Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Cell Microbiol ; 19(7)2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28103630

RESUMEN

Orientia tsutsugamushi causes scrub typhus, a potentially fatal infection that afflicts 1 million people annually. This obligate intracellular bacterium boasts one of the largest microbial arsenals of ankyrin repeat-containing protein (Ank) effectors, most of which target the endoplasmic reticulum (ER) by undefined mechanisms. Ank9 is the only one proven to function during infection. Here, we demonstrate that Ank9 bears a motif that mimics the GRIP domain of eukaryotic golgins and is necessary and sufficient for its Golgi localization. Ank9 reaches the ER exclusively by retrograde trafficking from the Golgi. Consistent with this observation, it binds COPB2, a host protein that mediates Golgi-to-ER transport. Ank9 destabilizes the Golgi and ER in a Golgi localization domain-dependent manner and induces the activating transcription factor 4-dependent unfolded protein response. The Golgi is also destabilized in cells infected with O. tsutsugamushi or treated with COPB2 small interfering RNA. COPB2 reduction and/or the cellular events that it invokes, such as Golgi destabilization, benefit Orientia replication. Thus, Ank9 or bacterial negative modulation of COPB2 might contribute to the bacterium's intracellular replication. This report identifies a novel microbial Golgi localization domain, links Ank9 to the ability of O. tsutsugamushi to perturb Golgi structure, and describes the first mechanism by which any Orientia effector targets the secretory pathway.


Asunto(s)
Ancirinas/metabolismo , Proteínas Bacterianas/metabolismo , Proteína Coatómero/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Orientia tsutsugamushi/patogenicidad , Factor de Transcripción Activador 4/metabolismo , Línea Celular Tumoral , Células HeLa , Humanos , Unión Proteica/fisiología , Dominios Proteicos/fisiología , Transporte de Proteínas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , Tifus por Ácaros/microbiología , Respuesta de Proteína Desplegada/fisiología
2.
Infect Immun ; 85(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27993973

RESUMEN

Anaplasma marginale causes bovine anaplasmosis, a debilitating and potentially fatal tick-borne infection of cattle. Because A. marginale is an obligate intracellular organism, its adhesins that mediate entry into host cells are essential for survival. Here, we demonstrate that A. marginale outer membrane protein A (AmOmpA; AM854) contributes to the invasion of mammalian and tick host cells. AmOmpA exhibits predicted structural homology to OmpA of A. phagocytophilum (ApOmpA), an adhesin that uses key lysine and glycine residues to interact with α2,3-sialylated and α1,3-fucosylated glycan receptors, including 6-sulfo-sialyl Lewis x (6-sulfo-sLex). Antisera against AmOmpA or its predicted binding domain inhibits A. marginale infection of host cells. Residues G55 and K58 are contributory, and K59 is essential for recombinant AmOmpA to bind to host cells. Enzymatic removal of α2,3-sialic acid and α1,3-fucose residues from host cell surfaces makes them less supportive of AmOmpA binding. AmOmpA is both an adhesin and an invasin, as coating inert beads with it confers adhesiveness and invasiveness. Recombinant forms of AmOmpA and ApOmpA competitively antagonize A. marginale infection of host cells, but a monoclonal antibody against 6-sulfo-sLex fails to inhibit AmOmpA adhesion and A. marginale infection. Thus, the two OmpA proteins bind related but structurally distinct receptors. This study provides a detailed understanding of AmOmpA function, identifies its essential residues that can be targeted by blocking antibody to reduce infection, and determines that it binds to one or more α2,3-sialylated and α1,3-fucosylated glycan receptors that are unique from those targeted by ApOmpA.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Anaplasma marginale/fisiología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Polisacáridos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Adhesinas Bacterianas/química , Secuencias de Aminoácidos , Aminoácidos , Animales , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/inmunología , Sitios de Unión , Línea Celular , Fucosa/metabolismo , Sueros Inmunes/inmunología , Modelos Moleculares , Conformación Molecular , Ácido N-Acetilneuramínico/metabolismo , Polisacáridos/química , Unión Proteica
3.
PLoS Pathog ; 11(2): e1004669, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25658707

RESUMEN

Anaplasma phagocytophilum causes granulocytic anaplasmosis, an emerging disease of humans and domestic animals. The obligate intracellular bacterium uses its invasins OmpA, Asp14, and AipA to infect myeloid and non-phagocytic cells. Identifying the domains of these proteins that mediate binding and entry, and determining the molecular basis of their interactions with host cell receptors would significantly advance understanding of A. phagocytophilum infection. Here, we identified the OmpA binding domain as residues 59 to 74. Polyclonal antibody generated against a peptide spanning OmpA residues 59 to 74 inhibited A. phagocytophilum infection of host cells and binding to its receptor, sialyl Lewis x (sLe(x)-capped P-selectin glycoprotein ligand 1. Molecular docking analyses predicted that OmpA residues G61 and K64 interact with the two sLe(x) sugars that are important for infection, α2,3-sialic acid and α1,3-fucose. Amino acid substitution analyses demonstrated that K64 was necessary, and G61 was contributory, for recombinant OmpA to bind to host cells and competitively inhibit A. phagocytophilum infection. Adherence of OmpA to RF/6A endothelial cells, which express little to no sLe(x) but express the structurally similar glycan, 6-sulfo-sLe(x), required α2,3-sialic acid and α1,3-fucose and was antagonized by 6-sulfo-sLe(x) antibody. Binding and uptake of OmpA-coated latex beads by myeloid cells was sensitive to sialidase, fucosidase, and sLe(x) antibody. The Asp14 binding domain was also defined, as antibody specific for residues 113 to 124 inhibited infection. Because OmpA, Asp14, and AipA each contribute to the infection process, it was rationalized that the most effective blocking approach would target all three. An antibody cocktail targeting the OmpA, Asp14, and AipA binding domains neutralized A. phagocytophilum binding and infection of host cells. This study dissects OmpA-receptor interactions and demonstrates the effectiveness of binding domain-specific antibodies for blocking A. phagocytophilum infection.


Asunto(s)
Anaplasma phagocytophilum , Proteínas de la Membrana Bacteriana Externa , Ehrlichiosis , Simulación del Acoplamiento Molecular , Sustitución de Aminoácidos , Anaplasma phagocytophilum/química , Anaplasma phagocytophilum/genética , Anaplasma phagocytophilum/metabolismo , Anaplasma phagocytophilum/patogenicidad , Animales , Anticuerpos Antibacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Células CHO , Cricetinae , Cricetulus , Ehrlichiosis/genética , Ehrlichiosis/metabolismo , Células HL-60 , Humanos , Mutación Missense , Unión Proteica , Estructura Terciaria de Proteína
4.
Infect Immun ; 84(7): 2051-2058, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27113359

RESUMEN

The Treponema denticola FhbB protein contributes to immune evasion by binding factor H (FH). Cleavage of FH by the T. denticola protease, dentilisin, may contribute to the local immune dysregulation that is characteristic of periodontal disease (PD). Although three FhbB phyletic types have been defined (FhbB1, FhbB2, and FhbB3), the in vivo expression patterns and antigenic heterogeneity of FhbB have not been assessed. Here, we demonstrate that FhbB is a dominant early antigen that elicits FhbB type-specific antibody (Ab) responses. Using the murine skin abscess model, we demonstrate that the presence or absence of FhbB or dentilisin significantly influences Ab responses to infection and skin abscess formation. Competitive binding analyses revealed that α-FhbB Ab can compete with FH for binding to T. denticola and block dentilisin-mediated FH cleavage. Lastly, we demonstrate that dentilisin cleavage sites reside within critical functional domains of FH, including the complement regulatory domain formed by CCPs 1 to 4. Analysis of the FH cleavage products revealed that they lack cofactor activity. The data presented here provide insight into the in vivo significance of dentilisin, FhbB and its antigenic diversity, and the potential impact of FH cleavage on the regulation of complement activation.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Péptido Hidrolasas/metabolismo , Treponema denticola/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Especificidad de Anticuerpos/inmunología , Antígenos Bacterianos/metabolismo , Unión Competitiva , Factor H de Complemento/química , Ratones , Unión Proteica/efectos de los fármacos , Unión Proteica/inmunología , Dominios y Motivos de Interacción de Proteínas , Proteolisis
5.
J Bacteriol ; 197(19): 3097-109, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26170417

RESUMEN

UNLABELLED: A rising theme among intracellular microbes is the delivery of ankyrin repeat-containing effectors (Anks) that interact with target proteins to co-opt host cell functions. Orientia tsutsugamushi, an obligate intracellular bacterium and the etiologic agent of scrub typhus, encodes one of the largest Ank repertoires of any sequenced microorganism. They have been previously identified as type 1 secretion system substrates. Here, in silico and manual sequence analyses revealed that a large proportion of O. tsutsugamushi strain Ikeda Anks bear a eukaryotic/poxvirus-like F-box motif, which is known to recruit host cell SCF1 ubiquitin ligase machinery. We assessed the Anks for the ability to serve as F-box proteins. Coimmunoprecipitation assays demonstrated that F-box-containing Anks interact with overexpressed and/or endogenous SCF1 components. When coexpressed with FLAG-Ank4_01 or FLAG-Ank9, a glutathione S-transferase (GST)-tagged version of the SCF1 component SKP1 localized to subcellular sites of FLAG-Ank accumulation. The abilities of recombinant Anks to interact and colocalize with SKP1 were F-box dependent. GST-SKP1 precipitated O. tsutsugamushi-derived Ank9 from infected host cells, verifying both that the pathogen expresses Ank9 during infection and the protein's capability to bind SKP1. Aligning O. tsutsugamushi, poxviral, and eukaryotic F-box sequences delineated three F-box residues that are highly conserved and likely to be functionally important. Substitution of these residues ablated the ability of GFP-Ank9 to interact with GST-SKP1. These results demonstrate that O. tsutsugamushi strain Ikeda Anks can co-opt host cell polyubiquitination machinery, provide the first evidence that an O. tsutsugamushi Ank does so during infection, and advance overall understanding of microbial F-box proteins. IMPORTANCE: Ankyrin repeat-containing proteins (Anks) are important virulence factors of intracellular bacteria that mediate protein-protein interactions with host cell targets. Orientia tsutsugamushi, which causes a debilitating infection called scrub typhus in one of the most densely populated regions of the world, encodes one of the largest Ank armamentariums of any sequenced bacterium. This study demonstrates that O. tsutsugamushi strain Ikeda Anks also bear F-box motifs that interact with host cell polyubiquitination machinery. By proving that an Orientia-derived Ank interacts with SKP1 in infected cells, this evidences the first bona fide Orientia effector and the first example of an endogenous F-box-containing Ank-mammalian-host ligand interaction for any intracellular bacterium. Also, importantly, this work identifies key residues that are essential for microbial F-box function.


Asunto(s)
Repetición de Anquirina , Proteínas Bacterianas/metabolismo , Proteínas F-Box/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Orientia tsutsugamushi/clasificación , Orientia tsutsugamushi/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas F-Box/genética , Células HeLa , Humanos , Datos de Secuencia Molecular , Orientia tsutsugamushi/genética , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Ubiquitina-Proteína Ligasas/genética
6.
Cell Microbiol ; 16(8): 1133-45, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24612118

RESUMEN

Anaplasma phagocytophilum, which causes granulocytic anaplasmosis in humans and animals, is a tick-transmitted obligate intracellular bacterium that mediates its own uptake into neutrophils and non-phagocytic cells. Invasins of obligate intracellular pathogens are attractive targets for protecting against or curing infection because blocking the internalization step prevents survival of these organisms. The complement of A. phagocytophilum invasins is incompletely defined. Here, we report the significance of a novel A. phagocytophilum invasion protein, AipA. A. phagocytophilum induced aipA expression during transmission feeding of infected ticks on mice. The bacterium upregulated aipA transcription when it transitioned from its non-infectious reticulate cell morphotype to its infectious dense-cored morphotype during infection of HL-60 cells. AipA localized to the bacterial surface and was expressed during in vivo infection. Of the AipA regions predicted to be surface-exposed, only residues 1 to 87 (AipA1-87 ) were found to be essential for host cell invasion. Recombinant AipA1-87 protein bound to and competitively inhibited A. phagocytophilum infection of mammalian cells. Antiserum specific for AipA1-87 , but not other AipA regions, antagonized infection. Additional blocking experiments using peptide-specific antisera narrowed down the AipA invasion domain to residues 9 to 21. An antisera combination targeting AipA1-87 together with two other A. phagocytophilum invasins, OmpA and Asp14, nearly abolished infection of host cells. This study identifies AipA as an A. phagocytophilum surface protein that is critical for infection, demarcates its invasion domain, and establishes a rationale for targeting multiple invasins to protect against granulocytic anaplasmosis.


Asunto(s)
Adhesinas Bacterianas/biosíntesis , Anaplasma phagocytophilum/patogenicidad , Anaplasmosis/microbiología , Proteínas de la Membrana Bacteriana Externa/biosíntesis , Ehrlichiosis/patología , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Anaplasma phagocytophilum/inmunología , Anaplasmosis/inmunología , Anaplasmosis/patología , Animales , Anticuerpos Monoclonales/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Células CHO , Línea Celular Tumoral , Cricetulus , Ehrlichiosis/inmunología , Ehrlichiosis/microbiología , Células HL-60 , Humanos , Sueros Inmunes/inmunología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Garrapatas , Regulación hacia Arriba
7.
Infect Immun ; 82(8): 3324-32, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24866803

RESUMEN

The primary causative agent of tick-borne relapsing fever in North America is Borrelia hermsii. It has been hypothesized that B. hermsii evades complement-mediated destruction by binding factor H (FH), a host-derived negative regulator of complement. In vitro, B. hermsii produces a single FH binding protein designated FhbA (FH binding protein A). The properties and ligand binding activity of FhbA suggest that it plays multiple roles in pathogenesis. It binds plasminogen and has been identified as a significant target of a B1b B cell-mediated IgM response in mice. FhbA has also been explored as a potential diagnostic antigen for B. hermsii infection in humans. The ability to test the hypothesis that FhbA is a critical virulence factor in vivo has been hampered by the lack of well-developed systems for the genetic manipulation of the relapsing fever spirochetes. In this report, we have successfully generated a B. hermsii fhbA deletion mutant (the B. hermsii YORΔfhbA strain) through allelic exchange mutagenesis. Deletion of fhbA abolished FH binding by the YORΔfhbA strain and eliminated cleavage of C3b on the cell surface. However, the YORΔfhbA strain remained infectious in mice and retained resistance to killing in vitro by human complement. Collectively, these results indicate that B. hermsii employs an FhbA/FH-independent mechanism of complement evasion that allows for resistance to killing by human complement and persistence in mice.


Asunto(s)
Actividad Bactericida de la Sangre , Borrelia/inmunología , Proteínas Portadoras/metabolismo , Proteínas del Sistema Complemento/inmunología , Fiebre Recurrente/inmunología , Fiebre Recurrente/microbiología , Factores de Virulencia/metabolismo , Animales , Borrelia/genética , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Eliminación de Gen , Humanos , Ratones , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda