Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
mBio ; 7(2): e00605-16, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-27084955
5.
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
6.
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
7.
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
8.
Mol Cancer Ther ; 12(10): 2273-81, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23945077

RESUMO

Chimeric protein toxins that act selectively on cells expressing a designated receptor may serve as investigational probes and/or antitumor agents. Here, we report use of the enzyme sortase A (SrtA) to create four chimeric toxins designed to selectively kill cells bearing the tumor marker HER2. We first expressed and purified: (i) a receptor recognition-deficient form of diphtheria toxin that lacks its receptor-binding domain and (ii) a mutated, receptor-binding-deficient form of anthrax-protective antigen. Both proteins carried at the C terminus the sortase recognition sequence LPETGG and a H6 affinity tag. Each toxin protein was mixed with SrtA plus either of two HER2-recognition proteins--a single-chain antibody fragment or an Affibody--both carrying an N-terminal G5 tag. With wild-type SrtA, the fusion reaction between the toxin and receptor-recognition proteins approached completion only after several hours, whereas with an evolved form of the enzyme, SrtA*, the reaction was virtually complete within 5 minutes. The four fusion toxins were purified and shown to kill HER2-positive cells in culture with high specificity. Sortase-mediated ligation of binary combinations of diverse natively folded proteins offers a facile way to produce large sets of chimeric proteins for research and medicine.


Assuntos
Aminoaciltransferases/genética , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Cisteína Endopeptidases/genética , Toxina Diftérica/genética , Imunotoxinas/genética , Proteínas Recombinantes de Fusão/genética , Sequência de Aminoácidos , Antígenos de Bactérias/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Sítios de Ligação , Linhagem Celular Tumoral , Toxina Diftérica/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imunotoxinas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Ligação Proteica , Receptor ErbB-2/genética , Proteínas Recombinantes de Fusão/uso terapêutico
9.
ACS Chem Biol ; 8(4): 812-22, 2013 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-23343607

RESUMO

Studying how pathogens subvert the host to cause disease has contributed to the understanding of fundamental cell biology. Bacillus anthracis, the causative agent of anthrax, produces the virulence factor lethal toxin to disarm host immunity and cause pathology. We conducted a phenotypic small molecule screen to identify inhibitors of lethal toxin-induced macrophage cell death and used an ordered series of secondary assays to characterize the hits and determine their effects on cellular function. We identified a structurally diverse set of small molecules that act at various points along the lethal toxin pathway, including inhibitors of endocytosis, natural product inhibitors of organelle acidification (e.g., the botulinum neurotoxin inhibitor, toosendanin), and a novel proteasome inhibitor, 4MNB (4-methoxy-2-[2-(5-methoxy-2-nitrosophenyl)ethyl]-1-nitrosobenzene). Many of the compounds, including three drugs approved for use in humans, also protected against the related Clostridium difficile toxin TcdB, further demonstrating their value as novel tools for perturbation and study of toxin biology and host cellular processes and highlighting potential new strategies for intervening on toxin-mediated diseases.


Assuntos
Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Morte Celular/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Macrófagos/efeitos dos fármacos , Animais , Linhagem Celular , Medicamentos de Ervas Chinesas/farmacologia , Endocitose/efeitos dos fármacos , Macrófagos/citologia , Camundongos
10.
Mol Oncol ; 7(3): 440-51, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23290417

RESUMO

Targeted therapeutics have emerged in recent years as an attractive approach to treating various types of cancer. One approach is to modify a cytocidal protein toxin to direct its action to a specific population of cancer cells. We created a targeted toxin in which the receptor-binding and pore-forming moiety of anthrax toxin, termed Protective Antigen (PA), was modified to redirect its receptor specificity to HER2, a marker expressed at the surface of a significant fraction of breast and ovarian tumors. The resulting fusion protein (mPA-ZHER2) delivered cytocidal effectors specifically into HER2-positive tumor cells, including a trastuzumab-resistant line, causing death of the cells. No off-target killing of HER2-negative cells was observed, either with homogeneous populations or with mixtures of HER2-positive and HER2-negative cells. A mixture of mPA variants targeting different receptors mediated killing of cells bearing either receptor, without affecting cells devoid of these receptors. Anthrax toxin may serve as an effective platform for developing therapeutics to ablate cells bearing HER2 or other tumor-specific cell-surface markers.


Assuntos
Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Neoplasias/tratamento farmacológico , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/genética , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/genética , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Humanos , Neoplasias/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Trastuzumab
11.
mBio ; 3(3)2012.
Artigo em Inglês | MEDLINE | ID: mdl-22550037

RESUMO

UNLABELLED: The actions of many bacterial toxins depend on their ability to bind to one or more cell-surface receptors. Anthrax toxin acts by a sequence of events that begins when the protective-antigen (PA) moiety of the toxin binds to either one of two cell-surface proteins, ANTXR1 and ANTXR2, and is proteolytically activated. The activated PA self-associates to form oligomeric pore precursors, which, in turn, bind the enzymatic moieties of the toxin and transport them to the cytosol. We introduced a double mutation into domain 4 of PA to ablate its native receptor-binding function and fused epidermal growth factor (EGF) to the C terminus of the mutated protein. The resulting fusion protein transported enzymatic effector proteins into a cell line that expressed the EGF receptor (A431 cells), but not into a line lacking this receptor (CHO-K1 cells). Addition of excess free EGF blocked transport of effector proteins into A431 cells via the fusion protein, but not via native PA. We also showed that fusing the diphtheria toxin receptor-binding domain to the C terminus of the mutated PA channeled effector-protein transport through the diphtheria toxin receptor. PA fusion proteins with altered receptor specificity may be useful in biological research and could have practical applications, including ablation or perturbation of selected populations of cells in vivo. IMPORTANCE: Bacterial toxins that act within mammalian cells have receptor-dependent mechanisms to transport their enzymatic components to the cytoplasmic compartment. By inactivating or otherwise modifying their respective intracellular targets, these intracellular effectors disrupt metabolic pathways and in some cases cause death of the cell. Our results show that the receptor specificity of the transport protein of anthrax toxin may be readily changed, raising the possibility that receptor-redirected forms of protective antigen (PA) and PA homologs may be useful for research and medical applications requiring modification or ablation of designated populations of cells.


Assuntos
Antraz/metabolismo , Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Animais , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Linhagem Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Peptídeos , Especificidade da Espécie
12.
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
13.
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
14.
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
16.
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
17.
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
18.
J Mol Biol ; 402(1): 1-7, 2010 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-20654627

RESUMO

The diphtheria toxin T domain translocates the catalytic C domain across the endosomal membrane in response to acidification. To elucidate the role of histidine protonation in modulating pH-dependent membrane action of the T domain, we have used site-directed mutagenesis coupled with spectroscopic and physiological assays. Replacement of H257 with an arginine (but not with a glutamine) resulted in dramatic unfolding of the protein at neutral pH, accompanied by a substantial loss of helical structure and greatly increased exposure of the buried residues W206 and W281. This unfolding and spectral shift could be reversed by the interaction of the H257R mutant with model lipid membranes. Remarkably, this greatly unfolded mutant exhibited wild-type-like activity in channel formation, N-terminus translocation, and cytotoxicity assays. Moreover, membrane permeabilization caused by the H257R mutant occurs already at pH 6, where wild type protein is inactive. We conclude that protonation of H257 acts as a major component of the pH-dependent conformational switch, resulting in destabilization of the folded structure in solution and thereby promoting the initial membrane interactions necessary for translocation.


Assuntos
Toxina Diftérica/química , Substituição de Aminoácidos , Membrana Celular/química , Cristalografia por Raios X , Toxina Diftérica/genética , Toxina Diftérica/metabolismo , Histidina/metabolismo , Concentração de Íons de Hidrogênio , Bicamadas Lipídicas , Mutagênese Sítio-Dirigida , Conformação Proteica , Estrutura Terciária de Proteína , Prótons
19.
PLoS One ; 5(5): e10553, 2010 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-20479891

RESUMO

BACKGROUND: Cell-surface receptors play essential roles in anthrax toxin action by providing the toxin with a high-affinity anchor and self-assembly site on the plasma membrane, mediating the toxin entry into cells through endocytosis, and shifting the pH threshold for prepore-to-pore conversion of anthrax toxin protective antigen (PA) to a more acidic pH, thereby inhibiting premature pore formation. Each of the two known anthrax toxin receptors, ANTXR1 and ANTXR2, has an ectodomain comprised of an N-terminal von Willebrand factor A domain (VWA), which binds PA, and an uncharacterized immunoglobulin-like domain (Ig) that connects VWA to the membrane-spanning domain. Potential roles of the receptor Ig domain in anthrax toxin action have not been investigated heretofore. METHODOLOGY/PRINCIPAL FINDINGS: We expressed and purified the ANTXR2 ectodomain (R2-VWA-Ig) in E. coli and showed that it contains three disulfide bonds: one in R2-VWA and two in R2-Ig. Reduction of the ectodomain inhibited functioning of the pore, as measured by K(+) release from liposomes or Chinese hamster ovary cells or by PA-mediated translocation of a model substrate across the plasma membrane. However, reduction did not affect binding of the ectodomain to PA or the transition of ectodomain-bound PA prepore to the pore conformation. The inhibitory effect depended specifically on reduction of the disulfides within R2-Ig. CONCLUSIONS/SIGNIFICANCE: We conclude that disulfide integrity within R2-Ig is essential for proper functioning of receptor-bound PA pore. This finding provides a novel venue to investigate the mechanism of anthrax toxin action and suggests new strategies for inhibiting toxin action.


Assuntos
Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Dissulfetos/metabolismo , Receptores de Peptídeos/química , Receptores de Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/química , Toxinas Bacterianas/química , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Escherichia coli/metabolismo , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Dados de Sequência Molecular , Oxirredução/efeitos dos fármacos , Potássio/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes/isolamento & purificação , Substâncias Redutoras/farmacologia , Dodecilsulfato de Sódio/farmacologia
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...