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










Publication year range
1.
Infect Immun ; 89(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33257533

ABSTRACT

The majority of Gram-negative bacteria elicit a potent immune response via recognition of lipid A expressed on the outer bacterial membrane by the host immune receptor Toll-like receptor 4 (TLR4). However, some Gram-negative bacteria evade detection by TLR4 or alter the outcome of TLR4 signaling by modification of lipid A species. Although the role of lipid A modifications on host innate immunity has been examined in some detail, it is currently unclear how lipid A remodeling influences host adaptive immunity. One prototypic Gram-negative bacterium that modifies its lipid A structure is Porphyromonas gingivalis, an anaerobic pathobiont that colonizes the human periodontium and induces chronic low-grade inflammation that is associated with periodontal disease as well as a number of systemic inflammatory disorders. P. gingivalis produces dephosphorylated and deacylated lipid A structures displaying altered activities at TLR4. Here, we explored the functional role of P. gingivalis lipid A modifications on TLR4-dependent innate and adaptive immune responses in mouse bone marrow-derived dendritic cells (BMDCs). We discovered that lipid A 4'-phosphate removal is required for P. gingivalis to evade BMDC-dependent proinflammatory cytokine responses and markedly limits the bacterium's capacity to induce beta interferon (IFN-ß) production. In addition, lipid A 4'-phosphatase activity prevents canonical bacterium-induced delay in antigen degradation, which leads to inefficient antigen cross-presentation and a failure to cross-prime CD8 T cells specific for a P. gingivalis-associated antigen. We propose that lipid A modifications produced by this bacterium alter host TLR4-dependent adaptive immunity to establish chronic infections associated with a number of systemic inflammatory disorders.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cross-Priming/physiology , Dendritic Cells/metabolism , Immunity, Innate/physiology , Lipopolysaccharides/metabolism , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/pathogenicity , Genetic Variation , Genotype , Host-Pathogen Interactions , Humans , Periodontium/microbiology , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/immunology
3.
Blood ; 121(9): 1574-83, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23297132

ABSTRACT

Antiviral immunity and cross-presentation is mediated constitutively through CD8α+ and CD103+ DCs. Development of these DC subsets is thought to require the transcription factors Irf8, Id2, Nfil3, and Batf3, although how this network is regulated is poorly defined. We addressed the nature of the differentiation blocks observed in the absence of these factors and found that although all 4 factors are required for CD103+ DC development, only Irf8 is essential for CD8α+ DCs. CD8α+ DCs emerged in the absence of Id2, Nfil3 and Batf3 in short-term bone marrow reconstitution. These "induced" CD8α+ DCs exhibit several hallmarks of classic CD8α+ DCs including the expression of CD24, Tlr3, Xcr1, Clec9A, and the capacity to cross-present soluble, cell-associated antigens and viral antigens even in the absence of Batf3. Collectively, these results uncover a previously undescribed pathway by which CD8α+ DCs emerge independent of Id2, Nfil3, and Batf3, but dependent on Irf8.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , CD8 Antigens/metabolism , Cell Differentiation , Dendritic Cells/physiology , Inhibitor of Differentiation Protein 2/physiology , Repressor Proteins/physiology , Animals , Antigens, Viral/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Cross-Priming/genetics , Cross-Priming/immunology , Cross-Priming/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Herpesvirus 1, Human/immunology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology
4.
Exp Dermatol ; 21(11): 876-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23163655

ABSTRACT

MicroRNAs (miRNAs) are evolutionarily conserved small non-coding RNAs that repress target genes at post-transcriptional level. Langerhans cells (LCs) are skin-residential dendritic cells (DCs) with a life cycle distinct from other types of DCs. miRNA deficiency interrupts the homoeostasis and function of epidermal LCs, suggesting the critical roles of miRNAs in LC development and function. However, the roles of individual miRNAs in regulating LC development and function remain completely unknown. MiRNA miR-150 is highly expressed in mature lymphocytes and regulates T- and B-cell development and function. Here, we reported that miR-150 is also expressed in epidermal LCs, and its expression is significantly down-regulated during in vitro LC maturation. Using a miR-150 knockout mouse model, we found that lack of miR-150 reduces the capacity of LCs to cross-present a soluble antigen to antigen-specific CD8(+) T cells, but does not disturb the development, maturation, migration and phagocytic capacity of LCs. Thus, our data indicate that miR-150 is required for LC cross-presentation.


Subject(s)
Cross-Priming/physiology , Epidermis/immunology , Langerhans Cells/immunology , MicroRNAs/genetics , MicroRNAs/physiology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Epidermal Cells , Gene Knockdown Techniques , In Vitro Techniques , Langerhans Cells/cytology , Mice , Mice, Knockout
5.
Blood ; 118(15): 4111-9, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21860028

ABSTRACT

Targeting antigens to dendritic cell (DC)-specific receptors, such as DC-SIGN, induces potent T cell-mediated immune responses. DC-SIGN is a transmembrane C-type lectin receptor with a long extracellular neck region and a carbohydrate recognition domain (CRD). Thus far, only antibodies binding the CRD have been used to target antigens to DC-SIGN. We evaluated the endocytic pathway triggered by antineck antibodies as well as their intracellular routing and ability to induce CD8(+) T-cell activation. In contrast to anti-CRD antibodies, antineck antibodies induced a clathrin-independent mode of DC-SIGN internalization, as demonstrated by the lack of colocalization with clathrin and the observation that silencing clathrin did not affect antibody internalization in human DCs. Interestingly, we observed that anti-neck and anti-CRD antibodies were differentially routed within DCs. Whereas anti-CRD antibodies were mainly routed to late endosomal compartments, anti-neck antibodies remained associated with early endosomal compartments positive for EEA-1 and MHC class I for up to 2 hours after internalization. Finally, cross-presentation of protein antigen conjugated to antineck antibodies was approximately 1000-fold more effective than nonconjugated antigen. Our studies demonstrate that anti-neck antibodies trigger a distinct mode of DC-SIGN internalization that shows potential for targeted vaccination strategies.


Subject(s)
Antigen Presentation/physiology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/immunology , Cross-Priming/physiology , Dendritic Cells/immunology , Endosomes/immunology , Lectins, C-Type/immunology , Lysosomes/immunology , Receptors, Cell Surface/immunology , Animals , Antibodies/genetics , Antibodies/immunology , Antigens/genetics , Antigens/immunology , CHO Cells , Cell Adhesion Molecules/genetics , Cricetinae , Cricetulus , Endosomes/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunity, Cellular/physiology , Lectins, C-Type/genetics , Lysosomes/genetics , Mice , Mice, Transgenic , Protein Structure, Tertiary , Receptors, Cell Surface/genetics , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/immunology
6.
Ann N Y Acad Sci ; 1183: 237-50, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20146719

ABSTRACT

Many prokaryotic and eukaryotic parasites multiply in specialized subcellular niches in the host cell. The invading microbes hijack key cellular functions to establish the intracellular niches but, unlike viruses, do not need the protein synthesis machinery of host cells to replicate. Circulating CD8+ T cells provide protective immunity by recognizing pathogen-derived peptide major histocompatibility complex class I molecules (pMHC I) expressed by infected cells. Here, we review studies on the complex and varied pathways that produce the appropriate pMHC I as ligands for the CD8+ T cells. We also discuss possible explanations for the curious observations that CD8+ T cells are specific for fewer pMHC I ligands in parasite infections compared to the diversity of pMHC I ligands in viral infections.


Subject(s)
Cross-Priming/physiology , Histocompatibility Antigens Class I/metabolism , Intracellular Space/microbiology , Peptides/immunology , Animals , Antigen Presentation/immunology , Antigens, Bacterial/immunology , Antigens, Viral/immunology , Cross-Priming/immunology , Histocompatibility Antigens Class I/immunology , Humans , Intracellular Space/immunology , Lymphocyte Activation/immunology , Models, Biological , T-Lymphocytes/immunology
7.
Allergol. immunopatol ; 36(2): 113-116, abr. 2008. ilus
Article in En | IBECS | ID: ibc-64444

ABSTRACT

The use of pine nuts, the seeds of Pinus pinea, is on the increasing in the modern Mediterranean diet. Little more than 20 cases of allergy to this tree nut have been published, and cross-reactivity with pine pollen, peanut and almond has already been reported. We describe the case of a young boy with several episodes of anaphylaxis after pine nut ingestion. Specific IgE to pine nut and Artemisia vulgaris was demonstrated by skin prick tests and in vitro determination of specific IgE, although no IgE to pine pollen or other nuts was detected. Immunoblotting of Artemisia vulgaris and pine nut revealed two matching diffuse bands, just below 14 kDa and 30 kDa. The ImmunoCAP® inhibition assays showed complete inhibition of pine nut specific IgE after serum incubation with Artemisia vulgaris extract. As far as we know, this is the first reported case of documented cross-reactivity between pine nut and Artemisia vulgaris


No disponible


Subject(s)
Humans , Male , Adult , Anaphylaxis/diagnosis , Anaphylaxis/epidemiology , Anaphylaxis/therapy , Pinus/adverse effects , Artemisia/adverse effects , Skin Tests/methods , Immunoblotting , Immunotherapy/methods , Cross-Priming , Antigens, Dermatophagoides/therapeutic use , Immunotherapy/instrumentation , Antigens, Dermatophagoides/immunology , Immunotherapy/trends , Cross-Priming/immunology , Cross-Priming/physiology , Hypersensitivity, Immediate/complications , Immunoglobulin E/immunology
8.
Hepatology ; 47(4): 1343-51, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18213574

ABSTRACT

UNLABELLED: The liver harbors a diversity of cell types that have been reported to stimulate T cells. Although most hepatic dendritic cells are immature, a small population of CD11c(high) conventional dendritic cells (cDCs) exists that expresses high levels of costimulatory molecules. We sought to determine the relative contribution of cDCs to cross-presentation by the liver. In vitro, liver nonparenchymal cells (NPCs) depleted of cDCs induced only minimal proliferation and activation of antigen-specific CD8(+) T cells when loaded with soluble protein antigen. Using a transgenic mouse with the CD11c promoter driving expression of the human diphtheria toxin receptor, we found that selective depletion of cDCs in vivo reduced the number and activation of antigen-specific CD8(+) T cells in the liver after intravenous administration of soluble protein antigen. Adoptive transfer of DCs, but not CD40 stimulation, restored the hepatic T-cell response. CONCLUSION: Our findings indicate that the ability of the liver to effectively cross-present soluble protein to antigen-specific CD8(+) T cells depends primarily on cDCs. Despite costimulation, other resident liver antigen-presenting cells cannot compensate for the absence of cDCs.


Subject(s)
Cross-Priming/physiology , Dendritic Cells/physiology , Liver/immunology , T-Lymphocytes/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin , Phenotype
9.
Cytokine Growth Factor Rev ; 19(1): 33-40, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18068417

ABSTRACT

Type I interferon (IFN-alpha/beta) is induced rapidly by infection and is well recognised for its crucial role in innate defence. However, it is evident that IFN-alpha/beta also serves as a signal for the generation of adaptive immune responses. In this review, we focus on the involvement of IFN-alpha/beta in the induction of CD8+ T cell responses by cross-priming.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/physiology , Interferon Type I/physiology , Antigen Presentation/physiology , Dendritic Cells/immunology , Humans , Interferon Type I/biosynthesis , Receptor, Interferon alpha-beta/physiology
10.
Immunology ; 119(1): 63-73, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16764686

ABSTRACT

CD8+ T cells play a crucial role in protective immunity to viruses and tumours. Antiviral CD8+ T cells are initially activated by professional antigen presenting cells (pAPCs) that are directly infected by viruses (direct-priming) or following uptake of exogenous antigen transferred from virus-infected or tumour cells (cross-priming). In order to efficiently target each of these antigen-processing pathways during vaccine design, it is necessary to delineate the properties of the natural substrates for either of these antigen-processing pathways. In this study, we utilized a novel T-cell receptor (TCR) transgenic mouse to examine the requirement for both antigen synthesis and synthesis of other cellular factors during direct or cross-priming. We found that direct presentation required ongoing synthesis of antigen, but that cross-priming favoured long-lived antigens and did not require ongoing antigen production. Even after prolonged blockade of protein synthesis in the donor cell, cross-priming was unaffected. In contrast, direct-presentation was almost undetectable in the absence of antigen neosynthesis and required ongoing protein synthesis. This suggests that the direct- and cross-priming pathways may utilize differing pools of antigen, an observation that has far-reaching implications for the rational design of vaccines aimed at the generation of protective CD8+ T cells.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/physiology , Virus Diseases/immunology , Adoptive Transfer/methods , Animals , Antigen-Presenting Cells/drug effects , Antigens/biosynthesis , Cell Line , Cycloheximide/pharmacology , Electroporation , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/prevention & control , Protein Synthesis Inhibitors/pharmacology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Virus Diseases/prevention & control
11.
J Neuroimmunol ; 174(1-2): 74-81, 2006 May.
Article in English | MEDLINE | ID: mdl-16504307

ABSTRACT

Central nervous system (CNS) tumors are thought to be poorly immunogenic. However, whether defective anti-tumor immunity is a consequence of a relative failure of T cell priming versus a deficient effector phase of the anti-tumor immune response is not clear. We utilized a well-defined model system of B16 melanoma expressing the model antigen SIY-GFP to evaluate tumor antigen cross-priming and tumor rejection from the CNS versus subcutaneous compartments. We observed that B16-SIY cells implanted in the CNS were capable of inducing T cell priming as measured by IFN-gamma ELISPOT in the spleen. Cross-priming occurred in the absence of detectable systemic dissemination of the tumor. Despite the induction of a T cell response, CNS tumors grew progressively and were fatal, whereas the same tumor implanted in the flank was rejected. To study the effector phase of the immune response in more detail, in vitro primed 2C/RAG2-/- TCR transgenic CD8+ cells, which recognize the SIY peptide, were adoptively transferred. In addition, the CNS microenvironment was modulated by intracranial delivery of IL-2. While mice that received primed 2C cells alone showed an increase in survival, co-administration of intracranial IL-2 led to a marked prolongation of survival, with 20% of mice surviving at least 120 days. Our results demonstrate that CD8+ T cell cross-priming does indeed occur in response to a CNS tumor, but that manipulation of the brain tumor microenvironment may be necessary to support the effector phase of the anti-tumor immune response.


Subject(s)
Abdominal Neoplasms/therapy , Antigens, Neoplasm/physiology , Brain Neoplasms/therapy , Cross-Priming/physiology , Immunotherapy/methods , Melanoma, Experimental/therapy , Abdominal Neoplasms/immunology , Animals , Brain Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Disease Models, Animal , Interleukin-2/immunology , Lymphocyte Activation , Male , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Survival Analysis , Time Factors
12.
Trends Immunol ; 25(2): 92-7, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15102368

ABSTRACT

At the beginning of this new millennium, pathogens and cancer remain the leading causes of death worldwide. The development of vaccines to prevent diseases for which no vaccine currently exists, such as AIDS or malaria, or to treat chronic infections or cancers, as well as the improvement of efficacy and safety of existing vaccines, remains a high priority. In most cases, the development of such vaccines requires strategies capable of stimulating CD8(+) cytotoxic T lymphocytes (CTLs) and thus, to deliver antigen to MHC class I molecules. There exists several different pathways for loading antigenic peptides onto MHC class I molecules, either based on the endogenous cytosolic MHC I pathway or on cross-presentation. The understanding of the relevance of each of these mechanisms in CTL activation will help vaccine design to progress more rationally.


Subject(s)
Antigen Presentation/physiology , Histocompatibility Antigens Class I/immunology , Vaccines/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Cross-Priming/immunology , Cross-Priming/physiology , Cytoplasm/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endocytosis/immunology , Endocytosis/physiology , Histocompatibility Antigens Class I/physiology , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Vaccines/metabolism , Vaccines, DNA/immunology , Vaccines, DNA/metabolism , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/metabolism
13.
Swiss Med Wkly ; 133(37-38): 501-6, 2003 Sep 26.
Article in English | MEDLINE | ID: mdl-14652798

ABSTRACT

The liver appears to be an organ favoring the induction of immune tolerance rather than immunity. Among the hepatic cell populations possibly involved in regulation of immune responses, liver sinusoidal endothelial cells (LSEC) are well suited to fulfill this role. LSEC are resident cells lining the hepatic sinusoidal wall and therefore are in intimate contact with leukocytes passing through the liver. They are equipped with numerous scavenger receptors rendering antigen-uptake in these cells extremely efficient. Antigen processing and MHC-restricted presentation of exogenous antigens for CD4 as well as CD8 T cells occurs equally with high efficiency. Importantly, CD4 and CD8 T cells that engaged in cognate interaction with LSEC have a tolerant phenotype. Thus LSEC contribute an important immune function to the liver: control of the immune response against circulating soluble antigens.


Subject(s)
Endothelial Cells/immunology , Immune Tolerance/physiology , Liver/cytology , Liver/immunology , Antigen Presentation/physiology , Cross-Priming/physiology , Endocytosis/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL