Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 59
1.
PLoS One ; 12(10): e0186561, 2017.
Article En | MEDLINE | ID: mdl-29036184

Brucella canis infection is an important cause of late-term abortion in pregnant bitches. The pathophysiological mechanisms leading to B. canis-induced abortion are unknown, but heavily infected trophoblasts are consistently observed. As trophoblasts responses to other pathogens contribute to placental inflammation leading to abortion, the aim of the present study was to characterize the cytokine response of canine trophoblasts to B. canis infection. To achieve this, trophoblasts isolated from term placenta of healthy female dogs were infected with B. canis, culture supernatants were harvested for cytokine determinations, and the load of intracellular viable B. canis was determined at different times post-infection. Additionally, cytokine responses were assessed in non-infected trophoblasts stimulated with conditioned media (CM) from B. canis-infected canine monocytes and neutrophils. Finally, cytokine response and bacteria replication were assessed in canine placental explants infected ex vivo. B. canis successfully infected and replicated in primary canine trophoblasts, eliciting an increase in IL-8 and RANTES (CCL5) secretion. Moreover, the stimulation of trophoblasts with CM from B. canis-infected monocytes and neutrophils induced a significant increase in IL-8, IL-6 and RANTES secretion. B. canis replication was confirmed in infected placental explants and the infection elicited an increased secretion of TNF-α, IL-8, IL-6 and RANTES. This study shows that canine trophoblasts produce proinflammatory cytokines in response to B. canis infection and/or to stimulation with factors produced by infected monocytes and neutrophils. These cytokines may contribute to placental inflammation leading to abortion in B. canis-infected pregnant bitches.


Brucella canis/physiology , Trophoblasts/microbiology , Animals , Antigens, Bacterial/immunology , Brucella canis/immunology , Chemokines/metabolism , Dogs , Female , Inflammation/microbiology , Phagocytes/cytology , Placenta/microbiology , Pregnancy , Toll-Like Receptors/agonists , Trophoblasts/cytology , Trophoblasts/metabolism
2.
Biol Reprod ; 94(2): 48, 2016 Feb.
Article En | MEDLINE | ID: mdl-26792938

Trophoblasts are targets of infection by Brucella spp. but their role in the pathophysiology of pregnancy complications of brucellosis is unknown. Here we show that Brucella abortus invades and replicates in the human trophoblastic cell line Swan-71 and that the intracellular survival of the bacterium depends on a functional virB operon. The infection elicited significant increments of interleukin 8 (IL8), monocyte chemotactic protein 1 (MCP-1), and IL6 secretion, but levels of IL1beta and tumor necrosis factor-alpha (TNF-alpha) did not vary significantly. Such proinflammatory response was not modified by the absence of the Brucella TIR domain-containing proteins BtpA and BtpB. The stimulation of Swan-71 cells with conditioned medium (CM) from B. abortus-infected human monocytes (THP-1 cells) or macrophages induced a significant increase of IL8, MCP-1 and IL6 as compared to stimulation with CM from non-infected cells. Similar results were obtained when stimulation was performed with CM from infected neutrophils. Neutralization studies showed that IL1beta and/or TNF-alpha mediated the stimulating effects of CM from infected phagocytes. Reciprocally, stimulation of monocytes and neutrophils with CM from Brucella-infected trophoblasts increased IL8 and/or IL6 secretion. These results suggest that human trophoblasts may provide a local inflammatory environment during B. abortus infections either through a direct response to the pathogen or through interactions with monocytes/macrophages or neutrophils, potentially contributing to the pregnancy complications of brucellosis.


Brucella abortus , Brucellosis/pathology , Inflammation/microbiology , Phagocytes/microbiology , Trophoblasts/microbiology , Brucellosis/metabolism , Cell Line , Chemokine CCL2/metabolism , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Monocytes/metabolism , Monocytes/microbiology , Monocytes/pathology , Phagocytes/metabolism , Phagocytes/pathology , Trophoblasts/metabolism , Trophoblasts/pathology , Tumor Necrosis Factor-alpha/metabolism
3.
PLoS One ; 10(10): e0140408, 2015.
Article En | MEDLINE | ID: mdl-26448160

Both CCL20 and human ß-defensin 2 (hBD2) interact with the same membrane receptor and display chemotactic and antimicrobial activities. They are produced by airway epithelia in response to infectious agents and proinflammatory cytokines. Whereas Brucella spp. can infect humans through inhalation, their ability to induce CCL20 and hBD2 in lung cells is unknown. Here we show that B. abortus induces CCL20 expression in human alveolar (A549) or bronchial (Calu-6) epithelial cell lines, primary alveolar epithelial cells, primary human monocytes, monocyte-derived macrophages and the monocytic cell line THP-1. CCL20 expression was mainly mediated by JNK1/2 and NF-kB in both Calu-6 and THP-1 cells. CCL20 secretion was markedly induced in A549, Calu-6 and THP-1 cells by heat-killed B. abortus or a model Brucella lipoprotein (L-Omp19) but not by the B. abortus lipopolysaccharide (LPS). Accordingly, CCL20 production by B. abortus-infected cells was strongly TLR2-dependent. Whereas hBD2 expression was not induced by B. abortus infection, it was significantly induced in A549 cells by conditioned media from B. abortus-infected THP-1 monocytes (CMB). A similar inducing effect was observed on CCL20 secretion. Experiments using blocking agents revealed that IL-1ß, but not TNF-α, was involved in the induction of hBD2 and CCL20 secretion by CMB. In the in vitro antimicrobial assay, the lethal dose (LD) 50 of CCL20 for B. abortus (>50 µg/ml) was markedly higher than that against E. coli (1.5 µg/ml) or a B. abortus mutant lacking the O polysaccharide in its LPS (8.7 ug/ml). hBD2 did not kill any of the B. abortus strains at the tested concentrations. These results show that human lung epithelial cells secrete CCL20 and hBD2 in response to B. abortus and/or to cytokines produced by infected monocytes. Whereas these molecules do not seem to exert antimicrobial activity against this pathogen, they could recruit immune cells to the infection site.


Alveolar Epithelial Cells/metabolism , Brucella abortus/immunology , Brucellosis/metabolism , Chemokine CCL20/biosynthesis , beta-Defensins/biosynthesis , Alveolar Epithelial Cells/microbiology , Anti-Bacterial Agents/pharmacology , Brucellosis/immunology , Brucellosis/microbiology , Cell Line , Cell Line, Tumor , Chemokine CCL20/metabolism , Chemokine CCL20/pharmacology , Humans , Immunity, Innate , Lipopolysaccharides/pharmacology , Lung/immunology , Lung/microbiology , Lung/pathology , MAP Kinase Signaling System , Microbial Sensitivity Tests , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Toll-Like Receptor 2/metabolism , beta-Defensins/metabolism , beta-Defensins/pharmacology
4.
Clin Vaccine Immunol ; 22(3): 274-81, 2015 Mar.
Article En | MEDLINE | ID: mdl-25540276

VirB proteins from Brucella spp. constitute the type IV secretion system, a key virulence factor mediating the intracellular survival of these bacteria. Here, we assessed whether a Th1-type immune response against VirB proteins may protect mice from Brucella infection and whether this response can be induced in the dog, a natural host for Brucella. Splenocytes from mice immunized with VirB7 or VirB9 responded to their respective antigens with significant and specific production of gamma interferon (IFN-γ), whereas interleukin-4 (IL-4) was not detected. Thirty days after an intraperitoneal challenge with live Brucella abortus, the spleen load of bacteria was almost 1 log lower in mice immunized with VirB proteins than in unvaccinated animals. As colonization reduction seemed to correlate with a Th1-type immune response against VirB proteins, we decided to assess whether such a response could be elicited in the dog. Peripheral blood mononuclear cells (PBMCs) from dogs immunized with VirB proteins (three subcutaneous doses in QuilA adjuvant) produced significantly higher levels of IFN-γ than cells from control animals upon in vitro stimulation with VirB proteins. A skin test to assess specific delayed-type hypersensitivity was positive in 4 out of 5 dogs immunized with either VirB7 or VirB9. As both proteins are predicted to locate in the outer membrane of Brucella organisms, the ability of anti-VirB antibodies to mediate complement-dependent bacteriolysis of B. canis was assessed in vitro. Sera from dogs immunized with either VirB7 or VirB9, but not from those receiving phosphate-buffered saline (PBS), produced significant bacteriolysis. These results suggest that VirB-specific responses that reduce organ colonization by Brucella in mice can be also elicited in dogs.


Bacterial Proteins/immunology , Bacterial Secretion Systems , Brucella Vaccine/immunology , Brucella/growth & development , Brucella/immunology , Spleen/microbiology , Th1 Cells/immunology , Adjuvants, Immunologic , Animals , Antibodies, Bacterial/immunology , Bacterial Load , Bacterial Proteins/administration & dosage , Bacteriolysis , Brucella/pathogenicity , Brucella Vaccine/administration & dosage , Brucella abortus/immunology , Brucella canis/immunology , Dogs , Hypersensitivity, Delayed , Injections, Subcutaneous , Interferon-gamma/immunology , Interleukin-4/immunology , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Spleen/cytology , Spleen/immunology , Vaccination
5.
PLoS One ; 9(1): e82341, 2014.
Article En | MEDLINE | ID: mdl-24416141

BACKGROUND: Cross-reactivity between soybean allergens and bovine caseins has been previously reported. In this study we aimed to map epitopes of the major soybean allergen Gly m 5 that are co-recognized by casein specific antibodies, and to identify a peptide responsible for the cross-reactivity. METHODS: Cow's milk protein (CMP)-specific antibodies were used in different immunoassays (immunoblotting, ELISA, ELISA inhibition test) to evaluate the in vitro recognition of soybean proteins (SP). Recombinant Gly m 5 (α), a truncated fragment containing the C-terminal domain (α-T) and peptides of α-T were obtained and epitope mapping was performed with an overlapping peptide assay. Bioinformatics tools were used for epitope prediction by sequence alignment, and for modelling the cross-recognized soy proteins and peptides. The binding of SP to a monoclonal antibody was studied by surface Plasmon resonance (SPR). Finally, the in vivo cross-recognition of SP was assessed in a mouse model of milk allergy. RESULTS: Both α and α-T reacted with the different CMP-specific antibodies. α-T contains IgG and IgE epitopes in several peptides, particularly in the peptide named PA. Besides, we found similar values of association and dissociation constants between the α-casein specific mAb and the different milk and soy components. The food allergy mouse model showed that SP and PA contain the cross-reactive B and T epitopes, which triggered hypersensitivity reactions and a Th2-mediated response on CMP-sensitized mice. CONCLUSIONS: Gly m 5 is a cross-reactive soy allergen and the α-T portion of the molecule contains IgG and IgE immunodominant epitopes, confined to PA, a region with enough conformation to be bound by antibodies. These findings contribute to explain the intolerance to SP observed in IgE-mediated CMA patients, primarily not sensitised to SP, as well as it sets the basis to propose a mucosal immunotherapy for milk allergy using this soy peptide.


Cross Reactions/immunology , Milk Hypersensitivity/immunology , Peptides/immunology , Soybean Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Antigens, Plant/chemistry , Antigens, Plant/immunology , Cattle , Computer Simulation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Globulins/chemistry , Globulins/immunology , Immunohistochemistry , Kinetics , Mice , Milk Proteins/immunology , Molecular Sequence Data , Peptide Fragments/immunology , Protein Subunits/immunology , Recombinant Proteins/immunology , Seed Storage Proteins/chemistry , Seed Storage Proteins/immunology , Soybean Proteins/chemistry
6.
Innate Immun ; 20(6): 626-38, 2014 Aug.
Article En | MEDLINE | ID: mdl-24055879

The immunomodulatory power of heat-killed Gordonia bronchialis was studied on gut epithelial cells activated with pro-inflammatory stimuli (flagellin, TNF-α or IL-1ß). Light emission of luciferase-transfected epithelial cells and mRNA expression of IL-1ß, TNF-α, IL-6, CCL20, IL-8 and MCP-1 were measured. NF-κB activation was assessed by immunofluorescence and immunoblotting, and induction of reactive oxygen species (ROS) was evaluated. In vivo inhibitory properties of G. bronchialis were studied with ligated intestinal loop assay and in a mouse model of food allergy. G. bronchialis promoted the down-regulation of the expression of CCL20 and IL-1ß on activated epithelial cells in a dose-dependent manner. A concomitant blocking of nuclear p65 translocation with increased production of ROS was found. In vivo experiments confirmed the inhibition of CCL20 expression and the suppression of IgE sensitization and hypersensitivity symptoms in the food allergy mouse model. In conclusion, heat-killed G. bronchialis inhibited the activation of NF-κB pathway in human epithelial cells, and suppressed the expression of CCL20. These results indicate that G. bronchialis may be used to modulate the initial steps of innate immune activation, which further suppress the allergic sensitization. This approach may be exploited as a therapy for intestinal inflammation.


Epithelial Cells/immunology , Epithelium/immunology , Gordonia Bacterium/immunology , Transcription Factor RelA/biosynthesis , Animals , Caco-2 Cells , Chemokine CCL20/biosynthesis , Chemokine CCL20/metabolism , Cholera Toxin/pharmacology , Dose-Response Relationship, Drug , Down-Regulation , Genes, Reporter/genetics , Humans , Inflammation Mediators , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics
7.
Am J Pathol ; 183(6): 1918-1927, 2013 Dec.
Article En | MEDLINE | ID: mdl-24113459

In patients with active brucellosis, the liver is frequently affected by histopathologic lesions, such as granulomas, inflammatory infiltrations, and parenchymal necrosis. Herein, we examine some potential mechanisms of liver damage in brucellosis. We demonstrate that Brucella abortus infection inhibits matrix metalloproteinase-9 (MMP-9) secretion and induces collagen deposition and tissue inhibitor of matrix metalloproteinase-1 secretion induced by hepatic stellate cells (LX-2). These phenomena depend on transforming growth factor-ß1 induction. In contrast, supernatants from B. abortus-infected hepatocytes and monocytes induce MMP-9 secretion and inhibit collagen deposition in hepatic stellate cells. Yet, if LX-2 cells are infected with B. abortus, the capacity of supernatants from B. abortus-infected hepatocytes and monocytes to induce MMP-9 secretion and inhibit collagen deposition is abrogated. These results indicate that depending on the balance between interacting cells and cytokines of the surrounding milieu, the response of LX-2 cells could be turned into an inflammatory or fibrogenic phenotype. Livers from mice infected with B. abortus displayed a fibrogenic phenotype with patches of collagen deposition and transforming growth factor-ß1 induction. This study provides potential mechanisms of liver immune response induced by B. abortus-infected hepatic stellate cells. In addition, these results demonstrate that the cross talk of these cells with hepatocytes and macrophages implements a series of interactions that may contribute to explaining some of mechanisms of liver damage observed in human brucellosis.


Brucella abortus , Brucellosis , Collagen/metabolism , Gene Expression Regulation, Enzymologic , Hepatic Stellate Cells , Liver Cirrhosis , Matrix Metalloproteinase 9/biosynthesis , Transforming Growth Factor beta1/metabolism , Animals , Brucellosis/metabolism , Brucellosis/pathology , Cell Line , Down-Regulation , Female , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Inbred BALB C
8.
Gene ; 532(2): 186-91, 2013 Dec 15.
Article En | MEDLINE | ID: mdl-24076352

Gaucher disease (GD) is caused by mutations in the GBA gene that confer a deficient level of activity of glucocerebrosidase (GCase). This deficiency leads to accumulation of the glycolipid glucocerebroside in the lysosomes of cells of monocyte/macrophage system. Type I GD is the mildest form and is characterized by the absence of neuronopathic affection. Bone compromise in Gaucher disease patients is the most disabling aspect of the disease. However, pathophysiological aspects of skeletal alterations are still poorly understood. The homeostasis of bone tissue is maintained by the balanced processes of bone resorption by osteoclasts and formation by osteoblasts. We decided to test whether bone resorption and/or bone formation could be altered by the use of a chemical in vitro murine model of Gaucher disease. We used two sources of cells from monocyte/macrophages lineage isolated from normal mice, splenocytes (S) and peritoneal macrophages (PM), and were exposed to CBE, the inhibitor of GCase (S-CBE and PM-CBE, respectively). Addition of both conditioned media (CM) from S-CBE and PM-CBE induced the differentiation of osteoclasts precursors from bone marrow to mature and functional osteoclasts. TNF-α could be one of the factors responsible for this effect. On the other hand, addition of CM to an osteoblast cell culture resulted in a reduction in expression of alkaline phosphatase and mineralization process. In conclusion, these results suggest implication of changes in both bone formation and bone resorption and are consistent with the idea that both sides of the homeostatic balance are affected in GD.


Gaucher Disease/pathology , Osteoblasts/metabolism , Osteoclasts/physiology , Animals , Antigens, Differentiation/metabolism , Bone Marrow Cells/physiology , Calcification, Physiologic , Cell Differentiation , Cells, Cultured , Culture Media, Conditioned , Gaucher Disease/chemically induced , Gaucher Disease/metabolism , Inositol/analogs & derivatives , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteolysis/metabolism , Tumor Necrosis Factor-alpha/physiology
9.
Mol Genet Metab ; 109(1): 93-9, 2013 May.
Article En | MEDLINE | ID: mdl-23452955

Fabry disease is an X-linked lysosomal disorder (LD) due to deficiency of the enzyme α-galactosidase A (αGal), which leads to the accumulation of neutral glycosphingolipids, mainly globotriaosylceramide (Gb3). Several mechanisms contribute to the diverse physiopathological alterations observed in this disease, and it has been suggested that an underlying proinflammatory state could play a significant role. The aim of this study is to investigate the presence of a proinflammatory state in the different subsets of peripheral blood mononuclear cells (PBMC) and to understand the mechanisms that contribute to its onset and perpetuation. We have shown that cultured PBMC from Fabry patients present a higher proinflammatory cytokine expression and production. Moreover, we determined that among PBMC, dendritic cells and monocytes present a basal proinflammatory cytokine production profile, which is further exacerbated with an inflammatory stimulus. Finally we established that normal, monocyte-derived dendritic cells and macrophages display the same proinflammatory profile when cultured in the presence of Gb3 and an inhibitor of αGal. Furthermore, this effect can be abolished using a TLR4 blocking antibody, indicating that TLR4 is necessary in the process. In summary, our results demonstrate the presence of a proinflammatory state involving two key subsets of innate immunity, and provide direct evidence of Gb3 having a proinflammatory role, likely mediated by TLR4, a finding that could help in the understanding of the underlying causes of the inflammatory pathogenesis of Fabry disease.


Cytokines/metabolism , Fabry Disease/blood , Trihexosylceramides/blood , alpha-Galactosidase/blood , Adolescent , Adult , Aged , Child , Child, Preschool , Dendritic Cells/metabolism , Fabry Disease/enzymology , Fabry Disease/immunology , Fabry Disease/pathology , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Male , Middle Aged , Toll-Like Receptor 4/metabolism , Trihexosylceramides/immunology
10.
PLoS One ; 7(11): e50214, 2012.
Article En | MEDLINE | ID: mdl-23189190

Outer membrane vesicles (OMVs) released by some gram-negative bacteria have been shown to exert immunomodulatory effects that favor the establishment of the infection. The aim of the present study was to assess the interaction of OMVs from Brucella abortus with human epithelial cells (HeLa) and monocytes (THP-1), and the potential immunomodulatory effects they may exert. Using confocal microscopy and flow cytometry, FITC-labeled OMVs were shown to be internalized by both cell types. Internalization was shown to be partially mediated by clathrin-mediated endocytosis. Pretreatment of THP-1 cells with Brucella OMVs inhibited some cytokine responses (TNF-α and IL-8) to E. coli LPS, Pam3Cys or flagellin (TLR4, TLR2 and TLR5 agonists, respectively). Similarly, pretreatment with Brucella OMVs inhibited the cytokine response of THP-1 cells to B. abortus infection. Treatment of THP-1 cells with OMVs during IFN-γ stimulation reduced significantly the inducing effect of this cytokine on MHC-II expression. OMVs induced a dose-dependent increase of ICAM-1 expression on THP-1 cells and an increased adhesion of these cells to human endothelial cells. The addition of OMVs to THP-1 cultures before the incubation with live B. abortus resulted in increased numbers of adhered and internalized bacteria as compared to cells not treated with OMVs. Overall, these results suggest that OMVs from B. abortus exert cellular effects that promote the internalization of these bacteria by human monocytes, but also downregulate the innate immune response of these cells to Brucella infection. These effects may favor the persistence of Brucella within host cells.


Brucella abortus/immunology , Brucella abortus/metabolism , Immunity, Innate , Monocytes/immunology , Phagocytosis/immunology , Transport Vesicles/metabolism , Bacterial Adhesion/immunology , Cell Adhesion Molecules/metabolism , Cell Line , Clathrin/metabolism , Cytokines/metabolism , Endocytosis/physiology , HeLa Cells , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Interferon-gamma/pharmacology , Monocytes/drug effects , Monocytes/microbiology , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Transport Vesicles/immunology
11.
Gene ; 509(1): 51-9, 2012 Nov 01.
Article En | MEDLINE | ID: mdl-23010424

Gaucher disease is a lysosomal storage disorder caused by deficiency of glucocerebrosidase enzymatic activity leading to accumulation of its substrate glucocerebrosidase mainly in macrophages. Skeletal disorder of Gaucher disease is the major cause of morbidity and is highly refractory to enzyme replacement therapy. However, pathological mechanisms of bone alterations in Gaucher disease are still poorly understood. We hypothesized that cellular alteration in Gaucher disease produces a proinflammatory milieu leading to bone destruction through enhancement of monocyte differentiation to osteoclasts and osteoclasts resorption activity. Against this background we decided to investigate in an in vitro chemical model of Gaucher disease, the capacity of secreted soluble mediators to induce osteoclastogenesis, and the mechanism responsible for this phenomena. We demonstrated that soluble factors produced by CBE-treated PBMC induced differentiation of osteoclasts precursors into mature and active osteoclasts that express chitotriosidase and secrete proinflammatory cytokines. We also showed a role of TNF-α in promoting osteoclastogenesis in Gaucher disease chemical model. To analyze the biological relevance of T cells in osteoclastogenesis of Gaucher disease, we investigated this process in T cell-depleted PBMC cultures. The findings suggest that T cells play a role in osteoclast formation in Gaucher disease. In conclusion, our data suggests that in vitro GCASE deficiency, along with concomitant glucosylceramide accumulation, generates a state of osteoclastogenesis mediated in part by pro-resorptive cytokines, especially TNF-α. Moreover, T cells are involved in osteoclastogenesis in Gaucher disease chemical model.


Gaucher Disease/immunology , Osteoclasts/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism , Cell Differentiation , Culture Media, Conditioned , Cytokines/metabolism , Gaucher Disease/genetics , Gaucher Disease/metabolism , Gaucher Disease/pathology , Glucosylceramides/metabolism , Humans , In Vitro Techniques , Models, Biological , Osteoclasts/metabolism , Osteoclasts/pathology , Osteolysis/immunology , Osteolysis/metabolism , Osteolysis/pathology
12.
Am J Pathol ; 181(3): 887-96, 2012 Sep.
Article En | MEDLINE | ID: mdl-22901753

The pathogenic mechanisms of bone loss caused by Brucella species have not been completely deciphered. Although T lymphocytes (LTs) are considered important to control infection, the mechanism of Brucella-induced T-cell responses to immunopathological features is not known. We present in vitro and in vivo evidence showing that Brucella abortus-induced inflammatory response leads to the activation of LTs, which further promote osteoclastogenesis. Pre-activated murine LTs treated with culture supernatant from macrophages infected with B. abortus induced bone marrow-derived monocytes (BMMs) to undergo osteoclastogenesis. Furthermore, osteoclastogenesis was mediated by CD4(+) T cells. Although B. abortus-activated T cells actively secreted the pro-osteoclastogenic cytokines RANKL and IL-17, osteoclastogenesis depended on IL-17, because osteoclast generation induced by Brucella-activated T cells was completely abrogated when these cells were cultured with BMMs from IL-17 receptor knockout mice. Neutralization experiments indicated that IL-6, generated by Brucella infection, induced the production of pro-osteoclastogenic IL-17 from LTs. By using BMMs from tumor necrosis factor receptor p55 knockout mice, we also demonstrated that IL-17 indirectly induced osteoclastogenesis through the induction of tumor necrosis factor-α from osteoclast precursors. Finally, extensive and widespread osteoclastogenesis was observed in the knee joints of mice injected with Brucella-activated T cells. Our results indicate that activated T cells, elicited by B. abortus-infected macrophages and influenced by the inflammatory milieu, promote the generation of osteoclasts, leading to bone loss.


Brucella abortus/immunology , Interleukin-17/metabolism , Macrophages/microbiology , Osteoclasts/immunology , Osteogenesis/immunology , T-Lymphocytes/immunology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Brucellosis/immunology , Brucellosis/microbiology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Interleukin-17/biosynthesis , Interleukin-6/metabolism , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/metabolism , RANK Ligand/metabolism , Receptors, Interleukin-17/metabolism , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/metabolism , Subcellular Fractions/metabolism , Tibia/immunology , Tibia/pathology , Tumor Necrosis Factor Decoy Receptors/deficiency , Tumor Necrosis Factor Decoy Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
Infect Immun ; 80(7): 2333-45, 2012 Jul.
Article En | MEDLINE | ID: mdl-22547546

Osteoarticular brucellosis is the most common presentation of the active disease in humans. Loss of bone is a serious complication of localized bacterial infection of bones or the adjacent tissue, and brucellosis proved not to be the exception. The skeleton is a dynamic organ system which is constantly remodeled. Osteoblasts are responsible for the deposition of bone matrix and are thought to facilitate the calcification and mineralization of the bone matrix, and their function could be altered under infectious conditions. In this article, we describe immune mechanisms whereby Brucella abortus may invade and replicate within osteoblasts, inducing apoptosis, inhibiting mineral and organic matrix deposition, and inducing upregulation of RANKL expression. Additionally, all of these mechanisms contributed in different ways to bone loss. These processes implicate the activation of signaling pathways (mitogen-activated protein kinases [MAPK] and caspases) involved in cytokine secretion, expression of activating molecules, and cell death of osteoblasts. In addition, considering the relevance of macrophages in intracellular Brucella survival and proinflammatory cytokine secretion in response to infection, we also investigated the role of these cells as modulators of osteoblast survival, differentiation, and function. We demonstrated that supernatants from B. abortus-infected macrophages may also mediate osteoblast apoptosis and inhibit osteoblast function in a process that is dependent on the presence of tumor necrosis factor alpha (TNF-α). These results indicate that B. abortus may directly and indirectly harm osteoblast function, contributing to the bone and joint destruction observed in patients with osteoarticular complications of brucellosis.


Apoptosis , Brucella abortus/pathogenicity , Osteoblasts/metabolism , Osteoblasts/microbiology , Osteogenesis , Animals , Cells, Cultured , Macrophages/immunology , Macrophages/microbiology , Mice
14.
FEMS Immunol Med Microbiol ; 66(1): 45-57, 2012 Oct.
Article En | MEDLINE | ID: mdl-22553918

In spite of the frequent acquisition of Brucella infection by the oral route in humans, the interaction of the bacterium with cells of the intestinal mucosa has been poorly studied. Here, we show that different Brucella species can invade human colonic epithelial cell lines (Caco-2 and HT-29), in which only smooth species can replicate efficiently. Infection with smooth strains did not produce a significant cytotoxicity, while the rough strain RB51 was more cytotoxic. Infection of Caco-2 cells or HT-29 cells with either smooth or rough strains of Brucella did not result in an increased secretion of TNF-α, IL-1ß, MCP-1, IL-10 or TGF-ß as compared with uninfected controls, whereas all the infections induced the secretion of IL-8 and CCL20 by both cell types. The MCP-1 response to flagellin from Salmonella typhimurium was similar in Brucella-infected or uninfected cells, ruling out a bacterial inhibitory mechanism as a reason for the weak proinflammatory response. Infection did not modify ICAM-1 expression levels in Caco-2 cells, but increased them in HT-29 cells. These results suggest that Brucella induces only a weak proinflammatory response in gut epithelial cells, but produces a significant CCL20 secretion. The latter may be important for bacterial dissemination given the known ability of Brucella to survive in dendritic cells.


Brucella/immunology , Brucella/pathogenicity , Chemokine CCL20/metabolism , Epithelial Cells/immunology , Epithelial Cells/microbiology , Cell Line , Humans
15.
J Leukoc Biol ; 91(2): 285-98, 2012 Feb.
Article En | MEDLINE | ID: mdl-22075930

Osteoarticular complications are common in human brucellosis, but the pathogenic mechanisms involved are largely unknown. In this manuscript, we described an immune mechanism for inflammatory bone loss in response to infection by Brucella abortus. We established a requirement for MyD88 and TLR2 in TNF-α-elicited osteoclastogenesis in response to B. abortus infection. CS from macrophages infected with B. abortus induced BMM to undergo osteoclastogenesis. Although B. abortus-infected macrophages actively secreted IL-1ß, IL-6, and TNF-α, osteoclastogenesis depended on TNF-α, as CS from B. abortus-infected macrophages failed to induce osteoclastogenesis in BMM from TNFRp55⁻/⁻ mice. CS from B. abortus-stimulated MyD88⁻/⁻ and TLR2⁻/⁻ macrophages failed to express TNF-α, and these CS induced no osteoclast formation compared with that of the WT or TLR4⁻/⁻ macrophages. Omp19, a B. abortus lipoprotein model, recapitulated the cytokine production and subsequent osteoclastogenesis induced by the whole bacterium. All phenomena were corroborated using human monocytes, indicating that this mechanism could play a role in human osteoarticular brucellosis. Our results indicate that B. abortus, through its lipoproteins, may be involved in bone resorption through the pathological induction of osteoclastogenesis.


Brucella abortus/physiology , Macrophages, Peritoneal/physiology , Myeloid Differentiation Factor 88/physiology , Osteoclasts/physiology , Toll-Like Receptor 2/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Antigens, Bacterial/pharmacology , Bacterial Outer Membrane Proteins/pharmacology , Brucellosis/immunology , Cell Differentiation/drug effects , Cells, Cultured/drug effects , Cells, Cultured/physiology , Culture Media, Conditioned/pharmacology , Cytokines/biosynthesis , Female , Humans , Lipoproteins/pharmacology , Macrophages, Peritoneal/drug effects , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/physiology , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , RANK Ligand/biosynthesis , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/deficiency , Transforming Growth Factor beta/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
16.
Mol Genet Metab ; 104(3): 319-24, 2011 Nov.
Article En | MEDLINE | ID: mdl-21724436

Fabry disease is an X-linked lysosomal storage disorder (LSD) due to deficiency of the enzyme α-galactosidase A, resulting in intracellular deposition of globotriaosylceramide (Gb3). Accumulation of Gb3 is probably related to tissue and organ dysfunctions. Diverse pathological mechanisms are elicited in LSDs, giving together the phenotypic expression of each disease. The purpose of the present study is to investigate if apoptosis could play a role in Fabry disease pathogenesis and to understand the mechanisms involved in the proapoptotic state. We have demonstrated that Fabry disease peripheral blood mononuclear cells display a higher apoptotic state, which is reduced by enzyme replacement therapy (ERT), and is mediated, at least in part, by activation of the intrinsic pathway of caspases. We could rule out the implication of "unfolded protein response-ER stress" in this apoptotic process. To further confirm the suggestion that Gb3 is associated to apoptotic cell death, we treated normal cells with Gb3 at concentrations found in Fabry patients. Addition of Gb3 resulted in a dose-dependent induction of apoptosis involving the intrinsic pathway. In summary, PBMC from Fabry patients display a higher apoptotic state, which could be mainly related to elevated Gb3.


Apoptosis/physiology , Fabry Disease/physiopathology , Leukocytes, Mononuclear/metabolism , Trihexosylceramides/metabolism , Adolescent , Adult , Analysis of Variance , Annexin A5 , Apoptosis/drug effects , Child , Child, Preschool , Dose-Response Relationship, Drug , Enzyme Replacement Therapy , Fabry Disease/drug therapy , Female , Flow Cytometry , Humans , Immunoblotting , In Situ Nick-End Labeling , Isoenzymes , Leukocytes, Mononuclear/physiology , Male , Middle Aged , Polymerase Chain Reaction , Trihexosylceramides/pharmacology , alpha-Galactosidase
17.
Infect Immun ; 79(9): 3619-32, 2011 Sep.
Article En | MEDLINE | ID: mdl-21730088

Arthritis is one of the most common complications of human brucellosis, but its pathogenic mechanisms have not been elucidated. Fibroblast-like synoviocytes (FLS) are known to be central mediators of joint damage in inflammatory arthritides through the production of matrix metalloproteinases (MMPs) that degrade collagen and of cytokines and chemokines that mediate the recruitment and activation of leukocytes. In this study we show that Brucella abortus infects and replicates in human FLS (SW982 cell line) in vitro and that infection results in the production of MMP-2 and proinflammatory mediators (interleukin-6 [IL-6], IL-8, monocyte chemotactic protein 1 [MCP-1], and granulocyte-macrophage colony-stimulating factor [GM-CSF]). Culture supernatants from Brucella-infected FLS induced the migration of monocytes and neutrophils in vitro and also induced these cells to secrete MMP-9 in a GM-CSF- and IL-6-dependent fashion, respectively. Reciprocally, culture supernatants from Brucella-infected monocytes and neutrophils induced FLS to produce MMP-2 in a tumor necrosis factor alpha (TNF-α)-dependent fashion. The secretion of proinflammatory mediators and MMP-2 by FLS did not depend on bacterial viability, since it was also induced by heat-killed B. abortus (HKBA) and by a model Brucella lipoprotein (L-Omp19). These responses were mediated by the recognition of B. abortus antigens through Toll-like receptor 2. The intra-articular injection of HKBA or L-Omp19 into the knee joint of mice resulted in the local induction of the proinflammatory mediators MMP-2 and MMP-9 and in the generation of a mixed inflammatory infiltrate. These results suggest that FLS, and phagocytes recruited by them to the infection focus, may be involved in joint damage during brucellar arthritis through the production of MMPs and proinflammatory mediators.


Arthritis, Infectious/immunology , Brucella abortus/immunology , Brucellosis/immunology , Joints/microbiology , Joints/pathology , Matrix Metalloproteinases/biosynthesis , Synovial Membrane/immunology , Animals , Antigens, Bacterial/immunology , Arthritis, Infectious/enzymology , Arthritis, Infectious/microbiology , Arthritis, Infectious/pathology , Bacterial Outer Membrane Proteins/immunology , Brucella abortus/growth & development , Brucella abortus/pathogenicity , Brucellosis/enzymology , Brucellosis/microbiology , Brucellosis/pathology , Cell Line , Cell Movement/drug effects , Chemokines/biosynthesis , Culture Media, Conditioned , Cytokines/biosynthesis , Cytokines/metabolism , Enzyme Induction , Enzyme-Linked Immunosorbent Assay , Female , Humans , Knee Joint/microbiology , Lipoproteins/immunology , Lymphocyte Activation , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred BALB C , Monocytes/physiology , Neutrophils/physiology , Synovial Membrane/cytology , Synovial Membrane/microbiology , Toll-Like Receptor 2/metabolism
18.
Microbes Infect ; 13(10): 852-61, 2011 Sep.
Article En | MEDLINE | ID: mdl-21621633

Although vascular pathologies such as vasculitis, endocarditis and mycotic aneurysms have been described in brucellosis patients, the interaction of Brucella with the endothelium has not been characterized. In this study we show that Brucella abortus and Brucella suis can infect and replicate in primary human umbilical vein endothelial cells (HUVEC) and in the microvascular endothelial cell line HMEC-1. Infection led to an increased production of IL-8, MCP-1 and IL-6 in HUVEC and HMEC-1 cells, and an increased expression of adhesion molecules (CD54 in both cells, CD106 and CD62E in HUVEC). Experiments with purified antigens from the bacterial outer membrane revealed that lipoproteins (Omp19) but not lipopolysaccharide mediate these proinflammatory responses. Infection of polarized HMEC-1 cells resulted in an increased capacity of these cells to promote the transmigration of neutrophils from the apical to the basolateral side of the monolayer, and the same phenomenon was observed when the cells were stimulated with live bacteria from the basolateral side. Overall, these results suggest that Brucella spp. can infect and survive within endothelial cells, and can induce a proinflammatory response that might be involved in the vascular manifestations of brucellosis.


Brucella abortus/immunology , Brucella abortus/pathogenicity , Brucella suis/immunology , Brucella suis/pathogenicity , Cytokines/metabolism , Endothelial Cells/immunology , Endothelial Cells/microbiology , Antigens, Bacterial/immunology , Antigens, CD/biosynthesis , Bacterial Outer Membrane Proteins/immunology , Cells, Cultured , Humans , Lipoproteins/immunology , Neutrophils/immunology , Transendothelial and Transepithelial Migration
19.
Infect Immun ; 79(1): 192-202, 2011 Jan.
Article En | MEDLINE | ID: mdl-20956574

Osteoarticular complications are common in human brucellosis, but the pathogenic mechanisms involved are largely unknown. Since matrix metalloproteinases (MMPs) are involved in joint and bone damage in inflammatory and infectious diseases, we investigated the production of MMPs by human osteoblasts and monocytes, either upon Brucella abortus infection or upon reciprocal stimulation with factors produced by each infected cell type. B. abortus infection of the normal human osteoblastic cell line hFOB 1.19 triggered a significant release of MMP-2, which was mediated in part by granulocyte-macrophage colony-stimulating factor (GM-CSF) acting on these same cells. Supernatants from infected osteoblasts exhibited increased levels of monocyte chemoattractant protein 1 and induced the migration of human monocytes (THP-1 cell line). Infection with B. abortus induced a high MMP-9 secretion in monocytes, which was also induced by heat-killed B. abortus and by the Omp19 lipoprotein from B. abortus. These effects were mediated by Toll-like receptor 2 and by the action of tumor necrosis factor alpha (TNF-α) produced by these same cells. Supernatants from B. abortus-infected monocytes induced MMP-2 secretion in uninfected osteoblasts, and this effect was mediated by TNF-α. Similarly, supernatants from infected osteoblasts induced MMP-9 secretion in uninfected monocytes. This effect was mediated by GM-CSF, which induced TNF-α production by monocytes, which in turn induced MMP-9 in these cells. These results suggest that MMPs could be potentially involved in the tissue damage observed in osteoarticular brucellosis.


Brucella abortus/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Matrix Metalloproteinases/metabolism , Monocytes/microbiology , Osteoblasts/microbiology , Tumor Necrosis Factor-alpha/metabolism , Cell Line , Gene Expression Regulation/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Monocytes/metabolism , Osteoblasts/metabolism
20.
J Med Microbiol ; 59(Pt 12): 1514-1518, 2010 Dec.
Article En | MEDLINE | ID: mdl-20724508

A case of prepatellar bursitis in a man with chronic brucellosis is presented. Brucella abortus biotype 1 was isolated from the abundant yellowish fluid obtained from the bursa. Clinical and epidemiological data did not suggest a direct inoculation of the agent in the bursa. However, the patient mentioned occasional local trauma due to recreational sports, which may have constituted a predisposing factor. As determined by ELISA, there were higher levels of IgG against Brucella LPS and cytosolic proteins detected in the patient's bursal synovial fluid when compared with serum. Levels of proinflammatory cytokines (tumour necrosis factor alpha, interleukin 1 beta, gamma interferon, interleukin 8 and MCP-1) were higher than in synovial fluids obtained from patients with rheumatoid arthritis and a patient with septic arthritis, and a zymographic analysis revealed a gelatinase of about 92 kDa. These findings indicate that it may be possible to diagnose brucellar bursitis by measuring specific antibodies in the bursal synovial fluid. In addition, our findings suggest a role of increased local levels of proinflammatory cytokines and gelatinases in the inflammatory manifestations of brucellar bursitis.


Brucella abortus/isolation & purification , Brucellosis/pathology , Bursitis/microbiology , Anti-Bacterial Agents/therapeutic use , Biomarkers/analysis , Brucellosis/drug therapy , Cytokines/analysis , Humans , Male , Middle Aged , Synovial Fluid/chemistry , Synovial Fluid/microbiology
...