Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Virol ; 96(4): e0200521, 2022 02 23.
Article in English | MEDLINE | ID: mdl-34878889

ABSTRACT

Birnaviruses are members of the Birnaviridae family, responsible for major economic losses to poultry and aquaculture. The family is composed of nonenveloped viruses with a segmented double-stranded RNA (dsRNA) genome. Infectious bursal disease virus (IBDV), the prototypic family member, is the etiological agent of Gumboro disease, a highly contagious immunosuppressive disease in the poultry industry worldwide. We previously demonstrated that IBDV hijacks the endocytic pathway for establishing the viral replication complexes on endosomes associated with the Golgi complex (GC). Here, we report that IBDV reorganizes the GC to localize the endosome-associated replication complexes without affecting its secretory functionality. By analyzing crucial proteins involved in the secretory pathway, we showed the essential requirement of Rab1b for viral replication. Rab1b comprises a key regulator of GC transport and we demonstrate that transfecting the negative mutant Rab1b N121I or knocking down Rab1b expression by RNA interference significantly reduces the yield of infectious viral progeny. Furthermore, we showed that the Rab1b downstream effector Golgi-specific BFA resistance factor 1 (GBF1), which activates the small GTPase ADP ribosylation factor 1 (ARF1), is required for IBDV replication, since inhibiting its activity by treatment with brefeldin A (BFA) or golgicide A (GCA) significantly reduces the yield of infectious viral progeny. Finally, we show that ARF1 dominant negative mutant T31N overexpression hampered IBDV infection. Taken together, these results demonstrate that IBDV requires the function of the Rab1b-GBF1-ARF1 axis to promote its replication, making a substantial contribution to the field of birnavirus-host cell interactions. IMPORTANCE Birnaviruses are unconventional members of the dsRNA viruses, with the lack of a transcriptionally active core being the main differential feature. This structural trait, among others that resemble those of the plus single-stranded (+ssRNA) viruses features, suggests that birnaviruses might follow a different replication program from that conducted by prototypical dsRNA members and the hypothesis that birnaviruses could be evolutionary links between +ssRNA and dsRNA viruses has been argued. Here, we present original data showing that IBDV-induced GC reorganization and the cross talk between IBDV and the Rab1b-GBF1-ARF1 mediate the intracellular trafficking pathway. The replication of several +ssRNA viruses depends on the cellular protein GBF1, but its role in the replication process is not clear. Thus, our findings make a substantial contribution to the field of birnavirus-host cell interactions and provide further evidence supporting the proposed evolutionary connection role of birnaviruses, an aspect which we consider especially relevant for researchers working in the virology field.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Infectious bursal disease virus/physiology , Secretory Pathway/physiology , Virus Replication/physiology , rab1 GTP-Binding Proteins/metabolism , ADP-Ribosylation Factor 1/genetics , Animals , Brefeldin A/pharmacology , Cell Line , Endosomes/metabolism , Golgi Apparatus/metabolism , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Host-Pathogen Interactions , Pyridines/pharmacology , Quinolines/pharmacology , Secretory Pathway/drug effects , Viral Replication Compartments/metabolism , Virus Replication/drug effects , rab1 GTP-Binding Proteins/genetics
2.
BMC Public Health ; 20(1): 1809, 2020 Nov 27.
Article in English | MEDLINE | ID: mdl-33246432

ABSTRACT

BACKGROUND: Mathematical modelling of infectious diseases is a powerful tool for the design of management policies and a fundamental part of the arsenal currently deployed to deal with the COVID-19 pandemic. METHODS: We present a compartmental model for the disease where symptomatic and asymptomatic individuals move separately. We introduced healthcare burden parameters allowing to infer possible containment and suppression strategies. In addition, the model was scaled up to describe different interconnected areas, giving the possibility to trigger regionalized measures. It was specially adjusted to Mendoza-Argentina's parameters, but is easily adaptable for elsewhere. RESULTS: Overall, the simulations we carried out were notably more effective when mitigation measures were not relaxed in between the suppressive actions. Since asymptomatics or very mildly affected patients are the vast majority, we studied the impact of detecting and isolating them. The removal of asymptomatics from the infectious pool remarkably lowered the effective reproduction number, healthcare burden and overall fatality. Furthermore, different suppression triggers regarding ICU occupancy were attempted. The best scenario was found to be the combination of ICU occupancy triggers (on: 50%, off: 30%) with the detection and isolation of asymptomatic individuals. In the ideal assumption that 45% of the asymptomatics could be detected and isolated, there would be no need for complete lockdown, and Mendoza's healthcare system would not collapse. CONCLUSIONS: Our model and its analysis inform that the detection and isolation of all infected individuals, without leaving aside the asymptomatic group is the key to surpass this pandemic.


Subject(s)
Asymptomatic Infections/epidemiology , Coronavirus Infections/diagnosis , Coronavirus Infections/prevention & control , Epidemics/prevention & control , Pandemics/prevention & control , Patient Isolation , Pneumonia, Viral/diagnosis , Pneumonia, Viral/prevention & control , Argentina/epidemiology , COVID-19 , Coronavirus Infections/epidemiology , Humans , Models, Theoretical , Pneumonia, Viral/epidemiology
3.
Article in English | MEDLINE | ID: mdl-33042861

ABSTRACT

Human Cytomegalovirus (HCMV) is a frequent opportunistic pathogen in immunosuppressed patients, which can be involved in kidney allograft dysfunction and rejection. In order to study the pathophysiology of HCMV renal diseases, we concentrated on the impact of HCMV infection on human renal tubular epithelial HK-2 cells. Our aim was to develop a model of infection of HK-2 cells by using the viral strain TB40/E, that contains the extended cell tropism of clinical isolates and the efficient viral multiplication in cell culture of laboratory-adapted strains. We observed that HK-2 cells can be infected by HCMV and expressed viral antigens, but they do not produce extracellular viral particles. We then studied the interplay of HCMV with ciliogenesis and autophagy. Primary cilium (PC) is a stress sensor important to maintain renal tissue homeostasis that projects from the apical side into the lumen of tubule cells. PC formation and length were not modified by HCMV infection. Autophagy, another stress response process critically required for normal kidney functions, was inhibited by HCMV in HK-2 cells with a reduction in the autophagic flux. HCMV classically induces an enlargement of infected cells in vivo and in vitro, and we observed that HCMV infection led to an enlargement of the HK-2 cell volume. Our results constitute therefore an excellent starting point to further explore the role of these mechanisms in renal cells dysfunction.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Autophagy , Cells, Cultured , Epithelial Cells , Humans
4.
Cell Mol Life Sci ; 76(7): 1397-1417, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30673822

ABSTRACT

Mitochondrial dysfunction represents an important cellular stressor and when intense and persistent cells must unleash an adaptive response to prevent their extinction. Furthermore, mitochondria can induce nuclear transcriptional changes and DNA methylation can modulate cellular responses to stress. We hypothesized that mitochondrial dysfunction could trigger an epigenetically mediated adaptive response through a distinct DNA methylation patterning. We studied cellular stress responses (i.e., apoptosis and autophagy) in mitochondrial dysfunction models. In addition, we explored nuclear DNA methylation in response to this stressor and its relevance in cell survival. Experiments in cultured human myoblasts revealed that intense mitochondrial dysfunction triggered a methylation-dependent pro-survival response. Assays done on mitochondrial disease patient tissues showed increased autophagy and enhanced DNA methylation of tumor suppressor genes and pathways involved in cell survival regulation. In conclusion, mitochondrial dysfunction leads to a "pro-survival" adaptive state that seems to be triggered by the differential methylation of nuclear genes.


Subject(s)
Cell Nucleus/genetics , Epigenesis, Genetic , Mitochondria/metabolism , Adolescent , Autophagy/drug effects , Case-Control Studies , Cell Nucleus/metabolism , Cell Shape/drug effects , Cell Survival/drug effects , Cells, Cultured , Child , Child, Preschool , DNA Methylation , Epigenesis, Genetic/drug effects , Female , Humans , Male , Mitochondria/drug effects , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Rotenone/pharmacology
5.
Infect Immun ; 81(6): 1940-51, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23509146

ABSTRACT

Arthritis is one of the most common complications of human active brucellosis, but its pathogenic mechanisms have not been completely elucidated. In this paper, we describe the role of synoviocytes in the pathogenesis of brucellar arthritis. Our results indicate that Brucella abortus infection inhibited synoviocyte apoptosis through the upregulation of antiapoptotic factors (cIAP-2, clusterin, livin, and P21/CIP/CDNK1A). In contrast, infection did not change the expression of proteins that have been involved in apoptosis induction such as Bad, Bax, cleaved procaspase 3, CytC, and TRAIL, among others; or their expression was reduced, as occurs in the case of P-p53(S15). In addition, B. abortus infection induced upregulation of adhesion molecules (CD54 and CD106), and the adhesion of monocytes and neutrophils to infected synoviocytes was significantly higher than to uninfected cells. Despite this increased adhesion, B. abortus-infected synoviocytes were able to inhibit apoptosis induced by supernatants from B. abortus-infected monocytes and neutrophils. Moreover, B. abortus infection increased soluble and membrane RANKL expression in synoviocytes that further induced monocytes to undergo osteoclastogenesis. The results presented here shed light on how the interactions of B. abortus with synovial fibroblasts may have an important role in the pathogenesis of brucellar arthritis.


Subject(s)
Apoptosis/physiology , Brucella abortus/physiology , Fibroblasts/microbiology , Gene Expression Regulation, Bacterial/physiology , RANK Ligand/metabolism , Synovial Membrane/cytology , Antigens, CD/metabolism , Bone Resorption/metabolism , Cell Adhesion , Cells, Cultured , Fibroblasts/cytology , Humans , Osteoclasts/metabolism , Osteoclasts/microbiology , RANK Ligand/genetics , Up-Regulation
6.
PLoS One ; 6(1): e16203, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21264260

ABSTRACT

As Brucella infections occur mainly through mucosal surfaces, the development of mucosal administered vaccines could be radical for the control of brucellosis. In this work we evaluated the potential of Brucella abortus 19 kDa outer membrane protein (U-Omp19) as an edible subunit vaccine against brucellosis. We investigated the protective immune response elicited against oral B. abortus infection after vaccination of mice with leaves from transgenic plants expressing U-Omp19; or with plant-made or E. coli-made purified U-Omp19. All tested U-Omp19 formulations induced protection against Brucella when orally administered without the need of adjuvants. U-Omp19 also induced protection against a systemic challenge when parenterally administered. This built-in adjuvant ability of U-Omp19 was independent of TLR4 and could be explained at least in part by its capability to activate dendritic cells in vivo. While unadjuvanted U-Omp19 intraperitoneally administered induced a specific Th1 response, following U-Omp19 oral delivery a mixed specific Th1-Th17 response was induced. Depletion of CD4(+) T cells in mice orally vaccinated with U-Omp19 resulted in a loss of the elicited protection, indicating that this cell type mediates immune protection. The role of IL-17 against Brucella infection has never been explored. In this study, we determined that if IL-17A was neutralized in vivo during the challenge period, the mucosal U-Omp19 vaccine did not confer mucosal protection. On the contrary, IL-17A neutralization during the infection did not influence at all the subsistence and growth of this bacterium in PBS-immunized mice. All together, our results indicate that an oral unadjuvanted vaccine based on U-Omp19 induces protection against a mucosal challenge with Brucella abortus by inducing an adaptive IL-17 immune response. They also indicate different and important new aspects i) IL-17 does not contribute to reduce the bacterial burden in non vaccinated mice and ii) IL-17 plays a central role in vaccine mediated anti-Brucella mucosal immunity.


Subject(s)
Antigens, Bacterial/therapeutic use , Bacterial Outer Membrane Proteins/therapeutic use , Brucella Vaccine/immunology , Brucella abortus/immunology , Interleukin-17/immunology , Lipoproteins/therapeutic use , Adaptive Immunity , Animals , Brucella Vaccine/administration & dosage , Brucellosis/prevention & control , CD4-Positive T-Lymphocytes/immunology , Mice , Th1 Cells/immunology , Th17 Cells/immunology
7.
Microbes Infect ; 13(3): 239-50, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21070860

ABSTRACT

The strategies that allow Brucella abortus to persist for years inside macrophages subverting host immune responses are not completely understood. Immunity against this bacterium relies on the capacity of IFN-γ to activate macrophages, endowing them with the ability to destroy intracellular bacteria. We report here that infection with B. abortus down-modulates the expression of the type I receptor for the Fc portion of IgG (FcγRI, CD64) and FcγRI-restricted phagocytosis regulated by IFN-γ in human monocytes/macrophages. Both phenomena were not dependent on bacterial viability, since they were also induced by heat-killed B. abortus (HKBA), suggesting that they were elicited by a structural bacterial component. Accordingly, a prototypical B. abortus lipoprotein (L-Omp19), but not its unlipidated form, inhibited both CD64 expression and FcγRI-restricted phagocytosis regulated by IFN-γ. Moreover, a synthetic lipohexapeptide that mimics the structure of the protein lipid moiety also inhibited CD64 expression, indicating that any Brucella lipoprotein could down-modulate CD64 expression and FcγRI-restricted phagocytosis. Pre-incubation of monocytes/macrophages with anti-TLR2 mAb blocked the inhibition of the CD64 expression mediated by HKBA and L-Omp19. These results, together with our previous observations establish that B. abortus utilizes its lipoproteins to inhibit the monocytes/macrophages activation mediated by IFN-γ and to subvert host immunonological responses.


Subject(s)
Brucella abortus/physiology , Interferon-gamma/antagonists & inhibitors , Macrophages/immunology , Monocytes/immunology , Receptors, IgG/biosynthesis , Toll-Like Receptor 2/metabolism , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Brucella abortus/chemistry , Brucella abortus/metabolism , Cells, Cultured , Flow Cytometry , Host-Pathogen Interactions , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lipoproteins/metabolism , Macrophages/microbiology , Monocytes/microbiology , Phagocytosis/physiology , Receptors, IgG/antagonists & inhibitors , Receptors, IgG/metabolism , Statistics, Nonparametric
8.
J Immunol ; 184(9): 5200-12, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20351187

ABSTRACT

Knowing the inherent stimulatory properties of the lipid moiety of bacterial lipoproteins, we first hypothesized that Brucella abortus outer membrane protein (Omp)16 lipoprotein would be able to elicit a protective immune response without the need of external adjuvants. In this study, we demonstrate that Omp16 administered by the i.p. route confers significant protection against B. abortus infection and that the protective response evoked is independent of the protein lipidation. To date, Omp16 is the first Brucella protein that without the requirement of external adjuvants is able to induce similar protection levels to the control live vaccine S19. Moreover, the protein portion of Omp16 (unlipidated Omp16 [U-Omp16]) elicits a protective response when administered by the oral route. Either systemic or oral immunization with U-Omp16 elicits a Th1-specific response. These abilities of U-Omp16 indicate that it is endowed with self-adjuvanting properties. The adjuvanticity of U-Omp16 could be explained, at least in part, by its capacity to activate dendritic cells in vivo. U-Omp16 is also able to stimulate dendritic cells and macrophages in vitro. The latter property and its ability to induce a protective Th1 immune response against B. abortus infection have been found to be TLR4 dependent. The facts that U-Omp16 is an oral protective Ag and possesses a mucosal self-adjuvanting property led us to develop a plant-made vaccine expressing U-Omp16. Our results indicate that plant-expressed recombinant U-Omp16 is able to confer protective immunity, when given orally, indicating that a plant-based oral vaccine expressing U-Omp16 could be a valuable approach to controlling this disease.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Brucella Vaccine/immunology , Brucellosis/prevention & control , Dendritic Cells/immunology , Host-Pathogen Interactions/immunology , Th1 Cells/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Administration, Oral , Animals , Antigens, Bacterial/administration & dosage , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/biosynthesis , Bacterial Outer Membrane Proteins/genetics , Brucella Vaccine/administration & dosage , Brucellosis/immunology , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Freund's Adjuvant/administration & dosage , Host-Pathogen Interactions/genetics , Immunity, Cellular , Injections, Intraperitoneal , Lipids/administration & dosage , Lipoproteins/administration & dosage , Lipoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells/microbiology , Nicotiana/genetics , Nicotiana/immunology
9.
Am J Pathol ; 176(3): 1323-38, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20093491

ABSTRACT

Central nervous system (CNS) invasion by bacteria of the genus Brucella results in an inflammatory disorder called neurobrucellosis. In this study we present in vivo and in vitro evidence that B. abortus and its lipoproteins activate the innate immunity of the CNS, eliciting an inflammatory response that leads to astrogliosis, a characteristic feature of neurobrucellosis. Intracranial injection of heat-killed B. abortus (HKBA) or outer membrane protein 19 (Omp19), a B. abortus lipoprotein model, induced astrogliosis in mouse striatum. Moreover, infection of astrocytes and microglia with B. abortus induced the secretion of interleukin (IL)-6, IL-1beta, tumor necrosis factor (TNF)-alpha, macrophage chemoattractant protein-1, and KC (CXCL1). HKBA also induced these inflammatory mediators, suggesting the involvement of a structural component of the bacterium. Accordingly, Omp19 induced the same cytokine and chemokine secretion pattern. B. abortus infection induced astrocyte, but not microglia, apoptosis. Indeed, HKBA and Omp19 elicited not only astrocyte apoptosis but also proliferation, two features observed during astrogliosis. Apoptosis induced by HKBA and L-Omp19 was completely suppressed in cells of TNF receptor p55-/- mice or when the general caspase inhibitor Z-VAD-FMK was added to cultures. Hence, TNF-alpha signaling via TNF receptor (TNFR) 1 through the coupling of caspases determines apoptosis. Our results provide proof of the principle that Brucella lipoproteins could be key virulence factors in neurobrucellosis and that astrogliosis might contribute to neurobrucellosis pathogenesis.


Subject(s)
Apoptosis , Astrocytes/microbiology , Astrocytes/pathology , Brucella abortus/physiology , Inflammation Mediators/metabolism , Animals , Antigens, Bacterial/pharmacology , Apoptosis/drug effects , Astrocytes/enzymology , Astrocytes/metabolism , Bacterial Outer Membrane Proteins/pharmacology , Brain/drug effects , Brain/microbiology , Brain/pathology , Brucella abortus/drug effects , Caspases/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chemokines/metabolism , Female , Hot Temperature , Immunohistochemistry , Lipopolysaccharides/pharmacology , Lipoproteins/pharmacology , Mice , Mice, Inbred BALB C , Microglia/drug effects , Microglia/microbiology , Microglia/pathology , Tumor Necrosis Factor-alpha/metabolism
10.
Microbes Infect ; 11(6-7): 689-97, 2009.
Article in English | MEDLINE | ID: mdl-19376263

ABSTRACT

Human brucellosis is caused by infection with certain species of the genus Brucella and is characterized by bacterial persistence and inflammation of many host tissues. Neutrophils are one of the predominant cell types present in the infiltrate of these inflamed tissues, and due to their potential effect on the inflammatory response and tissue damage, direct activation of neutrophils by Brucella abortus might contribute to the pathology associated with human brucellosis. B. abortus expresses outer membrane lipoproteins (Omp) with inflammatory properties on a variety of cell types. This study examines the effect of B. abortus and its lipoproteins on neutrophil functions. B. abortus induced an increment in CD35 and CD11b expression and a decline in CD62L accompanied by IL-8 secretion, a response compatible with neutrophil activation. B. abortus lipoprotein Omp19 (L-Omp19), but not its unlipidated form, mimicked the changes associated with neutrophil activation induced by B. abortus. L-Omp19 primed neutrophils for oxidative burst as well as promoted neutrophil migration and prolonged neutrophil survival. Thus, Brucella lipoproteins possess pro-inflammatory properties that could contribute to the localize tissue injury and inflammation by direct activation of neutrophils. Data presented here, together with our previous results implicate Brucella lipoproteins in the pathogenesis of human brucellosis.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Brucella abortus/immunology , Lipoproteins/immunology , Neutrophils/immunology , CD11b Antigen/analysis , Cell Migration Assays, Leukocyte , Cell Movement , Cell Survival , Cells, Cultured , Humans , Interleukin-8/metabolism , L-Selectin/analysis , Neutrophils/chemistry , Receptors, Complement 3b/analysis , Respiratory Burst
11.
Infect Immun ; 77(1): 436-45, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18981242

ABSTRACT

Available vaccines against Brucella spp. are live attenuated Brucella strains. In order to engineer a better vaccine to be used in animals and humans, our laboratory aims to develop an innocuous subunit vaccine. Particularly, we are interested in the outer membrane proteins (OMPs) of B. abortus: Omp16 and Omp19. In this study, we assessed the use of these proteins as vaccines against Brucella in BALB/c mice. Immunization with lipidated Omp16 (L-Omp16) or L-Omp19 in incomplete Freund's adjuvant (IFA) conferred significant protection against B. abortus infection. Vaccination with unlipidated Omp16 (U-Omp16) or U-Omp19 in IFA induced a higher degree of protection than the respective lipidated versions. Moreover, the level of protection induced after U-Omp16 or U-Omp19 immunization in IFA was similar to that elicited by live B. abortus S19 immunization. Flow cytometric analysis showed that immunization with U-Omp16 or U-Omp19 induced antigen-specific CD4(+) as well as CD8(+) T cells producing gamma interferon. In vivo depletion of CD4(+) or CD8(+) T cells in mice immunized with U-Omp16 or U-Omp19 plus IFA resulted in a loss of the elicited protection, indicating that both cell types are mediating immune protection. U-Omp16 or U-Omp19 vaccination induced a T helper 1 response, systemic protection in aluminum hydroxide formulation, and oral protection with cholera toxin adjuvant against B. abortus infection. Both immunization routes exhibited a similar degree of protection to attenuated Brucella vaccines (S19 and RB51, respectively). Overall these results indicate that U-Omp16 or U-Omp19 would be a useful candidate for a subunit vaccine against human and animal brucellosis.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Outer Membrane Proteins/immunology , Brucella Vaccine/immunology , Brucella abortus/immunology , Brucellosis/prevention & control , Freund's Adjuvant/administration & dosage , Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/pharmacology , Animals , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/administration & dosage , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cholera Toxin/administration & dosage , Cholera Toxin/pharmacology , Colony Count, Microbial , Female , Flow Cytometry , Freund's Adjuvant/pharmacology , Immunoglobulin G/blood , Interferon-gamma/biosynthesis , Leukocyte Reduction Procedures , Mice , Mice, Inbred BALB C , Spleen/microbiology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
12.
Microbes Infect ; 10(12-13): 1346-54, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18761420

ABSTRACT

Infection with Brucella abortus induces a pro-inflammatory response that drives T cell responses toward a Th1 profile. The mechanism by which this bacterium triggers this response is unknown. Dendritic cells (DC) are crucial mediators at the host-pathogen interface and are potent Th1-inducing antigen-presenting cells. Thus, we examined the mechanism whereby B. abortus stimulate human DC maturation. B. abortus-infected DC increased the expression of CD86, CD80, CCR7, CD83, MHCII, MHCI and CD40 and induced the production of TNF-alpha, IL-6, IL-10 and IL-12. Both phenomena were not dependent on bacterial viability since they were also induced by heat-killed B. abortus (HKBA). B. abortus LPS was unable to induce markers up-regulation or cytokine production. We next investigated the capacity of the outer membrane protein 19 (Omp19) as a B. abortus lipoprotein model to induce DC maturation. Lipidated Omp19 (L-Omp19), but not its unlipidated form, increased the expression of cell surface markers and the secretion of cytokines. L-Omp19-matured DC also have decreased endocytic activity and displayed enhanced T cell stimulatory activity in a MLR. Pre-incubation of DC with anti-TLR2 mAb blocked L-Omp19-mediated cytokine production. These results demonstrate that B. abortus lipoproteins can stimulate DC maturation providing a mechanism by which these bacteria generate a Th1-type immune response.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Brucella abortus/pathogenicity , Cell Differentiation , Dendritic Cells , Lipoproteins/immunology , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Flow Cytometry , Humans , Receptors, Cell Surface/metabolism
13.
Infect Immun ; 76(1): 250-62, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17984211

ABSTRACT

The strategies that allow Brucella abortus to survive inside macrophages for prolonged periods and to avoid the immunological surveillance of major histocompatibility complex class II (MHC-II)-restricted gamma interferon (IFN-gamma)-producing CD4+ T lymphocytes are poorly understood. We report here that infection of THP-1 cells with B. abortus inhibited expression of MHC-II molecules and antigen (Ag) processing. Heat-killed B. abortus (HKBA) also induced both these phenomena, indicating the independence of bacterial viability and involvement of a structural component of the bacterium. Accordingly, outer membrane protein 19 (Omp19), a prototypical B. abortus lipoprotein, inhibited both MHC-II expression and Ag processing to the same extent as HKBA. Moreover, a synthetic lipohexapeptide that mimics the structure of the protein lipid moiety also inhibited MHC-II expression, indicating that any Brucella lipoprotein could down-modulate MHC-II expression and Ag processing. Inhibition of MHC-II expression and Ag processing by either HKBA or lipidated Omp19 (L-Omp19) depended on Toll-like receptor 2 and was mediated by interleukin-6. HKBA or L-Omp19 also inhibited MHC-II expression and Ag processing of human monocytes. In addition, exposure to the synthetic lipohexapeptide inhibited Ag-specific T-cell proliferation and IFN-gamma production of peripheral blood mononuclear cells from Brucella-infected patients. Together, these results indicate that there is a mechanism by which B. abortus may prevent recognition by T cells to evade host immunity and establish a chronic infection.


Subject(s)
Antigen Presentation , Brucella abortus/physiology , Gene Expression Regulation , Genes, MHC Class II/genetics , Interleukin-6/metabolism , Toll-Like Receptor 2/metabolism , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Brucella abortus/immunology , Cells, Cultured , Down-Regulation , Humans , Interferon-gamma , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/microbiology , Lipoproteins/metabolism , Macrophages/metabolism , Macrophages/microbiology , Monocytes/metabolism , Monocytes/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL