Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
J Autoimmun ; 51: 30-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24602383

ABSTRACT

Although B cell activating factor (BAFF) and its receptor BR3 are produced and expressed by many cells, their role has been restricted to the lymphocyte lineage. Using various techniques (RT-PCR, indirect immunofluorescence, flow cytometry analysis), we observed the expression of BR3 and the production of BAFF by the human salivary gland cell line, by epithelial cells from biopsies of Sjögren's syndrome patients and their controls, but also by salivary gland epithelial cells in culture. To decipher the role of BAFF and BR3 on epithelial cells, BAFF and BR3 were neutralized by blocking antibodies or RNA specific inhibitor (siBR3) and epithelial cell survival was analyzed. Blocking BR3 promotes epithelial cell apoptosis in vitro. This apoptosis resulted in the nuclear translocation of PKCδ. BAFF neutralization by various anti-BAFF antibodies leads to different effects depending on the antibody used suggesting that only some forms of BAFF are required for epithelial cell survival. Our study demonstrates that BR3 is involved in the survival of cultured epithelial cells due to an autocrine effect of BAFF. It also suggests that epithelial cells produce different forms of BAFF and that only some of them are responsible for this effect.


Subject(s)
B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/metabolism , Epithelial Cells/metabolism , Adult , Aged , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , B-Cell Activating Factor/genetics , B-Cell Activation Factor Receptor/antagonists & inhibitors , B-Cell Activation Factor Receptor/genetics , Biopsy , Cell Survival/drug effects , Cell Survival/genetics , Female , Gene Expression , Humans , Immunohistochemistry , Immunophenotyping , Male , Middle Aged , Salivary Glands/pathology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/immunology , Sjogren's Syndrome/metabolism
2.
J Autoimmun ; 51: 23-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24582318

ABSTRACT

Toll-like receptors (TLRs) are positioned at the interface between innate and adaptive immunity. Unlike others, those such as TLR9, that recognize nucleic acids, are confined to the endosomal compartment and are scarce on the cell surface. Here, we present evidence for TLR9 expression on the plasma membrane of B cells. In contrast to endosomal TLR9, cell surface TLR9 does not bind CpG-B oligodeoxynucleotides. After B cell-receptor (BCR) stimulation, TLR9 was translocated into lipid rafts with the BCR, suggesting that it could serve as a co-receptor for BCR. Nevertheless, stimulation of B cells with anti-TLR9 antibodies did not modify the BCR-induced responses despite up-regulation of tyrosine phosphorylation of proteins. However, CpG-B activation of B cells, acting synergistically with BCR signals, was inhibited by anti-TLR9 stimulation. Induction of CD25 expression and proliferation of B cells were thus down-regulated by the engagement of cell surface TLR9. Overall, our results indicate that TLR9 expressed on the plasma membrane of B cells might be a negative regulator of endosomal TLR9, and could provide a novel control by which activation of autoreactive B cells is restrained.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Membrane/metabolism , Immunomodulation , Toll-Like Receptor 9/metabolism , Cells, Cultured , Humans , Immunophenotyping , Ligands , Lymphocyte Activation/immunology , Oligodeoxyribonucleotides/metabolism , Protein Binding
3.
Presse Med ; 41(9 Pt 2): e467-74, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22836197

ABSTRACT

The continuing progress in discovering lymphocyte subsets and the lengthening list of cytokines involved, together with how they are affected in primary Sjögren's syndrome (pSS), has further fuelled the debate on pSS pathogenesis. In this review the "interferon signature" observed in the salivary glands and the role of T-cell derived cytokines (Th1/Th2 polarization, Th17 and regulatory T cells) will be discussed. A particular emphasis has been placed on the B-cell derived cytokines and especially on FLT3-Ligand, a cytokine associated with lymphoma in pSS, and B-cell activating factor (BAFF) that prevents apoptosis of autoreactive B cells. It has indeed become a challenge to understand how the interaction between several interconnected networks of cytokines impact so different cell population in the immunopathogenesis of pSS.


Subject(s)
Cytokines/immunology , Sjogren's Syndrome/immunology , Autoimmune Diseases/immunology , B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Cell Polarity/immunology , Hematopoiesis/immunology , Humans , Interferons/immunology , Ligands , Membrane Proteins/immunology , Salivary Glands/immunology , T-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology
4.
J Autoimmun ; 39(3): 189-98, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22749832

ABSTRACT

The B-cell activating factor belonging to the tumor-necrosis factor family BAFF contributes to autoimmune disorders. As such, BAFF might become a therapeutic target. However, this molecule has pleiotropic effects that are as numerous as they are varied. The real effect of each form (spliced, glycosylated, membrane bound, soluble, homotrimerized, heterotrimerized, multimerized) has not been well characterized yet. Consequently, conflicting results, regarding the serum concentrations of BAFF or its functional effect, exist in literature. BAFF quantification based on ELISA commercial kits was indeed found to be inaccurate. The complexity of the various forms of BAFF will be reviewed by focusing on the different structural aspects of the molecule. These data have particular implications for autoimmunity, not only because of the role of these factors on B cell growth and survival, but also their influence on the onset and severity of several autoimmune diseases.


Subject(s)
Autoimmunity , B-Cell Activating Factor/immunology , B-Lymphocytes/immunology , Animals , B-Cell Activating Factor/genetics , B-Lymphocytes/pathology , Cytokine TWEAK , Exons , Humans , Introns , Mice , Polymorphism, Single Nucleotide , Protein Conformation , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Multimerization , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/immunology
5.
PLoS One ; 5(12): e15466, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21151933

ABSTRACT

Cationic amino acid transporters (mCAT1 and mCAT2B) regulate the arginine availability in macrophages. How in the infected cell a pathogen can alter the arginine metabolism of the host remains to be understood. We reveal here a novel mechanism by which Salmonella exploit mCAT1 and mCAT2B to acquire host arginine towards its own intracellular growth within antigen presenting cells. We demonstrate that Salmonella infected bone marrow derived macrophages and dendritic cells show enhanced arginine uptake and increased expression of mCAT1 and mCAT2B. We show that the mCAT1 transporter is in close proximity to Salmonella containing vacuole (SCV) specifically by live intracellular Salmonella in order to access the macrophage cytosolic arginine pool. Further, Lysosome associated membrane protein 1, a marker of SCV, also was found to colocalize with mCAT1 in the Salmonella infected cell. The intra vacuolar Salmonella then acquire the host arginine via its own arginine transporter, ArgT for growth. The argT knockout strain was unable to acquire host arginine and was attenuated in growth in both macrophages and in mice model of infection. Together, these data reveal survival strategies by which virulent Salmonella adapt to the harsh conditions prevailing in the infected host cells.


Subject(s)
Amino Acid Transport Systems, Basic/physiology , Arginine/chemistry , Salmonella typhimurium/metabolism , Animals , Arginine/metabolism , Bacterial Proteins/metabolism , Cells, Cultured , Cytosol/metabolism , Genetic Complementation Test , Macrophages/cytology , Mice , Mice, Inbred BALB C , Mycobacterium bovis/metabolism , Nitrites/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Salmonella Infections/metabolism , Virulence
6.
PLoS One ; 5(10): e13772, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-21048937

ABSTRACT

Synthetic CpG containing oligodeoxynucleotide Toll like receptor-9 agonist (CpG DNA) activates innate immunity and can stimulate antigen presentation against numerous intracellular pathogens. It was observed that Salmonella Typhimurium growth can be inhibited by the CpG DNA treatment in the murine dendritic cells. This inhibitory effect was mediated by an increased reactive oxygen species production. In addition, it was noted that CpG DNA treatment of dendritic cells during Salmonella infection leads to an increased antigen presentation. Further this increased antigen presentation was dependent on the enhanced reactive oxygen species production elicited by Toll like receptor-9 activation. With the help of an exogenous antigen it was shown that Salmonella antigen could also be cross-presented in a better way by CpG induction. These data collectively indicate that CpG DNA enhance the ability of murine dendritic cells to contain the growth of virulent Salmonella through reactive oxygen species dependent killing.


Subject(s)
Antigens, Bacterial/immunology , Dendritic Cells/metabolism , Reactive Oxygen Species/metabolism , Salmonella/immunology , Toll-Like Receptor 9/metabolism , CpG Islands , Dendritic Cells/immunology , Humans
7.
Microbes Infect ; 12(11): 809-18, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20538070

ABSTRACT

Salmonella, a Gram-negative facultative intracellular pathogen is capable of infecting vast array of hosts. The striking ability of Salmonella to overcome every hurdle encountered in the host proves that they are true survivors. In the host, Salmonella infects various cell types and needs to survive and replicate by countering the defense mechanism of the specific cell. In this review, we will summarize the recent insights into the cell biology of Salmonella infection. Here, we will focus on the findings that deal with the specific mechanism of various cell types to control Salmonella infection. Further, the survival strategies of the pathogen in response to the host immunity will also be discussed in detail. Better understanding of the mechanisms by which Salmonella evade the host defense system and establish pathogenesis will be critical in disease management.


Subject(s)
Host-Pathogen Interactions , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella/immunology , Salmonella/pathogenicity , Humans , Immune Evasion , Microbial Viability
8.
PLoS Pathog ; 6(6): e1000899, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20585552

ABSTRACT

Arginine is a crucial amino acid that serves to modulate the cellular immune response during infection. Arginine is also a common substrate for both inducible nitric oxide synthase (iNOS) and arginase. The generation of nitric oxide from arginine is responsible for efficient immune response and cytotoxicity of host cells to kill the invading pathogens. On the other hand, the conversion of arginine to ornithine and urea via the arginase pathway can support the growth of bacterial and parasitic pathogens. The competition between iNOS and arginase for arginine can thus contribute to the outcome of several parasitic and bacterial infections. There are two isoforms of vertebrate arginase, both of which catalyze the conversion of arginine to ornithine and urea, but they differ with regard to tissue distribution and subcellular localization. In the case of infection with Mycobacterium, Leishmania, Trypanosoma, Helicobacter, Schistosoma, and Salmonella spp., arginase isoforms have been shown to modulate the pathology of infection by various means. Despite the existence of a considerable body of evidence about mammalian arginine metabolism and its role in immunology, the critical choice to divert the host arginine pool by pathogenic organisms as a survival strategy is still a mystery in infection biology.


Subject(s)
Arginase/metabolism , Bacteria/pathogenicity , Bacterial Infections/pathology , Immunologic Factors/metabolism , Signal Transduction , Animals , Bacteria/enzymology , Bacterial Infections/enzymology , Bacterial Infections/immunology , Humans , Isoenzymes
9.
Microbiology (Reading) ; 155(Pt 8): 2476-2489, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19520723

ABSTRACT

Activation of macrophages by interferon gamma (IFN-gamma) and the subsequent production of nitric oxide (NO) are critical for the host defence against Salmonella enterica serovar Typhimurium infection. We report here the inhibition of IFN-gamma-induced NO production in RAW264.7 macrophages infected with wild-type Salmonella. This phenomenon was shown to be dependent on the nirC gene, which encodes a potential nitrite transporter. We observed a higher NO output from IFN-gamma-treated macrophages infected with a nirC mutant of Salmonella. The nirC mutant also showed significantly decreased intracellular proliferation in a NO-dependent manner in activated RAW264.7 macrophages and in liver, spleen and secondary lymph nodes of mice, which was restored by complementing the gene in trans. Under acidified nitrite stress, a twofold more pronounced NO-mediated repression of SPI2 was observed in the nirC knockout strain compared to the wild-type. This enhanced SPI2 repression in the nirC knockout led to a higher level of STAT-1 phosphorylation and inducible nitric oxide synthase (iNOS) expression than seen with the wild-type strain. In iNOS knockout mice, the organ load of the nirC knockout strain was similar to that of the wild-type strain, indicating that the mutant is exclusively sensitive to the host nitrosative stress. Taken together, these results reveal that intracellular Salmonella evade killing in activated macrophages by downregulating IFN-gamma-induced NO production, and they highlight the critical role of nirC as a virulence gene.


Subject(s)
Anion Transport Proteins/physiology , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Macrophages/microbiology , Membrane Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Salmonella Infections, Animal/microbiology , Salmonella/physiology , Salmonella/pathogenicity , Animals , Cell Culture Techniques , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Interferon-gamma/metabolism , Macrophages/metabolism , Mice , Nitric Oxide/metabolism , Phosphorylation , STAT1 Transcription Factor/metabolism , Salmonella/genetics , Salmonella Infections, Animal/metabolism , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...