Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Elife ; 122024 Apr 09.
Article in English | MEDLINE | ID: mdl-38591522

ABSTRACT

Suppressive function of regulatory T cells (Treg) is dependent on signaling of their antigen receptors triggered by cognate self, dietary, or microbial peptides presented on MHC II. However, it remains largely unknown whether distinct or shared repertoires of Treg TCRs are mobilized in response to different challenges in the same tissue or the same challenge in different tissues. Here we use a fixed TCRß chain FoxP3-GFP mouse model to analyze conventional (eCD4) and regulatory (eTreg) effector TCRα repertoires in response to six distinct antigenic challenges to the lung and skin. This model shows highly 'digital' repertoire behavior with easy-to-track challenge-specific TCRα CDR3 clusters. For both eCD4 and eTreg subsets, we observe challenge-specific clonal expansions yielding homologous TCRα clusters within and across animals and exposure sites, which are also reflected in the draining lymph nodes but not systemically. Some CDR3 clusters are shared across cancer challenges, suggesting a response to common tumor-associated antigens. For most challenges, eCD4 and eTreg clonal response does not overlap. Such overlap is exclusively observed at the sites of certain tumor challenges, and not systematically, suggesting transient and local tumor-induced eCD4=>eTreg plasticity. This transition includes a dominant tumor-responding eCD4 CDR3 motif, as well as characteristic iNKT TCRα CDR3. In addition, we examine the homeostatic tissue residency of clonal eTreg populations by excluding the site of challenge from our analysis. We demonstrate that distinct CDR3 motifs are characteristic of eTreg cells residing in particular lymphatic tissues, regardless of the challenge. This observation reveals the tissue-resident, antigen-specific clonal Treg populations.


Subject(s)
CD4-Positive T-Lymphocytes , T-Lymphocytes, Regulatory , Mice , Animals , Receptors, Antigen, T-Cell/genetics , Peptides , Clone Cells
2.
J Exp Med ; 215(11): 2796-2814, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30355617

ABSTRACT

Regulatory T (Treg) cells prevent autoimmunity by limiting immune responses and inflammation in the secondary lymphoid organs and nonlymphoid tissues. While unique subsets of Treg cells have been described in some nonlymphoid tissues, their relationship to Treg cells in secondary lymphoid organs and circulation remains unclear. Furthermore, it is possible that Treg cells from similar tissue types share largely similar properties. We have identified a short-lived effector Treg cell subset that expresses the α2 integrin, CD49b, and exhibits a unique tissue distribution, being abundant in peripheral blood, vasculature, skin, and skin-draining lymph nodes, but uncommon in the intestines and in viscera-draining lymph nodes. CD49b+ Treg cells, which display superior functionality revealed by in vitro and in vivo assays, appear to develop after multiple rounds of cell division and TCR-dependent activation. Accordingly, single-cell RNA-seq analysis placed these cells at the apex of the Treg developmental trajectory. These results shed light on the identity and development of a functionally potent subset of mature effector Treg cells that recirculate through and survey peripheral tissues.


Subject(s)
Blood Vessels/immunology , Immunologic Surveillance , Integrin alpha2/immunology , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Integrin alpha2/genetics , Lymph Nodes/blood supply , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Transgenic , Skin/blood supply , Skin/cytology , T-Lymphocytes, Regulatory/cytology
3.
Elife ; 72018 05 31.
Article in English | MEDLINE | ID: mdl-29848443

ABSTRACT

Dynamic post-transcriptional control of RNA expression by RNA-binding proteins (RBPs) is critical during immune response. ZFP36 RBPs are prominent inflammatory regulators linked to autoimmunity and cancer, but functions in adaptive immunity are less clear. We used HITS-CLIP to define ZFP36 targets in mouse T cells, revealing unanticipated actions in regulating T-cell activation, proliferation, and effector functions. Transcriptome and ribosome profiling showed that ZFP36 represses mRNA target abundance and translation, notably through novel AU-rich sites in coding sequence. Functional studies revealed that ZFP36 regulates early T-cell activation kinetics cell autonomously, by attenuating activation marker expression, limiting T cell expansion, and promoting apoptosis. Strikingly, loss of ZFP36 in vivo accelerated T cell responses to acute viral infection and enhanced anti-viral immunity. These findings uncover a critical role for ZFP36 RBPs in restraining T cell expansion and effector functions, and suggest ZFP36 inhibition as a strategy to enhance immune-based therapies.


Subject(s)
Antiviral Agents/metabolism , Immunity , Lymphocyte Activation , RNA-Binding Proteins/metabolism , T-Lymphocytes/metabolism , Tristetraprolin/metabolism , Animals , Base Sequence , Bone Marrow/virology , CD4-Positive T-Lymphocytes/metabolism , Cell Lineage , Kinetics , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Ribosomes/metabolism , Transcriptome/genetics , Tristetraprolin/genetics
4.
Immunology ; 153(2): 133-144, 2018 02.
Article in English | MEDLINE | ID: mdl-29080364

ABSTRACT

For understanding the rules and laws of adaptive immunity, high-throughput profiling of T-cell receptor (TCR) repertoires becomes a powerful tool. The structure of TCR repertoires is instructive even before the antigen specificity of each particular receptor becomes available. It embodies information about the thymic and peripheral selection of T cells; the readiness of an adaptive immunity to withstand new challenges; the character, magnitude and memory of immune responses; and the aetiological and functional proximity of T-cell subsets. Here, we describe our current analytical approaches for the comparative analysis of murine TCR repertoires, and show several examples of how these approaches can be applied for particular experimental settings. We analyse the efficiency of different metrics used for estimation of repertoire diversity, repertoire overlap, V-gene and J-gene segments usage similarity, and amino acid composition of CDR3. We discuss basic differences of these metrics and their advantages and limitations in different experimental models, and we provide guidelines for choosing an efficient way to lead a comparative analysis of TCR repertoires. Applied to the various known and newly developed mouse models, such analysis should allow us to disentangle multiple sophisticated puzzles in adaptive immunity.


Subject(s)
Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Immunity, Cellular/physiology , T-Lymphocyte Subsets/immunology , Animals , Mice , T-Lymphocyte Subsets/cytology
6.
Nature ; 546(7658): 421-425, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28607488

ABSTRACT

Adaptive immune responses are tailored to different types of pathogens through differentiation of naive CD4 T cells into functionally distinct subsets of effector T cells (T helper 1 (TH1), TH2, and TH17) defined by expression of the key transcription factors T-bet, GATA3, and RORγt, respectively. Regulatory T (Treg) cells comprise a distinct anti-inflammatory lineage specified by the X-linked transcription factor Foxp3 (refs 2, 3). Paradoxically, some activated Treg cells express the aforementioned effector CD4 T cell transcription factors, which have been suggested to provide Treg cells with enhanced suppressive capacity. Whether expression of these factors in Treg cells-as in effector T cells-is indicative of heterogeneity of functionally discrete and stable differentiation states, or conversely may be readily reversible, is unknown. Here we demonstrate that expression of the TH1-associated transcription factor T-bet in mouse Treg cells, induced at steady state and following infection, gradually becomes highly stable even under non-permissive conditions. Loss of function or elimination of T-bet-expressing Treg cells-but not of T-bet expression in Treg cells-resulted in severe TH1 autoimmunity. Conversely, following depletion of T-bet- Treg cells, the remaining T-bet+ cells specifically inhibited TH1 and CD8 T cell activation consistent with their co-localization with T-bet+ effector T cells. These results suggest that T-bet+ Treg cells have an essential immunosuppressive function and indicate that Treg cell functional heterogeneity is a critical feature of immunological tolerance.


Subject(s)
Immune Tolerance/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Animals , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Separation , Female , Lymphocyte Activation , Male , Mice , T-Lymphocytes, Regulatory/cytology , Th1 Cells/cytology , Th17 Cells/cytology , Th17 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology
7.
J Exp Med ; 214(3): 609-622, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28130403

ABSTRACT

The regulatory T cell (T reg cell) T cell receptor (TCR) repertoire is highly diverse and skewed toward recognition of self-antigens. TCR expression by T reg cells is continuously required for maintenance of immune tolerance and for a major part of their characteristic gene expression signature; however, it remains unknown to what degree diverse TCR-mediated interactions with cognate self-antigens are required for these processes. In this study, by experimentally switching the T reg cell TCR repertoire to a single T reg cell TCR, we demonstrate that T reg cell function and gene expression can be partially uncoupled from TCR diversity. An induced switch of the T reg cell TCR repertoire to a random repertoire also preserved, albeit to a limited degree, the ability to suppress lymphadenopathy and T helper cell type 2 activation. At the same time, these perturbations of the T reg cell TCR repertoire led to marked immune cell activation, tissue inflammation, and an ultimately severe autoimmunity, indicating the importance of diversity and specificity for optimal T reg cell function.


Subject(s)
Autoimmunity , T-Cell Antigen Receptor Specificity , T-Lymphocytes, Regulatory/immunology , Animals , Forkhead Transcription Factors/analysis , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Receptors, Antigen, T-Cell/physiology
8.
Nature ; 528(7580): 132-136, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26605529

ABSTRACT

T-cell receptor (TCR) signalling has a key role in determining T-cell fate. Precursor cells expressing TCRs within a certain low-affinity range for complexes of self-peptide and major histocompatibility complex (MHC) undergo positive selection and differentiate into naive T cells expressing a highly diverse self-MHC-restricted TCR repertoire. In contrast, precursors displaying TCRs with a high affinity for 'self' are either eliminated through TCR-agonist-induced apoptosis (negative selection) or restrained by regulatory T (Treg) cells, whose differentiation and function are controlled by the X-chromosome-encoded transcription factor Foxp3 (reviewed in ref. 2). Foxp3 is expressed in a fraction of self-reactive T cells that escape negative selection in response to agonist-driven TCR signals combined with interleukin 2 (IL-2) receptor signalling. In addition to Treg cells, TCR-agonist-driven selection results in the generation of several other specialized T-cell lineages such as natural killer T cells and innate mucosal-associated invariant T cells. Although the latter exhibit a restricted TCR repertoire, Treg cells display a highly diverse collection of TCRs. Here we explore in mice whether a specialized mechanism enables agonist-driven selection of Treg cells with a diverse TCR repertoire, and the importance this holds for self-tolerance. We show that the intronic Foxp3 enhancer conserved noncoding sequence 3 (CNS3) acts as an epigenetic switch that confers a poised state to the Foxp3 promoter in precursor cells to make Treg cell lineage commitment responsive to a broad range of TCR stimuli, particularly to suboptimal ones. CNS3-dependent expansion of the TCR repertoire enables Treg cells to control self-reactive T cells effectively, especially when thymic negative selection is genetically impaired. Our findings highlight the complementary roles of these two main mechanisms of self-tolerance.


Subject(s)
Self Tolerance/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Cell Lineage , Conserved Sequence/genetics , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Female , Forkhead Transcription Factors/genetics , Introns/genetics , Male , Mice , Promoter Regions, Genetic/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin-2/immunology , Receptors, Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/deficiency , AIRE Protein
9.
Cell ; 162(5): 1078-89, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26317471

ABSTRACT

Regulatory T (Treg) cells suppress immune responses to a broad range of non-microbial and microbial antigens and indirectly limit immune inflammation-inflicted tissue damage by employing multiple mechanisms of suppression. Here, we demonstrate that selective Treg cell deficiency in amphiregulin leads to severe acute lung damage and decreased blood oxygen concentration during influenza virus infection without any measureable alterations in Treg cell suppressor function, antiviral immune responses, or viral load. This tissue repair modality is mobilized in Treg cells in response to inflammatory mediator IL-18 or alarmin IL-33, but not by TCR signaling that is required for suppressor function. These results suggest that, during infectious lung injury, Treg cells have a major direct and non-redundant role in tissue repair and maintenance-distinct from their role in suppression of immune responses and inflammation-and that these two essential Treg cell functions are invoked by separable cues.


Subject(s)
Influenza, Human/immunology , Lung/cytology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Amphiregulin/genetics , Animals , Autoimmunity , Disease Models, Animal , Humans , Influenza, Human/pathology , Lung/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Suppressor Factors, Immunologic/analysis , T-Lymphocytes, Regulatory/chemistry
10.
PLoS One ; 9(1): e86762, 2014.
Article in English | MEDLINE | ID: mdl-24466225

ABSTRACT

Regulatory T (Treg) cells enforce T cell homeostasis and maintain peripheral T cell tolerance. Here we report a previously unappreciated phenomenon of acute T cell lymphopenia in secondary lymphoid organs and non-lymphoid tissues triggered by Treg cell depletion that precedes the expansion of self-reactive T cells. Lymphopenia affects both neonates and adults indicating a dominant role of Treg cells in maintaining peripheral T cell numbers regardless of the developmental stage. The lymphopenia was neither triggered by caspase-dependent apoptosis nor macrophage-mediated clearance of T cells, nor diminished survival of naïve or recently activated T cells due to paucity of IL-7. It is possible that transient lymphopenia associated with congenital or acute Treg cell deficiency may contribute to the development of T cell mediated autoimmune disorders.


Subject(s)
Apoptosis/immunology , Forkhead Transcription Factors/physiology , Lymphopenia/immunology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Animals, Newborn , Caspases/metabolism , Female , Flow Cytometry , Interleukin-7/immunology , Interleukin-7/metabolism , Lymphopenia/metabolism , Macrophages/metabolism , Male , Mice , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/metabolism
11.
PLoS One ; 9(1): e87240, 2014.
Article in English | MEDLINE | ID: mdl-24466345

ABSTRACT

Hemocyanins, the huge oxygen-transporting glycoproteins of some mollusks, are used as immunomodulatory proteins with proven anti-cancer properties. The biodiversity of hemocyanins has promoted interest in identifying new anti-cancer candidates with improved immunological properties. Hemocyanins promote Th1 responses without known side effects, which make them ideal for long-term sustained treatment of cancer. In this study, we evaluated a novel hemocyanin from the limpet/gastropod Fissurella latimarginata (FLH). This protein has the typical hollow, cylindrical structure of other known hemocyanins, such as the keyhole limpet hemocyanin (KLH) and the Concholepas hemocyanin (CCH). FLH, like the KLH isoforms, is composed of a single type of polypeptide with exposed N- and O-linked oligosaccharides. However, its immunogenicity was significantly greater than that of KLH and CCH, as FLH induced a stronger humoral immune response and had more potent anti-tumor activity, delaying tumor growth and increasing the survival of mice challenged with B16F10 melanoma cells, in prophylactic and therapeutic settings. Additionally, FLH-treated mice demonstrated increased IFN-γ production and higher numbers of tumor-infiltrating CD4(+) lymphocytes. Furthermore, in vitro assays demonstrated that FLH, but not CCH or KLH, stimulated the rapid production of pro-inflammatory cytokines (IL-6, IL-12, IL-23 and TNF-α) by dendritic cells, triggering a pro-inflammatory milieu that may explain its enhanced immunological activity. Moreover, this effect was abolished when deglycosylated FLH was used, suggesting that carbohydrates play a crucial role in the innate immune recognition of this protein. Altogether, our data demonstrate that FLH possesses increased anti-tumor activity in part because it activates a more potent innate immune response in comparison to other known hemocyanins. In conclusion, FLH is a potential new marine adjuvant for immunization and possible cancer immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Gastropoda/chemistry , Hemocyanins/isolation & purification , Hemocyanins/pharmacology , Immunity, Innate/drug effects , Immunologic Factors/pharmacology , Melanoma/drug therapy , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Hemocyanins/ultrastructure , Kaplan-Meier Estimate , Melanoma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Rosaniline Dyes
12.
J Virol ; 86(22): 12304-12, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22951826

ABSTRACT

The majority of deaths following influenza virus infection result from secondary bacterial superinfection, most commonly caused by Streptococcus pneumoniae. Several models have been proposed to explain how primary respiratory viral infections exacerbate secondary bacterial disease, but the mechanistic explanations have been contradictory. In this study, mice were infected with S. pneumoniae at different days after primary influenza A (X31) virus infection. Our findings show that the induction of type I interferons (IFNs) during a primary nonlethal influenza virus infection is sufficient to promote a deadly S. pneumoniae secondary infection. Moreover, mice deficient in type I interferon receptor (IFNAR knockout [KO] mice) effectively cleared the secondary bacterial infection from their lungs, increased the recruitment of neutrophils, and demonstrated an enhanced innate expression of interleukin-17 (IL-17) relative to wild-type (WT) mice. Lung γδ T cells were responsible for almost all IL-17 production, and their function is compromised during secondary S. pneumoniae infection of WT but not IFNAR KO mice. Adoptive transfer of γδ T cells from IFNAR KO mice reduced the susceptibility to secondary S. pneumoniae infection in the lung of WT mice. Altogether, our study highlights the importance of type I interferon as a key master regulator that is exploited by opportunistic pathogens such as S. pneumoniae. Our findings may be utilized to design effective preventive and therapeutic strategies that may be beneficial for coinfected patients during influenza epidemics.


Subject(s)
Influenza, Human/metabolism , Interferon Type I/metabolism , Orthomyxoviridae/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Streptococcus pneumoniae/metabolism , T-Lymphocytes/immunology , Animals , Female , Flow Cytometry/methods , Humans , Influenza, Human/complications , Interleukin-17/metabolism , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Pneumonia, Pneumococcal/etiology , Pneumonia, Pneumococcal/virology , Signal Transduction
13.
PLoS Pathog ; 7(11): e1002345, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22072965

ABSTRACT

Migratory lung dendritic cells (DCs) transport viral antigen from the lungs to the draining mediastinal lymph nodes (MLNs) during influenza virus infection to initiate the adaptive immune response. Two major migratory DC subsets, CD103(+) DCs and CD11b(high) DCs participate in this function and it is not clear if these antigen presenting cell (APC) populations become directly infected and if so whether their activity is influenced by the infection. In these experiments we show that both subpopulations can become infected and migrate to the draining MLN but a difference in their response to type I interferon (I-IFN) signaling dictates the capacity of the virus to replicate. CD103(+) DCs allow the virus to replicate to significantly higher levels than do the CD11b(high) DCs, and they release infectious virus in the MLNs and when cultured ex-vivo. Virus replication in CD11b(high) DCs is inhibited by I-IFNs, since ablation of the I-IFN receptor (IFNAR) signaling permits virus to replicate vigorously and productively in this subset. Interestingly, CD103(+) DCs are less sensitive to I-IFNs upregulating interferon-induced genes to a lesser extent than CD11b(high) DCs. The attenuated IFNAR signaling by CD103(+) DCs correlates with their described superior antigen presentation capacity for naïve CD8(+) T cells when compared to CD11b(high) DCs. Indeed ablation of IFNAR signaling equalizes the competency of the antigen presenting function for the two subpopulations. Thus, antigen presentation by lung DCs is proportional to virus replication and this is tightly constrained by I-IFN. The "interferon-resistant" CD103(+) DCs may have evolved to ensure the presentation of viral antigens to T cells in I-IFN rich environments. Conversely, this trait may be exploitable by viral pathogens as a mechanism for systemic dissemination.


Subject(s)
Dendritic Cells/immunology , Influenza A Virus, H1N1 Subtype/immunology , Interferon Type I/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Adaptive Immunity , Animals , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , Antigens, CD/analysis , Antigens, Viral/immunology , CD11b Antigen/analysis , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Dendritic Cells/metabolism , Dendritic Cells/virology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Integrin alpha Chains/analysis , Interferon Type I/metabolism , Lymph Nodes/immunology , Lymph Nodes/virology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/virology , Receptors, Interferon/antagonists & inhibitors , Virus Replication
14.
Rev Med Chil ; 139(2): 236-46, 2011 Feb.
Article in Spanish | MEDLINE | ID: mdl-21773663

ABSTRACT

Hemocyanins, the giant oxygen transporter glycoproteins of diverse mollusks, are xenogenic to the mammalian immune system and they display a remarkable immuno-genicity. Therefore they are ideal non-specific immunostimulants to treat some types of cancer. They are used as an alternative therapy for superficial urinary bladder cancer (SBC), that has been traditionally treated with Bacillus Calmette-Guerin (BCG). In contrast to BCG, hemocyanins do not cause side-effects, making them ideal for long-term repetitive treatments. Hemocyanins have also been exploited as carriers to develop antibodies against hapten molecules and peptides, as carrier-adjuvants for cutting-edge vaccines against cancer, drug addiction, and infectious diseases and in the diagnosis of parasitic diseases, such as Schistosomiasis. The hemocyanin from Megathura crenulata, also known as keyhole limpet hemocyanin (KLH), has been used for over thirty years for the purposes described above. More recently, hemoc yanin from the Chilean mollusk Concholepas concholepas (CCH) has proved to be a reliable alternative to KLH, either as carrier protein, and as a likely alternative for the immunotherapy of SBC. Despite KLH and CCH differ significantly in their origin and structure, we have demonstrated that both hemocyanins stimulate the immune system of mammals in a similar way by inducing a potent Thl-polarized cellular and humoral response.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Hemocyanins/immunology , Mollusca/immunology , Vaccines/immunology , Animals , Cancer Vaccines/immunology
15.
Nat Chem Biol ; 6(8): 610-4, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20601941

ABSTRACT

Identification of immune effectors and the post-translational modifications that control their activity is essential for dissecting mechanisms of immunity. Here we demonstrate that the antiviral activity of interferon-induced transmembrane protein 3 (IFITM3) is post-translationally regulated by S-palmitoylation. Large-scale profiling of palmitoylated proteins in a dendritic cell line using a chemical reporter strategy revealed over 150 lipid-modified proteins with diverse cellular functions, including innate immunity. We discovered that S-palmitoylation of IFITM3 on membrane-proximal cysteines controls its clustering in membrane compartments and its antiviral activity against influenza virus. The sites of S-palmitoylation are highly conserved among the IFITM family of proteins in vertebrates, which suggests that S-palmitoylation of these immune effectors may be an ancient post-translational modification that is crucial for host resistance to viral infections. The S-palmitoylation and clustering of IFITM3 will be important for elucidating its mechanism of action and for the design of antiviral therapeutics.


Subject(s)
Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Metabolomics , Palmitic Acids/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/pharmacology , Cell Line , Cell Membrane/metabolism , Cysteine/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , HeLa Cells , Humans , Immunity, Innate/physiology , Immunoprecipitation , Membrane Proteins/genetics , Protein Modification, Translational , Proteomics , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
16.
Cell Host Microbe ; 7(5): 343-53, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20478536

ABSTRACT

Respiratory viral infections trigger a robust inflammatory response in the lung, producing cytokines, chemokines, and growth factors that promote infiltration of effector leukocytes. Whereas the role of chemokines and infiltrating leukocytes in antiviral immunity is well studied, the effect that lung cytokines have on leukocytes in distal hematopoietic and lymphoid tissues and their role in antiviral immunity is unknown. We show that, during infection with influenza or Sendai virus, the lung communicates with the sterile bone marrow, the primary site of hematopoiesis, through type I interferons. While in the bone marrow, leukocytes exposed to type I interferons activate an antiviral transcriptional program and become resistant to infection with different viruses. The protected bone marrow leukocytes are capable of migrating to the infected lung and contribute to virus clearance. These findings show that appropriate instruction of cells during their development in the bone marrow is needed for effective control of infection.


Subject(s)
Bone Marrow/immunology , Cytokines/immunology , Leukocytes/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Respiratory Tract Infections/immunology , Respirovirus Infections/immunology , Animals , Mice , Mice, Inbred C57BL , Orthomyxoviridae/immunology , Sendai virus/immunology
17.
Viruses ; 2(11): 2541-58, 2010 11.
Article in English | MEDLINE | ID: mdl-21994630

ABSTRACT

Respiratory viruses cause disease in humans characterized by an abrupt onset of symptoms. Studies in humans and animal models have shown that symptoms are not immediate and appear days or even weeks after infection. Since the initial symptoms are a manifestation of virus recognition by elements of the innate immune response, early virus replication must go largely undetected. The interval between infection and the emergence of symptoms is called the incubation period and is widely used as a clinical score. While incubation periods have been described for many virus infections the underlying mechanism for this asymptomatic phase has not been comprehensively documented. Here we review studies of the interaction between human pathogenic respiratory RNA viruses and the host with a particular emphasis on the mechanisms used by viruses to inhibit immunity. We discuss the concept of the "stealth phase", defined as the time between infection and the earliest detectable inflammatory response. We propose that the "stealth phase" phenomenon is primarily responsible for the suppression of symptoms during the incubation period and results from viral antagonism that inhibits major pathways of the innate immune system allowing an extended time of unhindered virus replication.

18.
J Immunol ; 183(6): 3569-73, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717515

ABSTRACT

A timely immune response is crucial for the effective control of virus infection. The influenza virus NS1 protein interferes with the expression of key proinflammatory cytokines from infected cells in vitro. To investigate the effect of NS1 during the onset of immunity in vivo, we systematically studied the early events that occur in the lungs and draining lymph nodes upon infection with influenza virus. Strikingly, no sign of innate immunity was detected in the lungs for almost 2 days after infection until a sudden inflammatory burst, including IFNs, cytokines, and chemokines, occurred. This burst preceded the robust dendritic cell migration and T cell activation in the lymph nodes. An NS1-deficient virus triggered rapid inflammation in the lungs whereas a wild-type virus did not. Thus, we demonstrate that, in vivo, influenza virus uses the NS1 protein to replicate for almost 2 days after infection before detection by the immune system.


Subject(s)
Immunity , Influenza A virus/physiology , Viral Nonstructural Proteins/physiology , Virus Replication , Animals , Inflammation/virology , Influenza A virus/immunology , Lung/immunology , Lung/pathology , Lung/virology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/virology , Mice , Orthomyxoviridae Infections , Time Factors
19.
Int Immunopharmacol ; 9(3): 330-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19159699

ABSTRACT

Hemocyanin, the oxygen transporter metallo-glycoprotein from mollusks, shows strong relationship between its notable structural features and intrinsic immunomodulatory effects. Here we investigated the individual contribution of CCHA and CCHB subunits from Concholepas hemocyanin (CCH) to in vivo humoral immune response and their pre-clinical evaluation as immunotherapeutic agent in a mice bladder cancer model, in relation to their biochemical properties. To this end, subunits were purified and well characterized. Homogeneous subunits were obtained by anionic exchange chromatography, and its purity assessed by electrophoretic and immunochemical methods. While each CCH subunit contains eight functional units showing partial cross reaction, the vibrational spectral analysis showed several spectral differences, suggesting structural differences between them. In addition, we demonstrated differences in the carbohydrate content: CCHA had a 3.6% w/w sugar with both N- and O-linked moieties. In turn, CCHB had a 2.5% w/w sugar with N-linked, while O-linked moieties were nearly absent. Considering these differences, it was not possible to predict a priori whether the immunogenic and immunotherapeutic properties of subunits might be similar. Surprisingly, both subunits by itself induced a humoral response, and showed an antitumor effect in the bladder carcinoma cell line MBT-2. However, when immunologic parameters were analyzed, CCHA showed better efficiency than CCHB. No allergic reactions or any toxic effects were observed in mice treated with CCHA, sustaining its potential therapeutic use. Our study supports that CCHA subunit accounts for the most important features involved in the immunogenicity of CCH, such as better hydrophilicity and higher content of carbohydrates.


Subject(s)
Antineoplastic Agents/immunology , Carcinoma/drug therapy , Gastropoda/chemistry , Hemocyanins/immunology , Urinary Bladder Neoplasms/drug therapy , Animals , Antibody Formation , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Carcinoma/immunology , Cell Line, Tumor , Cross Reactions/immunology , Hemocyanins/chemistry , Hemocyanins/therapeutic use , Immunotherapy , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Protein Subunits/chemistry , Protein Subunits/immunology , Protein Subunits/therapeutic use , Urinary Bladder Neoplasms/immunology
20.
J Virol ; 83(2): 584-97, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19004953

ABSTRACT

One attractive strategy for the development of a human immunodeficiency virus (HIV) vaccine is the use of viral vectors with a proven safety profile and an absence of preexisting immunity in humans, such as Newcastle disease virus (NDV). Several NDV vaccine vectors have been generated, and their immunogenicities have been investigated with different animal models. However, a systematic study to evaluate the optimal insertion site of the foreign antigens into NDV that results in enhanced immune responses specific to the antigen has not yet been conducted. In this article, we describe the ability of NDV expressing HIV Gag to generate a Gag-specific immune response in mice. We also have determined the optimal insertion site into the NDV genome by generating recombinant NDV-HIVGag viruses in which HIV gag was located at different transcriptional positions throughout the NDV viral genome. All recombinant viruses were viable, grew to similar titers in embryonated chicken eggs, and expressed Gag in a stable manner. Our in vivo experiments revealed that higher HIV Gag protein expression positively correlates with an enhanced CD8(+) T-cell-mediated immune response and protective immunity against challenge with vaccinia virus expressing HIV Gag. We also inserted a codon-optimized version of HIV gag in the described best location, between the P and M genes. Virus expressing the codon-optimized version of HIV gag induced a higher expression of the protein and an enhanced immune response against HIV Gag in mice. These results indicate that strategies directed toward increasing antigen expression by NDV result in enhanced immunogenicity and vaccine efficacy.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Expression , Genetic Vectors , Newcastle disease virus/genetics , gag Gene Products, Human Immunodeficiency Virus/biosynthesis , AIDS Vaccines/genetics , Animals , Chick Embryo , Female , Mice , Mice, Inbred BALB C , Microbial Viability , Newcastle disease virus/growth & development , Recombination, Genetic , Vaccinia/prevention & control , Viral Plaque Assay , Viremia/prevention & control , gag Gene Products, Human Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...