Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Glycobiology ; 34(1)2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-37944064

RESUMEN

During the COVID-19 outbreak, numerous tools including protein-based vaccines have been developed. The methylotrophic yeast Pichia pastoris (synonymous to Komagataella phaffii) is an eukaryotic cost-effective and scalable system for recombinant protein production, with the advantages of an efficient secretion system and the protein folding assistance of the secretory pathway of eukaryotic cells. In a previous work, we compared the expression of SARS-CoV-2 Spike Receptor Binding Domain in P. pastoris with that in human cells. Although the size and glycosylation pattern was different between them, their protein structural and conformational features were indistinguishable. Nevertheless, since high mannose glycan extensions in proteins expressed by yeast may be the cause of a nonspecific immune recognition, we deglycosylated RBD in native conditions. This resulted in a highly pure, homogenous, properly folded and monomeric stable protein. This was confirmed by circular dichroism and tryptophan fluorescence spectra and by SEC-HPLC, which were similar to those of RBD proteins produced in yeast or human cells. Deglycosylated RBD was obtained at high yields in a single step, and it was efficient in distinguishing between SARS-CoV-2-negative and positive sera from patients. Moreover, when the deglycosylated variant was used as an immunogen, it elicited a humoral immune response ten times greater than the glycosylated form, producing antibodies with enhanced neutralizing power and eliciting a more robust cellular response. The proposed approach may be used to produce at a low cost, many antigens that require glycosylation to fold and express, but do not require glycans for recognition purposes.


Asunto(s)
COVID-19 , Saccharomycetales , Vacunas , Humanos , COVID-19/diagnóstico , COVID-19/prevención & control , Prueba de COVID-19 , Pichia/genética , Pichia/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Proteínas Recombinantes/química , Vacunas/metabolismo , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales
2.
Cell Microbiol ; 22(11): e13245, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32657513

RESUMEN

Adhesion to host cells is the first step in the virulence cycle of any pathogen. In Gram-negative bacteria, adhesion is mediated, among other virulence factors such as the lipopolysaccharides, by specific outer-membrane proteins generally termed adhesins that belong to a wide variety of families and have different evolutionary origins. In Brucella, a widespread zoonotic pathogen of animal and human health concern, adhesion is central as it may determine the intracellular fate of the bacterium, an essential stage in its pathogenesis. In the present paper, we further characterised a genomic locus that we have previously reported encodes an adhesin (BigA) with a bacterial immunoglobulin-like domain (BIg-like). We found that this region encodes a second adhesin, which we have named BigB; and PalA, a periplasmic protein necessary for the proper display in the outer membrane of BigA and BigB. Deletion of bigB or palA diminishes the adhesion of the bacterium and overexpression of BigB dramatically increases it. Incubation of cells with the recombinant BIg-like domain of BigB induced important cytoskeletal rearrangements and affected the focal adhesion sites indicating that the adhesin targets cell-cell or cell-matrix proteins. We additionally show that PalA has a periplasmic localisation and is completely necessary for the proper display of BigA and BigB, probably avoiding their aggregation and facilitating their transport to the outer membrane. Our results indicate that this genomic island is entirely devoted to the adhesion of Brucella to host cells.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana/genética , Proteínas Bacterianas/metabolismo , Brucella abortus/genética , Brucella abortus/patogenicidad , Islas Genómicas , Adhesinas Bacterianas/genética , Animales , Membrana Externa Bacteriana/metabolismo , Proteínas Bacterianas/genética , Brucella abortus/fisiología , Línea Celular , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Humanos , Periplasma/metabolismo , Virulencia
3.
J Virol ; 93(3)2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30404802

RESUMEN

After initiation of an infective cycle, spread of virus infection can occur in two fundamentally different ways: (i) viral particles can be released into the external environment and diffuse through the extracellular space until they interact with a new host cell, and (ii) virions can remain associated with infected cells, promoting the direct passage between infected and uninfected cells that is referred to as direct cell-to-cell transmission. Although evidence of cell-associated transmission has accumulated for many different viruses, the ability of members of the genus Pestivirus to use this mode of transmission has not been reported. In the present study, we used a novel recombinant virus expressing the envelope glycoprotein E2 fused to mCherry fluorescent protein to monitor the spreading of bovine viral diarrhea virus (BVDV) (the type member of the pestiviruses) infection. To demonstrate direct cell-to-cell transmission of BVDV, we developed a cell coculture system that allowed us to prove direct transmission from infected to uninfected cells in the presence of neutralizing antibodies. This mode of transmission requires cell-cell contacts and clathrin-mediated receptor-dependent endocytosis. Notably, it overcomes antibody blocking of the BVDV receptor CD46, indicating that cell-to-cell transmission of the virus involves the engagement of coreceptors on the target cell.IMPORTANCE BVDV causes one of the most economically important viral infections for the cattle industry. The virus is able to cross the placenta and infect the fetus, leading to the birth of persistently infected animals, which are reservoirs for the spread of BVDV. The occurrence of persistent infection has hampered the efficacy of vaccination because it requires eliciting levels of protection close to sterilizing immunity to prevent fetal infections. While vaccination prevents disease, BVDV can be detected if animals with neutralizing antibodies are challenged with the virus. Virus cell-to-cell transmission allows the virus to overcome barriers to free virus dissemination, such as antibodies or epithelial barriers. Here we show that BVDV exploits cell-cell contacts to propagate infection in a process that is resistant to antibody neutralization. Our results provide new insights into the mechanisms underlying the pathogenesis of BVDV infection and can aid in the design of effective control strategies.


Asunto(s)
Diarrea Mucosa Bovina Viral/virología , Comunicación Celular , Virus de la Diarrea Viral Bovina Tipo 1/patogenicidad , Interacciones Huésped-Patógeno , Proteínas del Envoltorio Viral/metabolismo , Replicación Viral , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Diarrea Mucosa Bovina Viral/genética , Diarrea Mucosa Bovina Viral/metabolismo , Bovinos , Células Cultivadas , Proteínas del Envoltorio Viral/genética , Internalización del Virus
4.
Infect Immun ; 87(8)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31182616

RESUMEN

The peptidoglycan in Gram-negative bacteria is a dynamic structure in constant remodeling. This dynamism, achieved through synthesis and degradation, is essential because the peptidoglycan is necessary to maintain the structure of the cell but has to have enough plasticity to allow the transport and assembly of macromolecular complexes in the periplasm and outer membrane. In addition, this remodeling has to be coordinated with the division process. Among the multiple mechanisms bacteria have to degrade the peptidoglycan are the lytic transglycosidases, enzymes of the lysozyme family that cleave the glycan chains generating gaps in the mesh structure increasing its permeability. Because these enzymes can act as autolysins, their activity has to be tightly regulated, and one of the mechanisms bacteria have evolved is the synthesis of membrane bound or periplasmic inhibitors. In the present study, we identify a periplasmic lytic transglycosidase inhibitor (PhiA) in Brucella abortus and demonstrate that it inhibits the activity of SagA, a lytic transglycosidase we have previously shown is involved in the assembly of the type IV secretion system. A phiA deletion mutant results in a strain with the incapacity to synthesize a complete lipopolysaccharide but with a higher replication rate than the wild-type parental strain, suggesting a link between peptidoglycan remodeling and speed of multiplication.


Asunto(s)
Brucella abortus/patogenicidad , N-Acetil Muramoil-L-Alanina Amidasa/antagonistas & inhibidores , Glicósido Hidrolasas/fisiología , Lipopolisacáridos/biosíntesis , Complejos Multienzimáticos/fisiología , Peptidoglicano/metabolismo , Transferasas/fisiología , Sistemas de Secreción Tipo IV/fisiología , Virulencia
5.
J Infect Dis ; 217(8): 1257-1266, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29325043

RESUMEN

Brucellaceae are stealthy pathogens with the ability to survive and replicate in the host in the context of a strong immune response. This capacity relies on several virulence factors that are able to modulate the immune system and in their structural components that have low proinflammatory activities. Lipopolysaccharide (LPS), the main component of the outer membrane, is a central virulence factor of Brucella, and it has been well established that it induces a low inflammatory response. We describe here the identification and characterization of a novel periplasmic protein (RomA) conserved in alpha-proteobacteria, which is involved in the homeostasis of the outer membrane. A mutant in this gene showed several phenotypes, such as membrane defects, altered LPS composition, reduced adhesion, and increased virulence and inflammation. We show that RomA is involved in the synthesis of LPS, probably coordinating part of the biosynthetic complex in the periplasm. Its absence alters the normal synthesis of this macromolecule and affects the homeostasis of the outer membrane, resulting in a strain with a hyperinflammatory phenotype. Our results suggest that the proper synthesis of LPS is central to maximize virulence and minimize inflammation.


Asunto(s)
Proteínas Bacterianas/fisiología , Brucella/metabolismo , Brucelosis/microbiología , Lipopolisacáridos/biosíntesis , Animales , Brucella/patogenicidad , Gentamicinas , Inflamación/metabolismo , Ratones , Transporte de Proteínas , Virulencia
6.
J Biol Chem ; 291(23): 12383-93, 2016 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-27059960

RESUMEN

The VirB secretion apparatus in Brucella belongs to the type IV secretion systems present in many pathogenic bacteria and is absolutely necessary for the efficient evasion of the Brucella-containing vacuole from the phagocytic route in professional phagocytes. This system is responsible for the secretion of a plethora of effector proteins that alter the biology of the host cell and promote the intracellular replication process. Although many VirB substrates have been identified in Brucella, we still know very little about the secretion mechanism that mediates their translocation across the two membranes and the periplasmic space. In this manuscript, we describe the identification of a gene, virJ, that codes for a protein with periplasmic localization that is involved in the intracellular replication process and virulence in mice. Our analysis revealed that this protein is necessary for the secretion of at least two VirB substrates that have a periplasmic intermediate and that it directly interacts with them. We additionally show that VirJ also associates with the apparatus per se and that its absence affects the assembly of the complex. We hypothesize that VirJ is part of a secretion platform composed of the translocon and several secretion substrates and that it probably coordinates the proper assembly of this macromolecular complex.


Asunto(s)
Proteínas Bacterianas/metabolismo , Periplasma/metabolismo , Sistemas de Secreción Tipo IV/metabolismo , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Brucella abortus/patogenicidad , Brucelosis/virología , Línea Celular , Células Cultivadas , Interacciones Huésped-Patógeno , Macrófagos/virología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Fluorescente , Unión Proteica , Sistemas de Secreción Tipo IV/genética , Virulencia , Factores de Virulencia/genética
7.
Cell Microbiol ; 18(4): 500-13, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26400021

RESUMEN

Adhesion to cells is the initial step in the infectious cycle of basically all pathogenic bacteria, and to do so, microorganisms have evolved surface molecules that target different cellular receptors. Brucella is an intracellular pathogen that infects a wide range of mammals whose virulence is completely dependent on the capacity to replicate in phagocytes. Although much has been done to elucidate how Brucella multiplies in macrophages, we still do not understand how bacteria invade epithelial cells to perform a replicative cycle or what adhesion molecules are involved in the process. We report the identification in Brucella abortus of a novel adhesin that harbours a bacterial immunoglobulin-like domain and demonstrate that this protein is involved in the adhesion to polarized epithelial cells such as the Caco-2 and Madin-Darby canine kidney models targeting the bacteria to the cell-cell interaction membrane. While deletion of the gene significantly reduced adhesion, over-expression dramatically increased it. Addition of the recombinant protein to cells induced cytoskeleton rearrangements and showed that this adhesin targets proteins of the cell-cell interaction membrane in confluent cultures.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Brucella abortus/fisiología , Células Epiteliales/microbiología , Animales , Brucella abortus/metabolismo , Células CACO-2 , Perros , Eliminación de Gen , Expresión Génica , Humanos , Células de Riñón Canino Madin Darby
8.
Cell Microbiol ; 16(3): 396-410, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24119283

RESUMEN

Brucella abortus, the aetiological agent of bovine brucellosis, is an intracellular pathogen whose virulence is completely dependent on a type IV secretion system. This secretion system translocates effector proteins into the host cell to modulate the intracellular fate of the bacterium in order to establish a secure niche were it actively replicates. Although much has been done in understanding how this secretion system participates in the virulence process, few effector proteins have been identified to date. We describe here the identification of a type IV secretion substrate (SepA) that is only present in Brucella spp. and has no detectable homology to known proteins. This protein is secreted in a virB-dependent manner in a two-step process involving a periplasmic intermediate and secretion is necessary for its function. The deletion mutant showed a defect in the early stages of intracellular replication in professional and non-professional phagocytes although it invades the cells more efficiently than the wild-type parental strain. Our results indicate that, even though the mutant was more invasive, it had a defect in excluding the lysosomal marker Lamp-1 and was inactivated more efficiently during the early phases of the intracellular life cycle.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Brucella abortus/patogenicidad , Animales , Sistemas de Secreción Bacterianos , Brucella abortus/genética , Brucelosis/microbiología , Brucelosis/patología , Línea Celular , Femenino , Regulación Bacteriana de la Expresión Génica , Células HeLa , Humanos , Proteínas de Membrana de los Lisosomas , Ratones , Ratones Endogámicos BALB C , Factores de Virulencia/metabolismo
9.
mBio ; : e0072624, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38847540

RESUMEN

The modulation of actin polymerization is a common theme among microbial pathogens. Even though microorganisms show a wide repertoire of strategies to subvert the activity of actin, most of them converge in the ones that activate nucleating factors, such as the Arp2/3 complex. Brucella spp. are intracellular pathogens capable of establishing chronic infections in their hosts. The ability to subvert the host cell response is dependent on the capacity of the bacterium to attach, invade, avoid degradation in the phagocytic compartment, replicate in an endoplasmic reticulum-derived compartment and egress. Even though a significant number of mechanisms deployed by Brucella in these different phases have been identified and characterized, none of them have been described to target actin as a cellular component. In this manuscript, we describe the identification of a novel virulence factor (NpeA) that promotes niche formation. NpeA harbors a short linear motif (SLiM) present within an amphipathic alpha helix that has been described to bind the GTPase-binding domain (GBD) of N-WASP and stabilizes the autoinhibited state. Our results show that NpeA is secreted in a Type IV secretion system-dependent manner and that deletion of the gene diminishes the intracellular replication capacity of the bacterium. In vitro and ex vivo experiments demonstrate that NpeA binds N-WASP and that the short linear motif is required for the biological activity of the protein.IMPORTANCEThe modulation of actin-binding effectors that regulate the activity of this fundamental cellular protein is a common theme among bacterial pathogens. The neural Wiskott-Aldrich syndrome protein (N-WASP) is a protein that several pathogens target to hijack actin dynamics. The highly adapted intracellular bacterium Brucella has evolved a wide repertoire of virulence factors that modulate many activities of the host cell to establish successful intracellular replication niches, but, to date, no effector proteins have been implicated in the modulation of actin dynamics. We present here the identification of a virulence factor that harbors a short linear motif (SLiM) present within an amphipathic alpha helix that has been described to bind the GTPase-binding domain (GBD) of N-WASP stabilizing its autoinhibited state. We demonstrate that this protein is a Type IV secretion effector that targets N-WASP-promoting intracellular survival and niche formation.

10.
Infect Immun ; 81(3): 956-64, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23319555

RESUMEN

Secretion of proteins in Gram-negative bacteria is a high-energy-consuming process that requires translocation across two membranes and a periplasmic space composed of a mesh-like layer, the peptidoglycan. To achieve this, bacteria have evolved complex secretion systems that cross these barriers, and in many cases there are specific peptidoglycanases that degrade the peptidoglycan to allow the proper assembly of the secretion machinery. We describe here the identification and characterization of a muramidase in Brucella abortus that participates in the intracellular multiplication in professional and nonprofessional phagocytes. We demonstrated that this protein has peptidoglycanase activity, that a strain with a clean deletion of the gene displayed a defect in the early stages of the intracellular multiplication curve, and that this is dependent on the lytic activity. While neither the attachment nor the invasion of the strain was affected, we demonstrated that it had a defect in excluding the lysosomal marker LAMP-1 but not in acquiring the reticulum endoplasmic marker calnexin, indicating that the gene participates in the early stages of the intracellular trafficking but not in the establishment of the replicative niche. Analysis of the assembly status and functionality of the VirB secretion apparatus indicated that the mutant has affected the proper function of this central virulence factor.


Asunto(s)
Proteínas Bacterianas/metabolismo , Brucella abortus/fisiología , Regulación Bacteriana de la Expresión Génica/fisiología , Muramidasa/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/genética , Brucella abortus/citología , Línea Celular , Proliferación Celular , ADN Bacteriano/genética , ADN Recombinante , Células Epiteliales/microbiología , Humanos , Macrófagos/microbiología , Ratones , Datos de Secuencia Molecular , Muramidasa/clasificación , Muramidasa/genética , Mutagénesis Sitio-Dirigida , Plásmidos
11.
mBio ; 14(1): e0333822, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36622142

RESUMEN

Host cell egress is a critical step in the life cycle of intracellular pathogens, especially in microbes capable of establishing chronic infections. The Gram-negative bacterium Brucella belongs to such a group of pathogens. Even though much has been done to understand how Brucella avoids killing and multiplies in its intracellular niche, the mechanism that this bacterium deploys to egress from the cell to complete its cycle has been poorly studied. In the manuscript, we quantify the kinetics of bacterial egress and show that Brucella exploits multivesicular bodies to exit host cells. For the first time, we visualized the process of egress in real time by live video microscopy and showed that a population of intracellular bacteria exit from host cells in vacuoles containing multivesicular body-like features. We observed the colocalization of Brucella with two multivesicular markers, namely, CD63 and LBPA, both during the final stages of the intracellular life cycle and in egressed bacteria. Moreover, drugs that either promote or inhibit multivesicular bodies either increased or decreased the number of extracellular bacteria, respectively. Our results strongly suggest that Brucella hijacks multivesicular bodies to exit the host cells to initiate new infection events. IMPORTANCE How intracellular bacterial pathogens egress from host cells has been poorly studied. This is particularly important because this stage of the infectious cycle can have a strong impact on how the host resolves the infection. Brucella is an intracellular pathogen that infects mammals, including humans, and causes a chronic debilitating illness. The bacterium has evolved a plethora of mechanisms to invade host cells, avoid degradation in the endocytic pathway, and actively multiply within a specialized intracellular compartment. However, how this pathogen exits from infected cells to produce reinfection and complete its life cycle is poorly understood. In the manuscript, we shed some light on the mechanisms that are exploited by Brucella to egress from host cells. We observed for the first time the egress of Brucella from infected cells by time-lapse video microscopy, and we found that the bacterium exits in vesicles containing multivesicular bodies (MVBs) features. Moreover, the drug manipulation of MVBs resulted in the alteration of bacterial egress efficiency. Our results indicate that Brucella hijacks MVBs to exit host cells and that this strongly contributes to the reinfection cycle.


Asunto(s)
Brucella , Humanos , Animales , Cuerpos Multivesiculares , Reinfección/metabolismo , Vacuolas/metabolismo , Bacterias , Mamíferos
12.
Vaccine ; 41(23): 3534-3543, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37149444

RESUMEN

Brucellosis remains one of the most worldwide distributed zoonosis inflicting serious economical and human health problems in many areas of the world. The disease is caused by different species of the genus Brucella that have different tropisms towards different mammals being the most relevant for human health Brucella abortus, Brucella melitensis and Brucella suis that infect cows, goats/sheep, and swine respectively. For B. melitensis, considered the species with more zoonotic potential and highly aggressive for animals, only one vaccine is available to date in the market: Rev 1. This attenuated strain has the disadvantage that is has a very high residual virulence for animals and humans and, for this reason, it is applied by ocular instillation which is technically challenging in many productive settings. For this reason, the search for new vaccines for caprine and ovine brucellosis is an active topic of research. We describe here the construction of a novel highly attenuated vaccine strain (Bm Delta-pgm) that confers excellent levels of protection against B. melitensis in the mouse model of infection. This strain is a clean deletion of the phosphoglucomutase (pgm) gene that codes for a protein that catalyzes the conversion of glucose-6-P to glucose-1-P, which is used as a precursor for the biosynthesis of many polysaccharides, including the O-antigen of the lipopolysaccharide and cyclic beta glucans. Our results indicate that vaccination with Bm Delta-pgm induces a robust memory cellular immune response but no antibody production against the O-antigen. Cross protection experiments show that this new vaccine protects against B. abortus and B. suis raising the possibility that Bm Delta-pgm could be used as a universal vaccine for the most important Brucella species.


Asunto(s)
Vacuna contra la Brucelosis , Brucella melitensis , Brucelosis , Femenino , Ratones , Animales , Ovinos , Bovinos , Humanos , Porcinos , Brucella melitensis/genética , Fosfoglucomutasa/genética , Cabras , Antígenos O , Brucelosis/prevención & control , Brucella abortus
13.
Microb Cell Fact ; 11: 13, 2012 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-22276812

RESUMEN

BACKGROUND: Immune responses directed towards surface polysaccharides conjugated to proteins are effective in preventing colonization and infection of bacterial pathogens. Presently, the production of these conjugate vaccines requires intricate synthetic chemistry for obtaining, activating, and attaching the polysaccharides to protein carriers. Glycoproteins generated by engineering bacterial glycosylation machineries have been proposed to be a viable alternative to traditional conjugation methods. RESULTS: In this work we expressed the C. jejuni oligosaccharyltansferase (OTase) PglB, responsible for N-linked protein glycosylation together with a suitable acceptor protein (AcrA) in Yersinia enterocolitica O9 cells. MS analysis of the acceptor protein demonstrated the transfer of a polymer of N-formylperosamine to AcrA in vivo. Because Y. enterocolitica O9 and Brucella abortus share an identical O polysaccharide structure, we explored the application of the resulting glycoprotein in vaccinology and diagnostics of brucellosis, one of the most common zoonotic diseases with over half a million new cases annually. Injection of the glycoprotein into mice generated an IgG response that recognized the O antigen of Brucella, although this response was not protective against a challenge with a virulent B. abortus strain. The recombinant glycoprotein coated onto magnetic beads was efficient in differentiating between naïve and infected bovine sera. CONCLUSION: Bacterial engineered glycoproteins show promising applications for the development on an array of diagnostics and immunoprotective opportunities in the future.


Asunto(s)
Vacunas Bacterianas/inmunología , Brucelosis Bovina/diagnóstico , Campylobacter jejuni/enzimología , Glicoproteínas/biosíntesis , Ingeniería de Proteínas , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Vacunas Bacterianas/biosíntesis , Vacunas Bacterianas/genética , Brucelosis Bovina/prevención & control , Bovinos , Glicoproteínas/química , Glicoproteínas/genética , Glicosilación , Hexosaminas/metabolismo , Hexosiltransferasas/biosíntesis , Hexosiltransferasas/genética , Inmunoglobulina G/inmunología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Antígenos O/inmunología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Yersinia enterocolitica/metabolismo
14.
J Cell Biol ; 174(7): 997-1007, 2006 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-16982801

RESUMEN

Synaptotagmin (Syt) VII is a ubiquitously expressed member of the Syt family of Ca2+ sensors. It is present on lysosomes in several cell types, where it regulates Ca2+-dependent exocytosis. Because [Ca2+]i and exocytosis have been associated with phagocytosis, we investigated the phagocytic ability of macrophages from Syt VII-/- mice. Syt VII-/- macrophages phagocytose normally at low particle/cell ratios but show a progressive inhibition in particle uptake under high load conditions. Complementation with Syt VII rescues this phenotype, but only when functional Ca2+-binding sites are retained. Reinforcing a role for Syt VII in Ca2+-dependent phagocytosis, particle uptake in Syt VII-/- macrophages is significantly less dependent on [Ca2+]i. Syt VII is concentrated on peripheral domains of lysosomal compartments, from where it is recruited to nascent phagosomes. Syt VII recruitment is rapidly followed by the delivery of Lamp1 to phagosomes, a process that is inhibited in Syt VII-/- macrophages. Thus, Syt VII regulates the Ca2+-dependent mobilization of lysosomes as a supplemental source of membrane during phagocytosis.


Asunto(s)
Calcio/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Macrófagos/metabolismo , Fagocitosis , Fagosomas/metabolismo , Sinaptotagminas/metabolismo , Animales , Membrana Celular/química , Membrana Celular/metabolismo , Células Cultivadas , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Sinaptotagminas/genética
15.
J Am Soc Mass Spectrom ; 28(12): 2716-2723, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28924631

RESUMEN

Brucellaceae are Gram-negative bacteria that cause brucellosis, one of the most distributed worldwide zoonosis, transmitted to humans by contact with either infected animals or their products. The lipopolysaccharide exposed on the cell surface has been intensively studied and is considered a major virulence factor of Brucella. In the last years, structural studies allowed the determination of new structures in the core oligosaccharide and the O-antigen of this lipopolysaccharide. In this work, we have reinvestigated the lipid A structure isolated from B. suis and B. abortus lipopolysaccharides. A detailed study by MALDI-TOF mass spectrometry in the positive and negative ion modes of the lipid A moieties purified from both species was performed. Interestingly, a new feature was detected: the presence of a pyrophosphorylethanolamine residue substituting the backbone. LID-MS/MS analysis of some of the detected ions allowed assurance that the Lipid A structure composed by the diGlcN3N disaccharide, mainly hexa-acylated and penta-acylated, bearing one phosphate and one pyrophosphorylethanolamine residue. Graphical abstract ᅟ.


Asunto(s)
Brucella abortus/química , Brucella suis/química , Lípido A/química , Acilación , Brucelosis/microbiología , Disacáridos/análisis , Etanolaminas/análisis , Humanos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem
16.
Vaccine ; 34(13): 1524-1530, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26899373

RESUMEN

Brucellosis is one of the most widespread zoonosis in the world affecting many domestic and wild animals including bovines, goats, pigs and dogs. Each species of the Brucella genus has a particular tropism toward different mammals being the most relevant for human health Brucella abortus, Brucella melitensis and Brucella suis that infect bovines, goats/camelids and swine respectively. Although for B. abortus and B. melitensis there are vaccines available, there is no efficient vaccine to protect swine from B. suis infection so far. We describe here the construction of a novel vaccine strain that confers excellent protection against B. suis in a mouse model of infection. This strain is a clean deletion of the phosphoglucomutase (pgm) gene that codes for a protein that catalyzes the conversion of glucose-6-P to glucose-1-P, which is used as a precursor for the biosynthesis of many polysaccharides. The Delta-pgm strain lacks a complete lipopolysaccharide, is unable to synthesize cyclic beta glucans and is sensitive to several detergents and Polymyxin B. We show that this strain replicates in cultured cells, is completely avirulent in the mouse model of infection but protects against a challenge of the virulent strain inducing the production of pro-inflammatory cytokines. This novel strain could be an excellent candidate for the control of swine brucellosis, a disease of emerging concern in many parts of the world.


Asunto(s)
Vacuna contra la Brucelosis/inmunología , Brucella suis/genética , Brucelosis/prevención & control , Eliminación de Gen , Fosfoglucomutasa/genética , Animales , Anticuerpos Antibacterianos/sangre , Brucella suis/enzimología , Línea Celular , Femenino , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Vacunas Atenuadas/inmunología , beta-Glucanos/química
17.
Microbes Infect ; 14(1): 79-85, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21911075

RESUMEN

Brucella, the causative agent of brucellosis, a major zoonotic disease affecting a broad range of mammals, is a gram-negative bacterium whose virulence is dependent on the capacity to attach and invade different cells of the host. The bacterium is able to infect through a diverse repertoire of epitheliums: skin, airways or gastric. Although much has been studied on the mechanisms Brucella uses to establish an intracellular replication niche, almost none is known on how the bacterium adheres and invades host cells. We report here the identification of a pathogenicity island that harbors a gene homologous to proteins with bacterial immunoglobulin-like domains present in other pathogens that play a role in attachment and invasion. Deletion of the entire island results in a mutant with a reduced attachment capacity measured by intracellular replication and adhesion assays. Intraperitoneal and oral experimental infection of mice strongly suggests that this island plays a role during the oral infection probably mediating attachment and trespassing of the gastric epithelium to establish a systemic infection.


Asunto(s)
Adhesión Bacteriana/genética , Brucella/genética , Brucelosis/microbiología , Genes Bacterianos , Familia de Multigenes , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Brucella/patogenicidad , Línea Celular , Femenino , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Virulencia
18.
J Cell Biol ; 191(3): 599-613, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21041449

RESUMEN

Syt VII is a Ca(2+) sensor that regulates lysosome exocytosis and plasma membrane repair. Because it lacks motifs that mediate lysosomal targeting, it is unclear how Syt VII traffics to these organelles. In this paper, we show that mutations or inhibitors that abolish palmitoylation disrupt Syt VII targeting to lysosomes, causing its retention in the Golgi complex. In macrophages, Syt VII is translocated simultaneously with the lysosomal tetraspanin CD63 from tubular lysosomes to nascent phagosomes in a Ca(2+)-dependent process that facilitates particle uptake. Mutations in Syt VII palmitoylation sites block trafficking of Syt VII, but not CD63, to lysosomes and phagosomes, whereas tyrosine replacement in the lysosomal targeting motif of CD63 causes both proteins to accumulate on the plasma membrane. Complexes of CD63 and Syt VII are detected only when Syt VII palmitoylation sites are intact. These findings identify palmitoylation-dependent association with the tetraspanin CD63 as the mechanism by which Syt VII is targeted to lysosomes.


Asunto(s)
Antígenos CD/metabolismo , Calcio/metabolismo , Lisosomas/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Sinaptotagminas/metabolismo , Animales , Células Cultivadas , Lipoilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sinaptotagminas/deficiencia , Sinaptotagminas/genética , Tetraspanina 30
19.
J Gen Virol ; 86(Pt 8): 2275-2280, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16033975

RESUMEN

Triatoma virus (TrV) belongs to a new family of RNA viruses known as Dicistroviridae. Nucleotide sequence comparisons between different dicistroviruses allowed two putative internal ribosomal entry sites (IRESs) in the TrV RNA to be defined: the 5'UTR IRES of 548 nt and the intergenic region (IGR) IRES of 172 nt. Using monocistronic and bicistronic RNAs, it was shown that the TrV genome contains two functional IRESs that mediate translation initiation in a cap-independent manner. In addition, it was found that the two TrV IRESs were able to direct efficient translation of reporter genes in microinjected Xenopus oocytes, suggesting minimum requirements for host factors. The IGR IRES begins with a non-canonical CUC; however, mutations of this triplet to AUG or CCU did not impair IRES function, indicating that the CUC is not essential for the initiation process. Furthermore, translation efficiency from two TrV IRESs was differentially modulated by IFN-alpha and viral infection.


Asunto(s)
Biosíntesis de Proteínas , Virus ARN/metabolismo , ARN Viral/metabolismo , Triatoma/virología , Regiones no Traducidas 5'/fisiología , Animales , Línea Celular , Cricetinae , Culicidae , ADN Intergénico/fisiología , Genes Reporteros/fisiología , Interferón-alfa/farmacología , Luciferasas/metabolismo , Ribosomas/metabolismo , Ribosomas/virología
20.
Microb Pathog ; 37(2): 95-105, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15312849

RESUMEN

Secreted as well as surface exposed proteins are assumed to play major roles in bacterial virulence. In this report we describe the construction of an N-terminal protein-capturing system and its use for the isolation of Brucella abortus S2308 genes coding for putative surface exposed or secreted proteins. For this purpose, a cloning vector that generates gene fusions to a ribosome binding site and start codon deficient Chloramphenicol Acetyl Transferase (CAT) reporter gene was constructed and the resulting library introduced into B. abortus S2308 and virB mutant strains. Secreted translational fusions were identified by determining CAT activity in culture supernatants. Secretion was confirmed by Western Blot using a polyclonal anti-CAT antibody. A total of 864 clones were screened and 10 genes encoding putative secreted/surface exposed proteins were identified. Seven are Brucella proteins with an assigned function, whereas three are hypothetical proteins. The number of amino acid residues that promotes CAT secretion varies from 5 to 386 and no conserved motifs were detected. Secretion in a virB mutant background of some of the isolated fusion proteins was also determined. Interestingly, some hybrid proteins seemed to require a full VirB system for their secretion.


Asunto(s)
Proteínas Bacterianas/genética , Brucella abortus/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Proteínas Bacterianas/metabolismo , Brucella abortus/enzimología , Brucella abortus/metabolismo , Brucella abortus/patogenicidad , Clonación Molecular , Vectores Genéticos , Mutagénesis Insercional , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA