracis lethal toxin consists of the protective antigen (PA) and the metalloprotease lethal factor (LF). During cellular uptake PA forms pores in membranes of endosomes, and unfolded LF translocates through the pores into the cytosol. We have investigated whether host cell chaperones facilitate translocation of LF and the fusion protein LF(N)DTA. LF(N) mediates uptake of LF(N)DTA into the cytosol, where DTA, the catalytic domain of diphtheria toxin, ADP-ribosylates elongation factor-2, allowing for detection of small amounts of translocated LF(N)DTA. Cyclosporin A, which inhibits peptidyl-prolyl cis/trans isomerase activity of cyclophilins, and radicicol, which inhibits Hsp90 activity, prevented uptake of LF(N)DTA into the cytosol of CHO-K1 cells and protected cells from intoxication by LF(N)DTA/PA. Both inhibitors, as well as an antibody against cyclophilin A blocked the release of active LF(N)DTA from endosomal vesicles into the cytosol in vitro. In contrast, the inhibitors did not inhibit cellular uptake of LF. In vitro, cyclophilin A and Hsp90 bound to LF(N)DTA and DTA but not to LF, implying that DTA determines this interaction. In conclusion, cyclophilin A and Hsp90 facilitate translocation of LF(N)DTA, but not of LF, across endosomal membranes, and thus they function selectively in promoting translocation of certain proteins, but not of others.


Assuntos
Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Ciclofilina A/metabolismo , Ciclosporina/farmacologia , Proteínas de Choque Térmico HSP90/metabolismo , Metaloproteases/metabolismo , Animais , Antígenos de Bactérias/genética , Toxinas Bacterianas/genética , Transporte Biológico , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Citosol/metabolismo , Toxina Diftérica/genética , Toxina Diftérica/metabolismo , Endossomos/metabolismo , Humanos , Macrolídeos/farmacologia , Fator 2 de Elongação de Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo
8.
Nat Neurosci ; 25(2): 168-179, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34931070

RESUMO

Bacterial products can act on neurons to alter signaling and function. In the present study, we found that dorsal root ganglion (DRG) sensory neurons are enriched for ANTXR2, the high-affinity receptor for anthrax toxins. Anthrax toxins are composed of protective antigen (PA), which binds to ANTXR2, and the protein cargoes edema factor (EF) and lethal factor (LF). Intrathecal administration of edema toxin (ET (PA + EF)) targeted DRG neurons and induced analgesia in mice. ET inhibited mechanical and thermal sensation, and pain caused by formalin, carrageenan or nerve injury. Analgesia depended on ANTXR2 expressed by Nav1.8+ or Advillin+ neurons. ET modulated protein kinase A signaling in mouse sensory and human induced pluripotent stem cell-derived sensory neurons, and attenuated spinal cord neurotransmission. We further engineered anthrax toxins to introduce exogenous protein cargoes, including botulinum toxin, into DRG neurons to silence pain. Our study highlights interactions between a bacterial toxin and nociceptors, which may lead to the development of new pain therapeutics.


Assuntos
Antraz , Bacillus anthracis , Toxinas Bacterianas , Células-Tronco Pluripotentes Induzidas , Animais , Antraz/microbiologia , Antraz/terapia , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Gânglios Espinais/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Nociceptores/metabolismo , Dor , Receptores de Peptídeos/metabolismo
9.
Biophys J ; 101(10): L41-3, 2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22098755

RESUMO

The translocation (T) domain plays a key role in the action of diphtheria toxin and is responsible for transferring the N-terminus-attached catalytic domain across the endosomal membrane into the cytosol in response to acidification. The T-domain undergoes a series of pH-triggered conformational changes that take place in solution and on the membrane interface, and ultimately result in transbilayer insertion and N-terminus translocation. Structure-function studies along this pathway have been hindered because the protein population occupies multiple conformations at the same time. Here we report that replacement of the three C-terminal histidine residues, H322, H323, and H372, in triple-R or triple-Q mutants prevents effective translocation of the N-terminus. Introduction of these mutations in the full-length toxin results in decrease of its potency. In the context of isolated T-domain, these mutations cause loss of characteristic conductance in planar bilayers. Surprisingly, these mutations do not affect general folding in solution, protein interaction with the membranes, insertion of the consensus transmembrane helical hairpin TH8-9, or the ability of the T-domain to destabilize vesicles to cause leakage of fluorescent markers. Thus, the C-terminal histidine residues are critical for the transition from the inserted intermediate state to the open-channel state in the insertion/translocation pathway of the T-domain.


Assuntos
Toxina Diftérica/química , Histidina/química , Bicamadas Lipídicas/química , Mutação/genética , Cristalografia por Raios X , Fenômenos Eletrofisiológicos , Modelos Moleculares , Proteínas Mutantes/química , Estrutura Terciária de Proteína , Transporte Proteico , Relação Estrutura-Atividade
10.
Biochemistry ; 50(17): 3512-6, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21425869

RESUMO

Electrophysiological studies of wild-type and mutated forms of anthrax protective antigen (PA) suggest that the Phe clamp, a structure formed by the Phe427 residues within the lumen of the oligomeric PA pore, binds the unstructured N-terminus of the lethal factor and the edema factor during initiation of translocation. We now show by electrophysiological measurements and gel shift assays that a single Cys introduced into the Phe clamp can form a disulfide bond with a Cys placed at the N-terminus of the isolated N-terminal domain of LF. These results demonstrate direct contact of these Cys residues, supporting a model in which the interaction of the unstructured N-terminus of the translocated moieties with the Phe clamp initiates N- to C-terminal threading of these moieties through the pore.


Assuntos
Antígenos de Bactérias/química , Bacillus anthracis , Toxinas Bacterianas/química , Reagentes de Ligações Cruzadas/química , Cisteína/química , Fenilalanina/química , Antígenos de Bactérias/genética , Toxinas Bacterianas/genética , Dissulfetos/química , Ensaio de Desvio de Mobilidade Eletroforética , Bicamadas Lipídicas , Potenciais da Membrana , Mutação , Multimerização Proteica , Transporte Proteico
11.
J Biol Chem ; 285(11): 8130-7, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20061382

RESUMO

Multimeric pores formed in the endosomal membrane by the Protective Antigen moiety of anthrax toxin translocate the enzymatic moieties of the toxin to the cytosolic compartment of mammalian cells. There is evidence that the side chains of the Phe(427) residues come into close proximity with one another in the lumen of the pore and form a structure, termed the Phe clamp, that catalyzes the translocation process. In this report we describe the effects of replacing Phe(427) in a single subunit of the predominantly heptameric pore with a basic or an acidic amino acid. Incorporating any charged residue at this position inhibited cytotoxicity >or=1,000-fold in our standard assay and caused strong inhibition of translocation in a planar phospholipid bilayer system. His and Glu were the most strongly inhibitory residues, ablating both cytotoxicity and translocation. Basic residues at position 427 prevented the Phe clamp from interacting with a translocation substrate to form a seal against the passage of ions and accelerated dissociation of the substrate from the pore. Acidic residues, in contrast, allowed the seal to form and the substrate to remain firmly bound, but blocked its passage, perhaps via electrostatic interactions with the positively charged N-terminal segment. Our findings are discussed in relation to the role of the Phe clamp in a Brownian ratchet model of translocation.


Assuntos
Antígenos de Bactérias , Toxinas Bacterianas , Endossomos/metabolismo , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Fenilalanina/genética , Substituição de Aminoácidos , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Glutamina/química , Histamina/química , Concentração de Íons de Hidrogênio , Ativação do Canal Iônico , Potenciais da Membrana , Mutagênese , Estrutura Quaternária de Proteína , Transporte Proteico
12.
Cell Microbiol ; 12(10): 1435-45, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20438574

RESUMO

To investigate the cell entry and intracellular trafficking of anthrax oedema factor (EF) and lethal factor (LF), they were C-terminally fused to the enhanced green fluorescent protein (EGFP) and monomeric Cherry (mCherry) fluorescent proteins. Both chimeras bound to the surface of BHK cells treated with protective antigen (PA) in a patchy mode. Binding was followed by rapid internalization, and the two anthrax factors were found to traffic along the same endocytic route and with identical kinetics, indicating that their intracellular path is essentially dictated by PA. Colocalization studies indicated that anthrax toxins enter caveolin-1 containing compartments and then endosomes marked by phoshatidylinositol 3-phoshate and Rab5, but not by early endosome antigen 1 and transferrin. After 40 min, both EF and LF chimeras were observed to localize within late compartments. Eventually, LF and EF appeared in the cytosol with a time-course consistent with translocation from late endosomes. Only the EGFP derivatives reached the cytosol because they are translocated by the PA channel, while the mCherry derivatives are not. This difference is attributed to a higher resistance of mCherry to unfolding. After translocation, LF disperses in the cytosol, while EF localizes on the cytosolic face of late endosomes.


Assuntos
Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Animais , Células Cultivadas , Cricetinae , Citosol/química , Endossomos/química , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Ligação Proteica , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Coloração e Rotulagem/métodos , Fatores de Tempo , Proteína Vermelha Fluorescente
13.
Proc Natl Acad Sci U S A ; 105(11): 4346-51, 2008 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-18334631

RESUMO

The protective antigen (PA) moiety of anthrax toxin forms a heptameric pore in endosomal membranes of mammalian cells and translocates the enzymatic moieties of the toxin to the cytosol of these cells. Phenylalanine-427 (F427), a solvent-exposed residue in the lumen of the pore, was identified earlier as being crucial for the transport function of PA. The seven F427 residues were shown in electrophysiological studies to form a clamp that catalyzes protein translocation through the pore. Here, we demonstrate by a variety of tests that certain F427 mutations also profoundly inhibit the conformational transition of the heptameric PA prepore to the pore and thereby block pore formation in membranes. Lysine, arginine, aspartic acid, or glycine at position 427 strongly inhibited this acidic pH-induced conformational transition, whereas histidine, serine, and threonine had virtually no effect on this step, but inhibited translocation instead. Thus, it is possible to inhibit pore formation or translocation selectively, depending on the choice of the side chain at position 427; and the net inhibition of the PA transport function by any given F427 mutation is the product of its effects on both steps. Mutations inhibiting either or both steps elicited a strong dominant-negative phenotype. These findings demonstrate the dual functions of F427 and underline its central role in transporting the enzymatic moieties of anthrax toxin across membranes.


Assuntos
Antígenos de Bactérias/metabolismo , Bacillus anthracis/citologia , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Animais , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Antígenos de Bactérias/toxicidade , Bacillus anthracis/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/toxicidade , Células CHO , Cátions/química , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Lipossomos/metabolismo , Modelos Moleculares , Mutação/genética , Fenótipo , Fenilalanina/genética , Fenilalanina/metabolismo , Transporte Proteico , Esporos Bacterianos/química , Esporos Bacterianos/citologia , Esporos Bacterianos/metabolismo
14.
Proc Natl Acad Sci U S A ; 105(7): 2439-44, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18268319

RESUMO

Vascular dysfunction has been reported in human cases of anthrax, in mammalian models of Bacillus anthracis, and in animals injected with anthrax toxin proteins. To examine anthrax lethal toxin effects on intact blood vessels, we developed a zebrafish model that permits in vivo imaging and evaluation of vasculature and cardiovascular function. Vascular defects monitored in hundreds of embryos enabled us to define four stages of phenotypic progression leading to circulatory dysfunction. We demonstrated increased endothelial permeability as an early consequence of toxin action by tracking the extravasation of fluorescent microspheres in toxin-injected embryos. Lethal toxin did not induce a significant amount of cell death in embryonic tissues or blood vessels, as shown by staining with acridine orange, and endothelial cells in lethal toxin-injected embryos continued to divide at the normal rate. Vascular permeability is strongly affected by the VEGF/vascular permeability factor (VPF) signaling pathway, and we were able to attenuate anthrax lethal toxin effects with chemical inhibitors of VEGFR function. Our study demonstrates the importance of vascular permeability in anthrax lethal toxin action and the need for further investigation of the cardiovascular component of human anthrax disease.


Assuntos
Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Permeabilidade da Membrana Celular , Neovascularização Fisiológica/efeitos dos fármacos , Peixe-Zebra , Animais , Morte Celular/efeitos dos fármacos , Embrião não Mamífero/irrigação sanguínea , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/embriologia , Dados de Sequência Molecular , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/embriologia
15.
Proc Natl Acad Sci U S A ; 105(16): 6150-5, 2008 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-18427110

RESUMO

Bacillus anthracis, the etiologic agent of anthrax, avoids immune surveillance and commandeers host macrophages as a vehicle for lymphatic spreading. Here, we show that B. anthracis edema toxin (ET), via its adenylyl cyclase activity, dramatically increases the motility of infected macrophages and the expression of vascular endothelial growth factor. The transcription factor CREB and the syndecan-1 gene, a CREB target, play crucial roles in ET-induced macrophage migration. These molecular and cellular responses occur in macrophages engaged in antiinflammatory G protein-coupled receptor activation, thus illustrating a common signaling circuitry controlling resolution of inflammation and host cell hijacking by B. anthracis.


Assuntos
Antraz/imunologia , Bacillus anthracis/enzimologia , Movimento Celular/genética , AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Macrófagos/imunologia , Adenilil Ciclases/metabolismo , Animais , Antraz/genética , Antraz/microbiologia , Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Macrófagos/microbiologia , Camundongos , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Sindecana-1/genética , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética
16.
Nat Struct Mol Biol ; 11(1): 60-6, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14718924

RESUMO

Recent events have created an urgent need for new therapeutic strategies to treat anthrax. We have applied a mixture-based peptide library approach to rapidly determine the optimal peptide substrate for the anthrax lethal factor (LF), a metalloproteinase with an important role in the pathogenesis of the disease. Using this approach we have identified peptide analogs that inhibit the enzyme in vitro and that protect cultured macrophages from LF-mediated cytolysis. The crystal structures of LF bound to an optimized peptide substrate and to peptide-based inhibitors provide a rationale for the observed selectivity and may be exploited in the design of future generations of LF inhibitors.


Assuntos
Antígenos de Bactérias , Bacillus anthracis/patogenicidade , Toxinas Bacterianas/antagonistas & inibidores , Toxinas Bacterianas/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Antraz/tratamento farmacológico , Bacillus anthracis/enzimologia , Bacillus anthracis/genética , Toxinas Bacterianas/genética , Humanos , Técnicas In Vitro , Cinética , Substâncias Macromoleculares , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Biblioteca de Peptídeos , Especificidade por Substrato
17.
Cancer Res ; 67(20): 9980-5, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17942931

RESUMO

Bacillus anthracis protective antigen (PA), the B subunit of the binary anthrax toxin, binds to the cellular receptors capillary morphogenesis gene 2 protein and tumor endothelial marker 8 with high affinity. Both receptors are expressed on endothelial cells during angiogenesis. We sought to determine whether one could inhibit angiogenesis by interfering with the binding of these receptors to their endogenous ligands. Here, we show that wild-type PA inhibits both vascular endothelial growth factor-induced and basic fibroblast growth factor-induced angiogenesis at moderate but statistically significant levels. Structure-activity studies identified a PA mutant that exhibited markedly enhanced inhibition of angiogenesis and also inhibited tumor growth in vivo. This mutant, PASSSR, is unable to undergo normal cellular processing and, thus, remains bound to the surface receptor. Further mutation of PASSSR so that it does not bind to these cell surface receptors abolished its ability to inhibit angiogenesis. We conclude that high-affinity anthrax toxin receptor (ATR) ligands, such as PA and PASSSR, are angiogenesis inhibitors and that ATRs are useful targets for antiangiogenic therapy. These results also suggest that endothelial cell-binding proteins from additional pathogens may inhibit angiogenesis and raise the question of the role of such inhibition in pathogenesis.


Assuntos
Inibidores da Angiogênese/farmacologia , Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Processos de Crescimento Celular/efeitos dos fármacos , Córnea/irrigação sanguínea , Neovascularização da Córnea/induzido quimicamente , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Humanos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos , Mutação , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/metabolismo , Receptores de Peptídeos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
18.
Biochemistry ; 47(32): 8406-13, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18636745

RESUMO

The actin-ADP-ribosylating Clostridium botulinum C2 toxin consists of the enzymatic component C2I and the binding component C2II. C2II forms heptameric channels involved in translocation of the enzymatic component into the target cell. On the basis of the heptameric toxin channel, we studied functional consequences of mutagenesis of amino acid residues probably lining the lumen of the toxin channel. Substitution of glutamate-399 of C2II with alanine blocked channel formation and cytotoxicity of the holotoxin. Although cytotoxicity and rounding up of cells by C2I were completely blocked by exchange of phenylalanine-428 with alanine, the mutation increased potassium conductance caused by C2II in artificial membranes by about 2-3-fold over that of wild-type toxin. In contrast to its effects on single-channel potassium conductance in artificial membranes, the F428A mutation delayed the kinetics of pore formation in lipid vesicles and inhibited the activity of C2II in promoting (86)Rb (+) release from preloaded intact cells after pH shift of the medium. Moreover, F428A C2II exhibited delayed and diminished formation of C2II aggregates at low pH, indicating major changes of the biophysical properties of the toxin. The data indicate that phenylalanine-428 of C2II plays a major role in conformational changes occurring during pore formation of the binding component of C2II.


Assuntos
Toxinas Botulínicas/química , Toxinas Botulínicas/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Porinas/química , Porinas/metabolismo , Sequência de Aminoácidos , Animais , Células CACO-2 , Bovinos , Células Cultivadas , Clostridium botulinum/química , Clostridium botulinum/genética , Células HeLa , Humanos , Mutagênese Sítio-Dirigida
19.
PLoS Pathog ; 2(10): e111, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17054395

RESUMO

Anthrax toxin receptors 1 and 2 (ANTXR1 and ANTXR2) have a related integrin-like inserted (I) domain which interacts with a metal cation that is coordinated by residue D683 of the protective antigen (PA) subunit of anthrax toxin. The receptor-bound metal ion and PA residue D683 are critical for ANTXR1-PA binding. Since PA can bind to ANTXR2 with reduced affinity in the absence of metal ions, we reasoned that D683 mutant forms of PA might specifically interact with ANTXR2. We show here that this is the case. The differential ability of ANTXR1 and ANTXR2 to bind D683 mutant PA proteins was mapped to nonconserved receptor residues at the binding interface with PA domain 2. Moreover, a D683K mutant form of PA that bound specifically to human and rat ANTXR2 mediated killing of rats by anthrax lethal toxin, providing strong evidence for the physiological importance of ANTXR2 in anthrax disease pathogenesis.


Assuntos
Antraz/microbiologia , Antígenos de Bactérias/toxicidade , Bacillus anthracis/patogenicidade , Toxinas Bacterianas/toxicidade , Receptores de Peptídeos/metabolismo , Animais , Antraz/imunologia , Antraz/metabolismo , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Bacillus anthracis/imunologia , Bacillus anthracis/metabolismo , Toxinas Bacterianas/imunologia , Sequência de Bases , Humanos , Longevidade/efeitos dos fármacos , Masculino , Proteínas de Membrana/imunologia , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Proteínas de Neoplasias/imunologia , Ratos , Ratos Endogâmicos F344 , Receptores de Superfície Celular/imunologia , Receptores de Peptídeos/imunologia
20.
Mol Cell Biol ; 25(13): 5492-8, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15964805

RESUMO

The enzymatic moieties of anthrax toxin enter the cytosol of mammalian cells via a pore in the endosomal membrane formed by the protective antigen (PA) moiety. Pore formation involves an acidic pH-induced conformational rearrangement of a heptameric precursor (the prepore), in which the seven 2beta2-2beta3 loops interact to generate a 14-strand transmembrane beta-barrel. To investigate this model in vivo, we labeled PA with the fluorophore 7-nitrobenz-2-oxa-1,3-diazole (NBD) at cysteine residues introduced into the 2beta2-2beta3 loop. Each labeled PA was bound to CHO cells, and NBD fluorescence was monitored over time in stirred cell suspensions or by confocal microscopy. A strong increase was observed with NBD at positions 305, 307, 309, and 311, sites where side chains are predicted to face the bilayer, and little change was seen at residues 304, 306, 308, 310, and 312, sites where side chains are predicted to face the pore lumen. The increase at position 305 was inhibited by membrane-restricted quenchers, low temperature, or various reagents known to affect toxin action. Of the 24 NBD attachment sites examined, all but three gave results qualitatively consistent with the beta-barrel model. Besides supporting the beta-barrel model of membrane insertion, our results describe the time course of insertion and identify PA residues where NBD gives a strong signal upon membrane insertion in vivo.


Assuntos
Antígenos de Bactérias/imunologia , Bacillus anthracis/imunologia , Toxinas Bacterianas/imunologia , Membrana Celular/metabolismo , 4-Cloro-7-nitrobenzofurazano , Animais , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Bacillus anthracis/química , Bacillus anthracis/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Cisteína/química , Escherichia coli/genética , Fluorescência , Corantes Fluorescentes , Concentração de Íons de Hidrogênio , Indicadores e Reagentes , Cinética , Bicamadas Lipídicas/química , Microscopia Confocal , Modelos Biológicos , Mutação , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA